Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 25(5): 886-901, 2024 May.
Article in English | MEDLINE | ID: mdl-38609547

ABSTRACT

Intestinal immune responses to microbes are controlled by the cytokine IL-10 to avoid immune pathology. Here, we use single-cell RNA sequencing of colon lamina propria leukocytes (LPLs) along with RNA-seq and ATAC-seq of purified CD4+ T cells to show that the transcription factors Blimp-1 (encoded by Prdm1) and c-Maf co-dominantly regulate Il10 while negatively regulating proinflammatory cytokines in effector T cells. Double-deficient Prdm1fl/flMaffl/flCd4Cