1
|
He B, Guo W, Shi R, Hoffman RD, Luo Q, Hu YJ, Gao J. Ruyong formula improves thymus function of CUMS-stimulated breast cancer mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117164. [PMID: 37717843 DOI: 10.1016/j.jep.2023.117164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/20/2023] [Accepted: 09/08/2023] [Indexed: 09/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ruyong Formula (RYF) is a famous Chinese herbal formula composed of 10 traditional Chinese herbs. It has been used as a therapeutic agent for breast cancer patients with depressive symptoms in China. However, its underlying pharmacological mechanism remains unclear. AIM OF THE STUDY This study aimed to explore the mechanism of RYF on the changes of thymus immune function in breast cancer body under mood disorders such as depression/anxiety. MATERIALS AND METHODS The chronic unpredictable mild stress (CUMS) was used to stimulate 4T1 breast cancer mice. The behavioral changes, 5-hydroxytryptamine (5-HT) level in brain, cytokeratin 5 (CK5) and 8 (CK8) expression in thymus, the proportion of T cell subsets, the thymic output, phenotypic changes of thymus epithelial cells (TECs), the expression levels of immune-related factors and downstream proteins of TSLP were analyzed after RYF treatment. RESULTS In CUMS stimulated group, the level of 5-HT in brain was significantly increased after RYF treatment. The output function of the thymus was improved, and the number of TECs in the medulla (CK5+), the proportion of CD3+CD4-CD8- (Double negative) and CD3+CD4+CD8+ (Double positive) T cells were all increased. The mRNA level of TSLP in mouse thymus was significantly decreased, but increased for IL-7. The protein levels of TSLP and Vimentin were decreased, but increased for p-STAT3, p-JAK2, E-cadherin, and p-PI3K p55 in vivo. In vitro study was showed the levels of Snail 1, Zeb 1 and Smad increased significantly in TGF-β1 group, and RYF could reverse their expression. CONCLUSIONS RYF could restore the structure and function of the thymus in depressed breast cancer mice by reversing the phenotypic changes of TECs and activating the JAK2/STAT3/PI3K pathway.
Collapse
Affiliation(s)
- Bingqian He
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Wenqin Guo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Rongzhen Shi
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Tangqi Branch of Traditional Chinese Medicine Hospital of Linping District, Hangzhou, Zhejiang, 311106, China.
| | - Robert D Hoffman
- Yo San University of Traditional Chinese Medicine, Los Angeles, CA, 90066, USA.
| | - Qihan Luo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Yuan-Jia Hu
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, 999078, China.
| | - Jianli Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
2
|
Roque S, de Sá-Calçada D, Cerqueira-Rodrigues B, Monteiro S, Guerreiro SG, Palha JA, Correia-Neves M. Chronic Mycobacterium avium infection differentially affects the cytokine expression profile of three mouse strains, but has no effect on behavior. Sci Rep 2023; 13:6199. [PMID: 37069180 PMCID: PMC10110542 DOI: 10.1038/s41598-023-33121-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 04/07/2023] [Indexed: 04/19/2023] Open
Abstract
One of the most remarkable findings in the immunology and neuroscience fields was the discovery of the bidirectional interaction between the immune and the central nervous systems. This interplay is tightly regulated to maintain homeostasis in physiological conditions. Disruption in this interplay has been suggested to be associated with several neuropsychiatric disorders. Most studies addressing the impact of an immune system disruption on behavioral alterations focus on acute pro-inflammatory responses. However, chronic infections are highly prevalent and associated with an altered cytokine milieu that persists over time. Studies addressing the potential effect of mycobacterial infections on mood behavior originated discordant results and this relationship needs to be further addressed. To increase our understanding on the effect of chronic infections on the central nervous system, we evaluated the role of Mycobacterium avium infection. A model of peripheral chronic infection with M. avium in female from three mouse strains (Balb/c, C57BL/6, and CD-1) was used. The effect of the infection was evaluated in the cytokine expression profile (spleen and hippocampus), hippocampal cell proliferation, neuronal plasticity, serum corticosterone production and mood behavior. The results show that M. avium peripheral chronic infection induces alterations not just in the peripheral immune system but also in the central nervous system, namely in the hippocampus. Interestingly, the cytokine expression profile alterations vary between mouse strains, and are not accompanied by hippocampal cell proliferation or neuronal plasticity changes. Accordingly, no differences were observed in locomotor, anxious and depressive-like behaviors, in any of the mouse strains used. We conclude that the M. avium 2447 infection-induced alterations in the cytokine expression profile, both in the periphery and the hippocampus, are insufficient to alter hippocampal plasticity and behavior.
Collapse
Affiliation(s)
- Susana Roque
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Daniela de Sá-Calçada
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Bruno Cerqueira-Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Susana Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Susana G Guerreiro
- Institute for Research and Innovation in Health (i3S), Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto-IPATIMUP, Porto, Portugal
- Biochemistry Unit, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Joana A Palha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Margarida Correia-Neves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
3
|
Rosichini M, Bordoni V, Silvestris DA, Mariotti D, Matusali G, Cardinale A, Zambruno G, Condorelli AG, Flamini S, Genah S, Catanoso M, Del Nonno F, Trezzi M, Galletti L, De Stefanis C, Cicolani N, Petrini S, Quintarelli C, Agrati C, Locatelli F, Velardi E. SARS-CoV-2 infection of thymus induces loss of function that correlates with disease severity. J Allergy Clin Immunol 2023; 151:911-921. [PMID: 36758836 PMCID: PMC9907790 DOI: 10.1016/j.jaci.2023.01.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 12/14/2022] [Accepted: 01/19/2023] [Indexed: 02/11/2023]
Abstract
BACKGROUND Lymphopenia, particularly when restricted to the T-cell compartment, has been described as one of the major clinical hallmarks in patients with coronavirus disease 2019 (COVID-19) and proposed as an indicator of disease severity. Although several mechanisms fostering COVID-19-related lymphopenia have been described, including cell apoptosis and tissue homing, the underlying causes of the decline in T-cell count and function are still not completely understood. OBJECTIVE Given that viral infections can directly target thymic microenvironment and impair the process of T-cell generation, we sought to investigate the impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on thymic function. METHODS We performed molecular quantification of T-cell receptor excision circles and κ-deleting recombination excision circles to assess, respectively, T- and B-cell neogenesis in SARS-CoV-2-infected patients. We developed a system for in vitro culture of primary human thymic epithelial cells (TECs) to mechanistically investigate the impact of SARS-CoV-2 on TEC function. RESULTS We showed that patients with COVID-19 had reduced thymic function that was inversely associated with the severity of the disease. We found that angiotensin-converting enzyme 2, through which SARS-CoV-2 enters the host cells, was expressed by thymic epithelium, and in particular by medullary TECs. We also demonstrated that SARS-CoV-2 can target TECs and downregulate critical genes and pathways associated with epithelial cell adhesion and survival. CONCLUSIONS Our data demonstrate that the human thymus is a target of SARS-CoV-2 and thymic function is altered following infection. These findings expand our current knowledge of the effects of SARS-CoV-2 infection on T-cell homeostasis and suggest that monitoring thymic activity may be a useful marker to predict disease severity and progression.
Collapse
Affiliation(s)
- Marco Rosichini
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy,Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Veronica Bordoni
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy,Cellular Immunology Laboratory, INMI L Spallanzani – IRCCS, Rome, Italy
| | - Domenico Alessandro Silvestris
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Davide Mariotti
- Cellular Immunology Laboratory, INMI L Spallanzani – IRCCS, Rome, Italy
| | - Giulia Matusali
- Virology Laboratory, INMI L Spallanzani – IRCCS, Rome, Italy
| | - Antonella Cardinale
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Giovanna Zambruno
- Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Angelo Giuseppe Condorelli
- Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Sara Flamini
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Shirley Genah
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Marialuigia Catanoso
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy,Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | | | - Matteo Trezzi
- Cardiac Surgery Unit, Department of Pediatric Cardiology and Cardiac Surgery, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Lorenzo Galletti
- Cardiac Surgery Unit, Department of Pediatric Cardiology and Cardiac Surgery, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Cristiano De Stefanis
- Pathology Unit, Core Research Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Nicolò Cicolani
- Confocal Microscopy Core Facility, Research Center, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Stefania Petrini
- Confocal Microscopy Core Facility, Research Center, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Concetta Quintarelli
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy,Department of Clinical Medicine and Surgery, University of Naples Federico II, Rome, Italy
| | - Chiara Agrati
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy,Cellular Immunology Laboratory, INMI L Spallanzani – IRCCS, Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy,Catholic University of the Sacred Heart, Rome, Italy
| | - Enrico Velardi
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| |
Collapse
|
4
|
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: 16] [Impact Index Per Article: 5.3] [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
|
5
|
Alterations in thymocyte populations under conditions of endotoxin tolerance. Chin Med J (Engl) 2021; 134:1855-1865. [PMID: 34133355 PMCID: PMC8367067 DOI: 10.1097/cm9.0000000000001598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background: Endotoxin tolerance (ET) is a protective phenomenon in which pre-treatment with a tolerance dose of lipopolysaccharide (LPS) leads to dramatically elevated survival. Accumulating evidence has shown that peripheral T cells contribute to the induction of ET. However, what happens to T cell development in the thymus under ET conditions remains unclear. The purpose of this study was to analyze the alterations in thymocyte populations (double-positive [DP] and single-positive [SP] cells) under ET conditions. Methods: Mice were intraperitoneally injected with LPS at a concentration of 5 mg/kg to establish an LPS tolerance model and were divided into two groups: a group examined 72 h after LPS injection (72-h group) and a group examined 8 days after LPS injection (8-day group). Injection of phosphate-buffered saline was used as a control (control group). Changes in thymus weight, cell counts, and morphology were detected in the three groups. Moreover, surface molecules such as CD4, CD8, CD44, CD69, and CD62L were analyzed using flow cytometry. Furthermore, proliferation, apoptosis, cytokine production, and extracellular signal-regulated kinase (ERK) pathway signaling were analyzed in thymocyte populations. The polymorphism and length of the T-cell receptor (TCR) β chain complementarity-determining region 3 (CDR3) were analyzed using capillary electrophoresis DNA laser scanning analysis (ABI 3730). Results: Thymus weight and cell counts were decreased in the early stage but recovered by the late stage in a murine model of LPS-induced ET. Moreover, the proportions of DP cells (control: 72.130 ± 4.074, 72-h: 10.600 ± 3.517, 8-day: 84.770 ± 2.228), CD4+ SP cells (control: 15.770 ± 4.419, 72-h: 44.670 ± 3.089, 8-day: 6.367 ± 0.513), and CD8+ SP cells (control: 7.000 ± 1.916, 72-h: 34.030 ± 3.850, 8-day: 5.133 ± 0.647) were obviously different at different stages of ET. The polymorphism and length of TCR β chain CDR3 also changed obviously, indicating the occurrence of TCR rearrangement and thymocyte diversification. Further analysis showed that the expression of surface molecules, including CD44, CD69, and CD62L, on thymocyte populations (DP and SP cells) were changed to different degrees. Finally, the proliferation, apoptosis, cytokine production, and ERK pathway signaling of thymocyte populations were changed significantly. Conclusion: These data reveal that alterations in thymocyte populations might contribute to the establishment of ET.
Collapse
|
6
|
Detection of Microbiota from Human Thymus of Myasthenia Gravis. Indian J Surg 2020. [DOI: 10.1007/s12262-020-02202-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
7
|
Elsaesser HJ, Mohtashami M, Osokine I, Snell LM, Cunningham CR, Boukhaled GM, McGavern DB, Zúñiga-Pflücker JC, Brooks DG. Chronic virus infection drives CD8 T cell-mediated thymic destruction and impaired negative selection. Proc Natl Acad Sci U S A 2020; 117:5420-5429. [PMID: 32094187 PMCID: PMC7071912 DOI: 10.1073/pnas.1913776117] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Chronic infection provokes alterations in inflammatory and suppressive pathways that potentially affect the function and integrity of multiple tissues, impacting both ongoing immune control and restorative immune therapies. Here we demonstrate that chronic lymphocytic choriomeningitis virus infection rapidly triggers severe thymic depletion, mediated by CD8 T cell-intrinsic type I interferon (IFN) and signal transducer and activator of transcription 2 (Stat2) signaling. Occurring temporal to T cell exhaustion, thymic cellularity reconstituted despite ongoing viral replication, with a rapid secondary thymic depletion following immune restoration by anti-programmed death-ligand 1 (PDL1) blockade. Therapeutic hematopoietic stem cell transplant (HSCT) during chronic infection generated new antiviral CD8 T cells, despite sustained virus replication in the thymus, indicating an impairment in negative selection. Consequently, low amounts of high-affinity self-reactive T cells also escaped the thymus following HSCT during chronic infection. Thus, by altering the stringency and partially impairing negative selection, the host generates new virus-specific T cells to replenish the fight against the chronic infection, but also has the potentially dangerous effect of enabling the escape of self-reactive T cells.
Collapse
Affiliation(s)
- Heidi J Elsaesser
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Mahmood Mohtashami
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8 Canada
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Ivan Osokine
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Laura M Snell
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Cameron R Cunningham
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Giselle M Boukhaled
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20824
| | - Juan Carlos Zúñiga-Pflücker
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2M9, Canada
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8 Canada
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - David G Brooks
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2M9, Canada;
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8 Canada
| |
Collapse
|
8
|
Losada-Barragán M, Umaña-Pérez A, Durães J, Cuervo-Escobar S, Rodríguez-Vega A, Ribeiro-Gomes FL, Berbert LR, Morgado F, Porrozzi R, Mendes-da-Cruz DA, Aquino P, Carvalho PC, Savino W, Sánchez-Gómez M, Padrón G, Cuervo P. Thymic Microenvironment Is Modified by Malnutrition and Leishmania infantum Infection. Front Cell Infect Microbiol 2019; 9:252. [PMID: 31355153 PMCID: PMC6639785 DOI: 10.3389/fcimb.2019.00252] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/28/2019] [Indexed: 01/23/2023] Open
Abstract
Detrimental effects of malnutrition on immune responses to pathogens have long been recognized and it is considered a main risk factor for various infectious diseases, including visceral leishmaniasis (VL). Thymus is a target of both malnutrition and infection, but its role in the immune response to Leishmania infantum in malnourished individuals is barely studied. Because we previously observed thymic atrophy and significant reduction in cellularity and chemokine levels in malnourished mice infected with L. infantum, we postulated that the thymic microenvironment is severely compromised in those animals. To test this, we analyzed the microarchitecture of the organ and measured the protein abundance in its interstitial space in malnourished BALB/c mice infected or not with L. infantum. Malnourished-infected animals exhibited a significant reduction of the thymic cortex:medulla ratio and altered abundance of proteins secreted in the thymic interstitial fluid. Eighty-one percent of identified proteins are secreted by exosomes and malnourished-infected mice showed significant decrease in exosomal proteins, suggesting that exosomal carrier system, and therefore intrathymic communication, is dysregulated in those animals. Malnourished-infected mice also exhibited a significant increase in the abundance of proteins involved in lipid metabolism and tricarboxylic acid cycle, suggestive of a non-proliferative microenvironment. Accordingly, flow cytometry analysis revealed decreased proliferation of single positive and double positive T cells in those animals. Together, the reduced cortical area, decreased proliferation, and altered protein abundance suggest a dysfunctional thymic microenvironment where T cell migration, proliferation, and maturation are compromised, contributing for the thymic atrophy observed in malnourished animals. All these alterations could affect the control of the local and systemic infection, resulting in an impaired response to L. infantum infection.
Collapse
Affiliation(s)
- Monica Losada-Barragán
- Laboratório de Pesquisa em Leishmanioses, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil.,Grupo de Investigación en Biología Celular y Funcional e Ingeniería de Biomoléculas, Departamento de Biologia, Universidad Antonio Nariño, Bogotá, Colombia
| | - Adriana Umaña-Pérez
- Grupo de Investigación en Hormonas, Departamento de Química, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Jonathan Durães
- Laboratório de Pesquisa em Leishmanioses, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Sergio Cuervo-Escobar
- Facultad de Ciencias, Universidad de Ciencias Aplicadas y Ambientales, Bogotá, Colombia
| | - Andrés Rodríguez-Vega
- Laboratório de Pesquisa em Leishmanioses, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Flávia L Ribeiro-Gomes
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Luiz R Berbert
- Laboratório de Pesquisas sobre o Timo, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Fernanda Morgado
- Laboratório de Pesquisa em Leishmanioses, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Renato Porrozzi
- Laboratório de Pesquisa em Leishmanioses, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Daniella Arêas Mendes-da-Cruz
- Laboratório de Pesquisas sobre o Timo, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Neuroimunomodulação, Fiocruz, Rio de Janeiro, Brazil
| | | | - Paulo C Carvalho
- Computational Mass Spectrometry and Proteomics Group, Fiocruz, Rio de Janeiro, Brazil
| | - Wilson Savino
- Laboratório de Pesquisas sobre o Timo, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Neuroimunomodulação, Fiocruz, Rio de Janeiro, Brazil
| | - Myriam Sánchez-Gómez
- Grupo de Investigación en Hormonas, Departamento de Química, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Gabriel Padrón
- Laboratório de Pesquisa em Leishmanioses, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Patricia Cuervo
- Laboratório de Pesquisa em Leishmanioses, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| |
Collapse
|
9
|
Starikova EA, Golovin AS, Vasilyev KA, Karaseva AB, Serebriakova MK, Sokolov AV, Kudryavtsev IV, Burova LA, Voynova IV, Suvorov AN, Vasilyev VB, Freidlin IS. Role of arginine deiminase in thymic atrophy during experimental Streptococcus pyogenes infection. Scand J Immunol 2019; 89:e12734. [PMID: 30471128 DOI: 10.1111/sji.12734] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 11/11/2018] [Accepted: 11/18/2018] [Indexed: 01/08/2023]
Abstract
Expression of gene of arginine deiminase (AD) allows adaptation of Streptococcus pyogenes to adverse environmental conditions. AD activity can lead to L-arginine deficiency in the host cells' microenvironment. Bioavailability of L-arginine is an important factor regulating the functions of the immune cells in mammals. By introducing a mutation into S pyogenes M46-16, we obtained a strain with inactivated arcA/sagp gene (M49-16 delArcA), deficient in AD. This allowed elucidating the function of AD in pathogenesis of streptococcal infection. The virulence of the parental and mutant strains was examined in a murine model of subcutaneous streptococcal infection. L-arginine concentration in the plasma of mice infected with S pyogenes M49-16 delArcA remained unchanged in course of the entire experiment. At the same time mice infected with S pyogenes M49-16 demonstrated gradual diminution of L-arginine concentration in the blood plasma, which might be due to the activity of streptococcal AD. Mice infected with S pyogenes M49-16 delArcA demonstrated less intensive bacterial growth in the primary foci and less pronounced bacterial dissemination as compared with animals infected with the parental strain S pyogenes M46-16. Similarly, thymus involution, alterations in apoptosis, thymocyte subsets and Treg cells differentiation were less pronounced in mice infected with S pyogenes M49-16 delArcA than in those infected with the parental strain. The results obtained showed that S pyogenes M49-16 delArcA, unable to produce AD, had reduced virulence in comparison with the parental S pyogenes M49-16 strain. AD is an important factor for the realization of the pathogenic potential of streptococci.
Collapse
Affiliation(s)
| | | | | | - Alena Borisovna Karaseva
- Federal State Budgetary Scientific Institution, Institute of Experimental Medicine, St. Petersburg, Russia
| | | | - Alexey Victorovich Sokolov
- Federal State Budgetary Scientific Institution, Institute of Experimental Medicine, St. Petersburg, Russia.,Saint-Petersburg State University, St. Petersburg, Russia
| | - Igor Vladimirovich Kudryavtsev
- Federal State Budgetary Scientific Institution, Institute of Experimental Medicine, St. Petersburg, Russia.,Far Eastern Federal University Vladivostok, Russia
| | | | - Irina Vitalyevna Voynova
- Federal State Budgetary Scientific Institution, Institute of Experimental Medicine, St. Petersburg, Russia
| | - Alexander Nikolaevich Suvorov
- Federal State Budgetary Scientific Institution, Institute of Experimental Medicine, St. Petersburg, Russia.,Saint-Petersburg State University, St. Petersburg, Russia
| | - Vadim Borisovich Vasilyev
- Federal State Budgetary Scientific Institution, Institute of Experimental Medicine, St. Petersburg, Russia.,Saint-Petersburg State University, St. Petersburg, Russia
| | - Irina Solomonovna Freidlin
- Federal State Budgetary Scientific Institution, Institute of Experimental Medicine, St. Petersburg, Russia.,Saint-Petersburg State University, St. Petersburg, Russia.,Pavlov First Saint-Petersburg State Medical University, St. Petersburg, Russia
| |
Collapse
|
10
|
Li T, Yan F, Meng X, Wang J, Ting Kam RK, Zeng X, Liu Z, Zhou H, Yang F, Ren R, Liao K, Liu L. Improvement of glucocorticoid-impaired thymus function by dihydromyricetin via up-regulation of PPARγ-associated fatty acid metabolism. Pharmacol Res 2018; 137:76-88. [PMID: 30227260 DOI: 10.1016/j.phrs.2018.09.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/10/2018] [Accepted: 09/12/2018] [Indexed: 12/27/2022]
Abstract
T lymphocytes produced by the thymus are essential mediators of immunity. Accelerated thymic atrophy appears in the patients with administration of glucocorticoids (GCs) which are commonly-used drugs to treat autoimmune and infectious diseases, leading to dysregulation of immunity with manifestation of progressive diminution of new T cell production. However, there is no ideal method to overcome such side effects of GCs. In the current study, we proposed a composition of dexamethasone (DEX) and dihydromyricetin (DMY) derived from a medicinal plant, which could protect from DEX-induced thymus damage and simultaneously enhance the anti-inflammatory effect of DEX. In the current study, we found that DEX-damaged thymic cellularity and architecture, reduced thymocyte numbers, induced thymocyte apoptosis and dropped CD4+ and CD8+ double positive T cell numbers in thymus which was effectively improved by co-treatment with DMY. Quantification of signal joint TCR delta excision circles (TRECs) and Vβ TCR spectratyping analysis were employed to determine the thymus function with indicated treatments. The results showed that DEX-impaired thymus output and decreased TCR cell diversity which was ameliorated by co-treatment with DMY. iTRAQ 2D LC-MS/MS was applied to analyze the proteomic profiling of thymus of mice treated with or without indicated agents, followed by informatics analysis to identify the correlated signaling pathway. After validated by Western blotting and Real-time PCR, we found that PPARγ-associated fatty acid metabolism was increased in the thymic tissues of the animals treated with DMY plus DEX than the animals treated with DEX alone. The agonist and antagonist of PPARγ were further employed to verify the role of PPARγ in the present study. Furthermore, DMY demonstrated a synergistic effect with co-administration of DEX on suppressing inflammation in vivo. Collectively, DMY relieved thymus function damaged by DEX via regulation of PPARγ-associated fatty acid metabolism. Our findings may provide a new strategy on protection of thymus from damage caused by GCs by using appropriate adjuvant natural agents through up-regulation of PPARγ-associated fatty acid metabolism.
Collapse
Affiliation(s)
- Ting Li
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Fenggen Yan
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China; Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Xiongyu Meng
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Jingrong Wang
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Richard Kin Ting Kam
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China; Department of Chemical Pathology, Chinese University of Hong Kong, Hong Kong, China
| | - Xing Zeng
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Zhongqiu Liu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Hua Zhou
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Fen Yang
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Rutong Ren
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Kangsheng Liao
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China.
| |
Collapse
|
11
|
Cerqueira-Rodrigues B, Mendes A, Correia-Neves M, Nobrega C. Ag85-focused T-cell immune response controls Mycobacterium avium chronic infection. PLoS One 2018; 13:e0193596. [PMID: 29499041 PMCID: PMC5834192 DOI: 10.1371/journal.pone.0193596] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/14/2018] [Indexed: 01/09/2023] Open
Abstract
CD4+ T cells are essential players for the control of mycobacterial infections. Several mycobacterial antigens have been identified for eliciting a relevant CD4+ T cell mediated-immune response, and numerous studies explored this issue in the context of Mycobacterium tuberculosis infection. Antigen 85 (Ag85), a highly conserved protein across Mycobacterium species, is secreted at the early phase of M. tuberculosis infection leading to the proliferation of Ag85-specific CD4+ T cells. However, in the context of Mycobacterium avium infection, little is known about the expression of this antigen and the elicited immune response. In the current work, we investigated if a T cell receptor (TCR) repertoire mostly, but not exclusively, directed at Ag85 is sufficient to mount a protective immune response against M. avium. We show that P25 mice, whose majority of T cells express a transgenic TCR specific for Ag85, control M. avium infection at the same level as wild type (WT) mice up to 20 weeks post-infection (wpi). During M. avium infection, Ag85 antigen is easily detected in the liver of 20 wpi mice by immunohistochemistry. In spite of the propensity of P25 CD4+ T cells to produce higher amounts of interferon-gamma (IFNγ) upon ex vivo stimulation, no differences in serum IFNγ levels are detected in P25 compared to WT mice, nor enhanced immunopathology is detected in P25 mice. These results indicate that a T cell response dominated by Ag85-specific T cells is appropriate to control M. avium infection with no signs of immunopathology.
Collapse
Affiliation(s)
- Bruno Cerqueira-Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Mendes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Margarida Correia-Neves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Claudia Nobrega
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, Braga/Guimarães, Portugal
- * E-mail:
| |
Collapse
|
12
|
D’Attilio L, Santucci N, Bongiovanni B, Bay ML, Bottasso O. Tuberculosis, the Disrupted Immune-Endocrine Response and the Potential Thymic Repercussion As a Contributing Factor to Disease Physiopathology. Front Endocrinol (Lausanne) 2018; 9:214. [PMID: 29765355 PMCID: PMC5938357 DOI: 10.3389/fendo.2018.00214] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 04/16/2018] [Indexed: 12/31/2022] Open
Abstract
Upon the pathogen encounter, the host seeks to ensure an adequate inflammatory reaction to combat infection but at the same time tries to prevent collateral damage, through several regulatory mechanisms, like an endocrine response involving the production of adrenal steroid hormones. Our studies show that active tuberculosis (TB) patients present an immune-endocrine imbalance characterized by an impaired cellular immunity together with increased plasma levels of cortisol, pro-inflammatory cytokines, and decreased amounts of dehydroepiandrosterone. Studies in patients undergoing specific treatment revealed that cortisol levels remained increased even after several months of initiating therapy. In addition to the well-known metabolic and immunological effects, glucocorticoids are involved in thymic cortical depletion with immature thymocytes being quite sensitive to such an effect. The thymus is a central lymphoid organ supporting thymocyte T-cell development, i.e., lineage commitment, selection events and thymic emigration. While thymic TB is an infrequent manifestation of the disease, several pieces of experimental and clinical evidence point out that the thymus can be infected by mycobacteria. Beyond this, the thymic microenvironment during TB may be also altered because of the immune-hormonal alterations. The thymus may be then an additional target of organ involvement further contributing to a deficient control of infection and disease immunopathology.
Collapse
|
13
|
Ansari AR, Liu H. Acute Thymic Involution and Mechanisms for Recovery. Arch Immunol Ther Exp (Warsz) 2017; 65:401-420. [PMID: 28331940 DOI: 10.1007/s00005-017-0462-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 03/12/2017] [Indexed: 12/14/2022]
Abstract
Acute thymic involution (ATI) is usually regarded as a virulence trait. It is caused by several infectious agents (bacteria, viruses, parasites, fungi) and other factors, including stress, pregnancy, malnutrition and chemotherapy. However, the complex mechanisms that operate during ATI differ substantially from each other depending on the causative agent. For instance, a transient reduction in the size and weight of the thymus and depletion of populations of T cell subsets are hallmarks of ATI in many cases, whereas severe disruption of the anatomical structure of the organ is also associated with some factors, including fungal, parasitic and viral infections. However, growing evidence shows that ATI may be therapeutically halted or reversed. In this review, we highlight the current progress in this field with respect to numerous pathological factors and discuss the possible mechanisms. Moreover, these new observations also show that ATI can be mechanistically reversed.
Collapse
Affiliation(s)
- Abdur Rahman Ansari
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, 430070, Wuhan, Hubei, China
- Section of Anatomy and Histology, Department of Basic Sciences, College of Veterinary and Animal Sciences (CVAS), Jhang, Pakistan
- University of Veterinary and Animal Sciences (UVAS), Lahore, Pakistan
| | - Huazhen Liu
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, 430070, Wuhan, Hubei, China.
| |
Collapse
|
14
|
Losada-Barragán M, Umaña-Pérez A, Cuervo-Escobar S, Berbert LR, Porrozzi R, Morgado FN, Mendes-da-Cruz DA, Savino W, Sánchez-Gómez M, Cuervo P. Protein malnutrition promotes dysregulation of molecules involved in T cell migration in the thymus of mice infected with Leishmania infantum. Sci Rep 2017; 7:45991. [PMID: 28397794 PMCID: PMC5387407 DOI: 10.1038/srep45991] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 03/07/2017] [Indexed: 12/18/2022] Open
Abstract
Protein malnutrition, the most deleterious cause of malnutrition in developing countries, has been considered a primary risk factor for the development of clinical visceral leishmaniasis (VL). Protein malnutrition and infection with Leishmania infantum leads to lymphoid tissue disorganization, including changes in cellularity and lymphocyte subpopulations in the thymus and spleen. Here we report that protein malnutrition modifies thymic chemotactic factors by diminishing the CCL5, CXCL12, IGF1, CXCL9 and CXCL10 protein levels in infected animals. Nevertheless, T cells preserve their migratory capability, as they were able to migrate ex vivo in response to chemotactic stimuli, indicating that malnutrition may compromise the thymic microenvironment and alter in vivo thymocyte migration. Decrease in chemotactic factors protein levels was accompanied by an early increase in the parasite load of the spleen. These results suggest that the precondition of malnutrition is affecting the cell-mediated immune response to L. infantum by altering T cell migration and interfering with the capacity of protein-deprived animals to control parasite spreading and proliferation. Our data provide evidence for a disturbance of T lymphocyte migration involving both central and peripheral T-cells, which likely contribute to the pathophysiology of VL that occurs in malnourished individuals.
Collapse
Affiliation(s)
- Monica Losada-Barragán
- Laboratório de Pesquisas em Leishmaniose, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brasil
| | - Adriana Umaña-Pérez
- Departamento de Química, Universidad Nacional de Colombia, Sede Bogotá, Facultad de Ciencias, Grupo de Investigación en Hormonas, Bogotá, Colombia
| | - Sergio Cuervo-Escobar
- Departamento de Química, Universidad Nacional de Colombia, Sede Bogotá, Facultad de Ciencias, Grupo de Investigación en Hormonas, Bogotá, Colombia
| | - Luiz Ricardo Berbert
- Laboratório de Pesquisas sobre o Timo, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brasil
| | - Renato Porrozzi
- Laboratório de Pesquisas em Leishmaniose, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brasil
| | - Fernanda N Morgado
- Laboratório de Pesquisas em Leishmaniose, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brasil
| | | | - Wilson Savino
- Laboratório de Pesquisas sobre o Timo, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brasil
| | - Myriam Sánchez-Gómez
- Departamento de Química, Universidad Nacional de Colombia, Sede Bogotá, Facultad de Ciencias, Grupo de Investigación en Hormonas, Bogotá, Colombia.
| | - Patricia Cuervo
- Laboratório de Pesquisas em Leishmaniose, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brasil.
| |
Collapse
|
15
|
Nuñez S, Moore C, Gao B, Rogers K, Hidalgo Y, Del Nido PJ, Restaino S, Naka Y, Bhagat G, Madsen JC, Bono MR, Zorn E. The human thymus perivascular space is a functional niche for viral-specific plasma cells. Sci Immunol 2016; 1. [PMID: 28459117 DOI: 10.1126/sciimmunol.aah4447] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The human thymus is susceptible to viral infections that can severely alter thymopoiesis and compromise the mechanisms of acquired tolerance to self-antigens. In humans, plasma cells residing primarily in the bone marrow confer long-lasting protection to common viruses by secreting antigen-specific antibodies. Since the thymus also houses B cells, we examined the phenotypic complexity of these thymic resident cells and their possible protective role against viral infections. Using tissue specimens collected from subjects ranging in age from 5 days to 71 years, we found that starting during the first year of life, CD138+ plasma cells (PC) begin accumulating in the thymic perivascular space (PVS) where they constitutively produce IgG without the need for additional stimulation. These, thymic PC secrete almost exclusively IgG1 and IgG3, the two main complement-fixing effector IgG subclasses. Moreover, using antigen-specific ELISpot assays, we demonstrated that thymic PC include a high frequency of cells reactive to common viral proteins. Our study reveals an unrecognized role of the PVS as a functional niche for viral-specific PCs. The PVS is located between the thymic epithelial areas and the circulation. PCs located in this compartment may therefore provide internal protection against pathogen infections and preserve the integrity and function of the organ.
Collapse
Affiliation(s)
- Sarah Nuñez
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, USA.,Department of Biology, University of Chile, Santiago, Chile
| | - Carolina Moore
- MGH Transplant Center and Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Baoshan Gao
- MGH Transplant Center and Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kortney Rogers
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Yessia Hidalgo
- Department of Biology, University of Chile, Santiago, Chile
| | - Pedro J Del Nido
- Department of Surgery, Boston Children Hospital, Boston, MA, USA
| | - Susan Restaino
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Yoshifumi Naka
- Department of Surgery, Columbia University Medical Center, New York, NY, USA
| | - Govind Bhagat
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Joren C Madsen
- MGH Transplant Center and Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Emmanuel Zorn
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
16
|
Kugler DG, Flomerfelt FA, Costa DL, Laky K, Kamenyeva O, Mittelstadt PR, Gress RE, Rosshart SP, Rehermann B, Ashwell JD, Sher A, Jankovic D. Systemic toxoplasma infection triggers a long-term defect in the generation and function of naive T lymphocytes. J Exp Med 2016; 213:3041-3056. [PMID: 27849554 PMCID: PMC5154934 DOI: 10.1084/jem.20151636] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 08/05/2016] [Accepted: 10/18/2016] [Indexed: 12/12/2022] Open
Abstract
Kugler et al. show that systemic infection with Toxoplasma gondii triggers a long-term impairment in thymic function, which leads to an immunodeficient state reflected in decreased antimicrobial resistance. Because antigen-stimulated naive T cells either die as effectors or enter the activated/memory pool, continuous egress of new T lymphocytes from thymus is essential for maintenance of peripheral immune homeostasis. Unexpectedly, we found that systemic infection with the protozoan Toxoplasma gondii triggers not only a transient increase in activated CD4+ Th1 cells but also a persistent decrease in the size of the naive CD4+ T lymphocyte pool. This immune defect is associated with decreased thymic output and parasite-induced destruction of the thymic epithelium, as well as disruption of the overall architecture of that primary lymphoid organ. Importantly, the resulting quantitative and qualitative deficiency in naive CD4+ T cells leads to an immunocompromised state that both promotes chronic toxoplasma infection and leads to decreased resistance to challenge with an unrelated pathogen. These findings reveal that systemic infectious agents, such as T. gondii, can induce long-term immune alterations associated with impaired thymic function. When accumulated during the lifetime of the host, such events, even when occurring at low magnitude, could be a contributing factor in immunological senescence.
Collapse
Affiliation(s)
- David G Kugler
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Francis A Flomerfelt
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Diego L Costa
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Karen Laky
- T Cell Development Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Olena Kamenyeva
- Biological Imaging, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Paul R Mittelstadt
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Ronald E Gress
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Stephan P Rosshart
- Immunology Section, Liver Diseases Branch, National Institute for Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Barbara Rehermann
- Immunology Section, Liver Diseases Branch, National Institute for Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jonathan D Ashwell
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Dragana Jankovic
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
17
|
Alves da Costa T, Di Gangi R, Thomé R, Barreto Felisbino M, Pires Bonfanti A, Lumi Watanabe Ishikawa L, Sartori A, Burger E, Verinaud L. Severe Changes in Thymic Microenvironment in a Chronic Experimental Model of Paracoccidioidomycosis. PLoS One 2016; 11:e0164745. [PMID: 27736987 PMCID: PMC5063316 DOI: 10.1371/journal.pone.0164745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 09/29/2016] [Indexed: 12/26/2022] Open
Abstract
T cell maturation takes place within the thymus, a primary lymphoid organ that is commonly targeted during infections. Previous studies showed that acute infection with Paracoccidioides brasiliensis (Pb), the causative agent of paracoccidioidomycosis (PCM), promotes thymic atrophy that is associated with the presence of yeast cells in the organ. However, as human PCM is a chronic infection, it is imperative to investigate the consequences of Pb infection over the thymic structure and function in chronic infection. In this sense, we developed a new experimental model where Pb yeast cells are injected through the intraperitoneal route and mice are evaluated over 120 days of infection. Thymuses were analyzed in chronically infected mice and we found that the thymus underwent extensive morphological alterations and severe infiltration of P. brasiliensis yeast cells. Further analyses showed an altered phenotype and function of thymocytes that are commonly found in peripheral mature T lymphocytes. We also observed activation of the NLRP3 inflammasome in the thymus. Our data provide new information on the severe changes observed in the thymic microenvironment in a model of PCM that more closely mimics the human infection.
Collapse
Affiliation(s)
- Thiago Alves da Costa
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Rosária Di Gangi
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Rodolfo Thomé
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Marina Barreto Felisbino
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Amanda Pires Bonfanti
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Larissa Lumi Watanabe Ishikawa
- Department of Microbiology and Immunology, Institute of Biosciences of Botucatu, Univ. Estadual Paulista (UNESP), Botucatu, São Paulo, Brazil
| | - Alexandrina Sartori
- Department of Microbiology and Immunology, Institute of Biosciences of Botucatu, Univ. Estadual Paulista (UNESP), Botucatu, São Paulo, Brazil
| | - Eva Burger
- Department of Microbiology and Immunology, Institute of Biomedical Sciences, Federal University of Alfenas (UNIFAL-MG), Alfenas, Minas Gerais, Brazil
| | - Liana Verinaud
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
18
|
TNF superfamily members play distinct roles in shaping the thymic stromal microenvironment. Mol Immunol 2016; 72:92-102. [PMID: 27011037 DOI: 10.1016/j.molimm.2016.02.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 02/10/2016] [Accepted: 02/29/2016] [Indexed: 12/31/2022]
Abstract
The differentiation and proper function of thymic epithelial cells (TECs) depend on various tumor necrosis factor superfamily (TNFSF) signals that are needed to maintain the thymic stromal microenvironment. Nevertheless, the direct transcriptional effects of these signals on TECs remain unclear. To address this issue, we stimulated murine embryonic thymus tissue with selected TNFSF ligands and performed a gene expression profiling study. We show that Aire expression is a direct and specific effect of RANKL stimulation, whereas LTβ and TNFα are major inducers of chemokines in the thymic stroma and we propose differential NF-κB binding as one possible cause of these gene expression patterns. Our work provides further insight into the complex molecular pathways that shape the thymic microenvironment and maintain central tolerance.
Collapse
|
19
|
The discovery of the blood–thymus barrier. Immunol Lett 2015; 168:325-8. [DOI: 10.1016/j.imlet.2015.10.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 10/16/2015] [Accepted: 10/23/2015] [Indexed: 12/11/2022]
|
20
|
The Attenuated Live Yellow Fever Virus 17D Infects the Thymus and Induces Thymic Transcriptional Modifications of Immunomodulatory Genes in C57BL/6 and BALB/C Mice. Autoimmune Dis 2015; 2015:503087. [PMID: 26457200 PMCID: PMC4589579 DOI: 10.1155/2015/503087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/17/2015] [Accepted: 08/26/2015] [Indexed: 12/28/2022] Open
Abstract
Thymus is involved in induction of self-tolerance in T lymphocytes, particularly due to Aire activity. In peripheral tissues, Treg cells and immunomodulatory molecules, like the major histocompatibility complex (MHC) class Ib molecules, are essential for maintenance of autotolerance during immune responses. Viral infections can trigger autoimmunity and modify thymic function, and YFV17D immunization has been associated with the onset of autoimmunity, being contraindicated in patients with thymic disorders. Aiming to study the influence of YFV17D immunization on the transcriptional profiles of immunomodulatory genes in thymus, we evaluated the gene expression of AIRE, FOXP3, H2-Q7 (Qa-2/HLA-G), H2-T23 (Qa-1/HLA-E), H2-Q10, and H2-K1 following immunization with 10,000 LD50 of YFV17D in C57BL/6 and BALB/c mice. The YFV17D virus replicated in thymus and induced the expression of H2-Q7 (Qa-2/HLA-G) and H2-T23 (Qa-1/HLA-E) transcripts and repressed the expression of AIRE and FOXP3. Transcriptional expression varied according to tissue and mouse strain analyzed. Expression of H2-T23 (Qa-1/HLA-E) and FOXP3 was induced in thymus and liver of C57BL/6 mice, which exhibited defective control of viral load, suggesting a higher susceptibility to YFV17D infection. Since the immunization with YFV17D modulated thymus gene expression in genetically predisposed individuals, the vaccine may be related to the onset of autoimmunity disorders.
Collapse
|
21
|
Martínez VG, Canseco NM, Hidalgo L, Valencia J, Entrena A, Fernández-Sevilla LM, Hernández-López C, Sacedón R, Vicente A, Varas A. A discrete population of IFN λ-expressing BDCA3hi dendritic cells is present in human thymus. Immunol Cell Biol 2015; 93:673-8. [PMID: 25753268 DOI: 10.1038/icb.2015.22] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 01/19/2015] [Accepted: 02/01/2015] [Indexed: 12/24/2022]
Abstract
Human thymus contains two major subpopulations of dendritic cells (DCs), conventional DCs (cDCs) and plasmacytoid DCs (pDCs), which are mainly involved in central tolerance and also in protecting the thymus against infections. In blood and peripheral organs cDCs include the subpopulation of BDCA3(hi) DCs, considered as equivalents to mouse CD8α(+) DCs. In this study we describe in human thymus the presence of a discrete population of BDCA3(hi) DCs that, like their peripheral counterparts, express CD13, low-intermediate levels of CD11c, CLEC9A, high levels of XCR1, IRF8 and TLR3, and mostly lack the expression of CD11b, CD14 and TLR7. Thymic BDCA3(hi) DCs display immature features with a low expression of costimulatory molecules and HLA-DR, and a low allostimulatory capacity. Also, BDCA3(hi) DCs exhibit a strong response to TLR3 stimulation, producing high levels of interferon (IFN)-λ1 and CXCL10, which indicates that, similarly to thymic pDCs, BDCA3(hi) DCs can have an important role in thymus protection against viral infections.
Collapse
MESH Headings
- Antigens, Differentiation/analysis
- Antigens, Surface/analysis
- Apoptosis
- Cells, Cultured
- Chemokine CXCL10/analysis
- Child, Preschool
- Coculture Techniques
- Dendritic Cells/chemistry
- Dendritic Cells/classification
- Dendritic Cells/cytology
- HLA-DR Antigens/analysis
- Humans
- Infant
- Infant, Newborn
- Interferons
- Interleukins/analysis
- Interleukins/biosynthesis
- Interleukins/genetics
- Lectins, C-Type/analysis
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Receptors, G-Protein-Coupled/analysis
- Receptors, Mitogen/analysis
- Thrombomodulin
- Thymus Gland/cytology
- Thymus Gland/immunology
- Toll-Like Receptor 3/analysis
Collapse
Affiliation(s)
- Víctor G Martínez
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Noelia M Canseco
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Laura Hidalgo
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Jaris Valencia
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Ana Entrena
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | | | | | - Rosa Sacedón
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Angeles Vicente
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Alberto Varas
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| |
Collapse
|
22
|
Nunes-Alves C, Booty MG, Carpenter SM, Rothchild AC, Martin CJ, Desjardins D, Steblenko K, Kløverpris HN, Madansein R, Ramsuran D, Leslie A, Correia-Neves M, Behar SM. Human and Murine Clonal CD8+ T Cell Expansions Arise during Tuberculosis Because of TCR Selection. PLoS Pathog 2015; 11:e1004849. [PMID: 25945999 PMCID: PMC4422591 DOI: 10.1371/journal.ppat.1004849] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 04/01/2015] [Indexed: 12/17/2022] Open
Abstract
The immune system can recognize virtually any antigen, yet T cell responses against several pathogens, including Mycobacterium tuberculosis, are restricted to a limited number of immunodominant epitopes. The host factors that affect immunodominance are incompletely understood. Whether immunodominant epitopes elicit protective CD8+ T cell responses or instead act as decoys to subvert immunity and allow pathogens to establish chronic infection is unknown. Here we show that anatomically distinct human granulomas contain clonally expanded CD8+ T cells with overlapping T cell receptor (TCR) repertoires. Similarly, the murine CD8+ T cell response against M. tuberculosis is dominated by TB10.44-11-specific T cells with extreme TCRβ bias. Using a retrogenic model of TB10.44-11-specific CD8+ T cells, we show that TCR dominance can arise because of competition between clonotypes driven by differences in affinity. Finally, we demonstrate that TB10.4-specific CD8+ T cells mediate protection against tuberculosis, which requires interferon-γ production and TAP1-dependent antigen presentation in vivo. Our study of how immunodominance, biased TCR repertoires, and protection are inter-related, provides a new way to measure the quality of T cell immunity, which if applied to vaccine evaluation, could enhance our understanding of how to elicit protective T cell immunity. While T cells are required for protection against Mycobacterium tuberculosis infection, attempts to prevent tuberculosis by vaccines designed to elicit memory T cells have only been partially successful. Several vaccine candidates are in clinical trials, but progress has been slow because their ability to prevent disease must be empirically tested. There is little understanding of why certain antigens are targets of protective immunity. We have characterized an immunodominant CD8+ T cell response to the M. tuberculosis antigen TB10.4 (EsxH). CD8+ T cells specific for the TB10.44–11 epitope are primed early during infection and account for 30–50% of lung CD8+ T cells during chronic infection. Now we have used deep sequencing to characterize the TCR repertoire of TB10.44-11-specific CD8+ T cells in the lungs of infected mice. Interestingly, TB10.44-11-specific CD8+ T cells exhibit extreme clonal expansion of certain TCRβ with common structural features, most likely because of affinity selection. Affinity selection of T cells is more important when antigen presentation is limiting. Although the lung contains numerous bacteria during infection, antigen-presentation by infected APC may be limiting, mimicking a “low antigen” state. Thus, even T cells that have the potential to mediate protection may function inefficiently because of suboptimal T cell activation.
Collapse
Affiliation(s)
- Cláudio Nunes-Alves
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Matthew G. Booty
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Program in Immunology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Stephen M. Carpenter
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Division of Infectious Disease, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Division of Infectious Disease, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Alissa C. Rothchild
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Program in Immunology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Constance J. Martin
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Danielle Desjardins
- Division of Infectious Disease, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Katherine Steblenko
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Henrik N. Kløverpris
- KwaZulu-Natal Research Institute for TB and HIV, Durban, South Africa
- Nelson Mandela School of Medicine, University of Kwa-Zulu-Natal, Durban, South Africa
- Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Rajhmun Madansein
- Nelson Mandela School of Medicine, University of Kwa-Zulu-Natal, Durban, South Africa
| | - Duran Ramsuran
- KwaZulu-Natal Research Institute for TB and HIV, Durban, South Africa
| | - Alasdair Leslie
- KwaZulu-Natal Research Institute for TB and HIV, Durban, South Africa
- Nelson Mandela School of Medicine, University of Kwa-Zulu-Natal, Durban, South Africa
| | - Margarida Correia-Neves
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Samuel M. Behar
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
23
|
Lopatina O, Yoshihara T, Nishimura T, Zhong J, Akther S, Fakhrul AAKM, Liang M, Higashida C, Sumi K, Furuhara K, Inahata Y, Huang JJ, Koizumi K, Yokoyama S, Tsuji T, Petugina Y, Sumarokov A, Salmina AB, Hashida K, Kitao Y, Hori O, Asano M, Kitamura Y, Kozaka T, Shiba K, Zhong F, Xie MJ, Sato M, Ishihara K, Higashida H. Anxiety- and depression-like behavior in mice lacking the CD157/BST1 gene, a risk factor for Parkinson's disease. Front Behav Neurosci 2014; 8:133. [PMID: 24795584 PMCID: PMC4001052 DOI: 10.3389/fnbeh.2014.00133] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 04/02/2014] [Indexed: 11/13/2022] Open
Abstract
CD157, known as bone marrow stromal cell antigen-1, is a glycosylphosphatidylinositol-anchored ADP-ribosyl cyclase that supports the survival and function of B-lymphocytes and hematopoietic or intestinal stem cells. Although CD157/Bst1 is a risk locus in Parkinson's disease (PD), little is known about the function of CD157 in the nervous system and contribution to PD progression. Here, we show that no apparent motor dysfunction was observed in young knockout (CD157 (-/-)) male mice under less aging-related effects on behaviors. CD157 (-/-) mice exhibited anxiety-related and depression-like behaviors compared with wild-type mice. These behaviors were rescued through treatment with anti-psychiatric drugs and oxytocin. CD157 was weakly expressed in the amygdala and c-Fos immunoreactivity in the amygdala was less evident in CD157 (-/-) mice than in wild-type mice. These results demonstrate for the first time that CD157 plays a role as a neuro-regulator and suggest a potential role in pre-motor symptoms in PD.
Collapse
Affiliation(s)
- Olga Lopatina
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University Kanazawa, Japan ; Core Research for Evolutional Science and Technology Tokyo, Japan ; Department of Biochemistry, Medical, Pharmaceutical and Toxicological Chemistry, Krasnoyarsk State Medical University Krasnoyarsk, Russia
| | - Toru Yoshihara
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University Kanazawa, Japan ; Advanced Science Research Center, Kanazawa University Kanazawa, Japan
| | - Tomoko Nishimura
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Jing Zhong
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Shirin Akther
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Azam A K M Fakhrul
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Mingkun Liang
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Chiharu Higashida
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University Kanazawa, Japan ; Core Research for Evolutional Science and Technology Tokyo, Japan
| | - Kohei Sumi
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Kazumi Furuhara
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Yuki Inahata
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Jian-Jung Huang
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Keita Koizumi
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Shigeru Yokoyama
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Takahiro Tsuji
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Yulia Petugina
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University Kanazawa, Japan ; Department of Biochemistry, Medical, Pharmaceutical and Toxicological Chemistry, Krasnoyarsk State Medical University Krasnoyarsk, Russia
| | - Andrei Sumarokov
- Department of Biochemistry, Medical, Pharmaceutical and Toxicological Chemistry, Krasnoyarsk State Medical University Krasnoyarsk, Russia
| | - Alla B Salmina
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University Kanazawa, Japan ; Department of Biochemistry, Medical, Pharmaceutical and Toxicological Chemistry, Krasnoyarsk State Medical University Krasnoyarsk, Russia
| | - Koji Hashida
- Core Research for Evolutional Science and Technology Tokyo, Japan ; Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences Kanazawa, Japan
| | - Yasuko Kitao
- Core Research for Evolutional Science and Technology Tokyo, Japan ; Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences Kanazawa, Japan
| | - Osamu Hori
- Core Research for Evolutional Science and Technology Tokyo, Japan ; Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences Kanazawa, Japan
| | - Masahide Asano
- Advanced Science Research Center, Kanazawa University Kanazawa, Japan
| | - Yoji Kitamura
- Advanced Science Research Center, Kanazawa University Kanazawa, Japan
| | - Takashi Kozaka
- Advanced Science Research Center, Kanazawa University Kanazawa, Japan
| | - Kazuhiro Shiba
- Advanced Science Research Center, Kanazawa University Kanazawa, Japan
| | - Fangfang Zhong
- Division of Cell Biology and Neuroscience, Department of Morphological and Physiological Sciences, Faculty of Medical Sciences, University of Fukui Fukui, Japan
| | - Min-Jue Xie
- Division of Cell Biology and Neuroscience, Department of Morphological and Physiological Sciences, Faculty of Medical Sciences, University of Fukui Fukui, Japan
| | - Makoto Sato
- Division of Cell Biology and Neuroscience, Department of Morphological and Physiological Sciences, Faculty of Medical Sciences, University of Fukui Fukui, Japan ; Research Center for Child Mental Development, University of Fukui Fukui, Japan
| | - Katsuhiko Ishihara
- Department of Immunology and Molecular Genetics, Kawasaki Medical School Kurashiki, Japan
| | - Haruhiro Higashida
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University Kanazawa, Japan ; Core Research for Evolutional Science and Technology Tokyo, Japan ; Department of Biochemistry, Medical, Pharmaceutical and Toxicological Chemistry, Krasnoyarsk State Medical University Krasnoyarsk, Russia
| |
Collapse
|
24
|
Doimo NTS, Zárate-Bladés CR, Rodrigues RF, Tefé-Silva C, Trotte MNS, Souza PRM, Soares LS, Rios WM, Floriano EM, Brandão IT, Masson AP, Coelho V, Ramos SG, Silva CL. Immunotherapy of tuberculosis with Mycobacterium leprae Hsp65 as a DNA vaccine triggers cross-reactive antibodies against mammalian Hsp60 but not pathological autoimmunity. Hum Vaccin Immunother 2014; 10:1238-43. [PMID: 24607935 DOI: 10.4161/hv.28249] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Despite substantial efforts in recent years toward the development of new vaccines and drugs against tuberculosis (TB), success has remained elusive. Immunotherapy of TB with mycobacterial Hsp65 as a DNA vaccine (DNA-hsp65) results in a reduction of systemic bacterial loads and lung tissue damage, but the high homology of Hsp65 with the mammalian protein raises concern that pathological autoimmune responses may also be triggered. We searched for autoimmune responses elicited by DNA-hsp65 immunotherapy in mice chronically infected with TB by evaluating the humoral immune response and comprehensive histopathology using stereology. Cross-reactive antibodies between mycobacterial and mammalian Hsp60/65 were detected; however, no signs of pathological autoimmunity were found up to 60 days after the end of the therapy.
Collapse
Affiliation(s)
- Nayara T S Doimo
- The Centre for Tuberculosis Research; Department of Biochemistry and Immunology; Ribeirão Preto School of Medicine; University of São Paulo; São Paulo, Brazil
| | - Carlos R Zárate-Bladés
- The Centre for Tuberculosis Research; Department of Biochemistry and Immunology; Ribeirão Preto School of Medicine; University of São Paulo; São Paulo, Brazil
| | - Rodrigo F Rodrigues
- The Centre for Tuberculosis Research; Department of Biochemistry and Immunology; Ribeirão Preto School of Medicine; University of São Paulo; São Paulo, Brazil
| | - Cristiane Tefé-Silva
- Department of Pathology; Ribeirão Preto School of Medicine; University of São Paulo; São Paulo, Brazil
| | - Marcele N S Trotte
- Veterinary Medical Department of Anatomical Pathology; State University of Rio de Janeiro; Rio de Janeiro, Brazil
| | - Patrícia R M Souza
- The Centre for Tuberculosis Research; Department of Biochemistry and Immunology; Ribeirão Preto School of Medicine; University of São Paulo; São Paulo, Brazil
| | - Luana S Soares
- The Centre for Tuberculosis Research; Department of Biochemistry and Immunology; Ribeirão Preto School of Medicine; University of São Paulo; São Paulo, Brazil
| | - Wendy M Rios
- The Centre for Tuberculosis Research; Department of Biochemistry and Immunology; Ribeirão Preto School of Medicine; University of São Paulo; São Paulo, Brazil
| | - Elaine M Floriano
- Department of Pathology; Ribeirão Preto School of Medicine; University of São Paulo; São Paulo, Brazil
| | - Izaira T Brandão
- The Centre for Tuberculosis Research; Department of Biochemistry and Immunology; Ribeirão Preto School of Medicine; University of São Paulo; São Paulo, Brazil
| | - Ana P Masson
- The Centre for Tuberculosis Research; Department of Biochemistry and Immunology; Ribeirão Preto School of Medicine; University of São Paulo; São Paulo, Brazil
| | - Verônica Coelho
- Laboratory of Immunology; Heart Institute (InCor); School of Medicine; University of São Paulo; São Paulo, Brazil; Institute for Investigation in Immunology (iii)-National Institute of Science and Technology; São Paulo, Brazil
| | - Simone G Ramos
- Department of Pathology; Ribeirão Preto School of Medicine; University of São Paulo; São Paulo, Brazil
| | - Celio L Silva
- The Centre for Tuberculosis Research; Department of Biochemistry and Immunology; Ribeirão Preto School of Medicine; University of São Paulo; São Paulo, Brazil
| |
Collapse
|
25
|
de Meis J, Savino W. Mature peripheral T cells are important to preserve thymus function and selection of thymocytes during Mycobacterium tuberculosis infection. Immunotherapy 2014; 5:573-6. [PMID: 23725281 DOI: 10.2217/imt.13.41] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Evaluation of: Nobrega C, Nunes-Alves C, Cerqueira-Rodrigues B et al. T cells home to the thymus and control infection. J. Immunol. 190, 1646-1658 (2013). It is well documented that the thymus is a target organ for a large variety of pathogens (virus, bacteria, fungi and protozoa). Moreover, the presence of pathogen-derived antigens in the thymus of infected mice seems to interfere with the capacity of mature T cells to respond to the invading organism. In this way, Nobrega and colleagues demonstrated in 2010 that Mycobacterium avium infection in the thymus leads to the appearance of differentiated T cells tolerogenic for bacterial antigens. In the present and elegant study, the same group demonstrates that T-cell recirculation from the periphery to the thymus is a mechanism that allows the immune system to respond to thymic infection. A Mycobacterium-infected thymus increases the production of Th1-effector chemokines, such as CXCL9 and CXCL10, which in turn recruit CXCR3(+) peripheral T cells involved in intrathymic bacterial control. Taken together, these findings may represent an important issue of the host response, in terms of different pathogens able to infect the thymus.
Collapse
Affiliation(s)
- Juliana de Meis
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Manguinhos, Rio de Janeiro, Brazil
| | | |
Collapse
|
26
|
Tolerance has its limits: how the thymus copes with infection. Trends Immunol 2013; 34:502-10. [PMID: 23871487 DOI: 10.1016/j.it.2013.06.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 05/25/2013] [Accepted: 06/17/2013] [Indexed: 01/11/2023]
Abstract
The thymus is required for T cell differentiation; a process that depends on which antigens are encountered by thymocytes, the environment surrounding the differentiating cells, and the thymic architecture. These features are altered by local infection of the thymus and by the inflammatory mediators that accompany systemic infection. Although once believed to be an immune privileged site, it is now known that antimicrobial responses are recruited to the thymus. Resolving infection in the thymus is important because chronic persistence of microbes impairs the differentiation of pathogen-specific T cells and diminishes resistance to infection. Understanding how these mechanisms contribute to disease susceptibility, particularly in infants with developing T cell repertoires, requires further investigation.
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Maintenance of T-cell function and modulation of tolerance are critical issues in organ transplantation. The thymus is the primary organ for T-cell generation, and a preserved thymic function is essential for a self-tolerant diverse T-cell repertoire. Transplant procedures and related immunosuppressive drugs may hinder thymic integrity and function. We review here the recent advances in understanding the regulation of the unique thymic microenvironment with relevance for the field of transplantation. RECENT FINDINGS Recent studies have assigned a role for IL-22 in the regeneration of thymic epithelium, and for microRNAs in the modulation of its survival and function. The interplay of key molecules in the cross-talk between thymic epithelial cells and thymocytes was depicted, opening new perspectives for the in-vitro recapitulation of T-cell development and for thymic transplantation. Additionally, the thymus was shown to be able to sustain thymocyte progenitor renewal. SUMMARY These findings open new venues of research toward therapeutic interventions in the endogenous thymus to modulate or reconstitute the immune system; thymic transplantation; and the future development of artificial thymus, which would represent an important tool to achieve tolerance across the histocompatibility barriers.
Collapse
|
28
|
Hofmann M, Oschowitzer A, Kurzhals SR, Krüger CC, Pircher H. Thymus-resident memory CD8+ T cells mediate local immunity. Eur J Immunol 2013; 43:2295-304. [PMID: 23715993 DOI: 10.1002/eji.201343519] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 04/23/2013] [Accepted: 05/23/2013] [Indexed: 01/24/2023]
Abstract
The thymus is a primary lymphoid organ responsible for production and selection of T cells. Nonetheless, mature T cells and in particular activated T cells can reenter the thymus. Here, we identified memory CD8(+) T cells specific for lymphocytic choriomeningitis virus or vaccinia virus in the thymus of mice long-time after the infection. CD8(+) T cells were mainly located in the thymic medulla, but also in the cortical areas. Interestingly, virus-specific memory CD8(+) T cells in the thymus expressed the cell surface markers CD69 and CD103 that are characteristic of tissue-resident memory T cells in a time-dependent manner. Kinetic analyses and selective depletion of peripheral CD8(+) T cells by antibodies further revealed that thymic virus-specific memory CD8(+) T cells did not belong to the circulating pool of lymphocytes. Finally, we demonstrate that these thymus-resident virus-specific memory CD8(+) T cells efficiently mounted a secondary proliferative response, exhibited immediate effector functions and were able to protect the thymus from lymphocytic choriomeningitis virus reinfection. In conclusion, the present study not only describes for the first time virus-specific memory CD8(+) T cells with characteristics of tissue-resident memory T (T(RM)) cells in a primary lymphoid organ but also extends our knowledge about local T-cell immunity in the thymus.
Collapse
Affiliation(s)
- Maike Hofmann
- Department of Immunology, Institute of Medical Microbiology and Hygiene, University of Freiburg, Freiburg, Germany
| | | | | | | | | |
Collapse
|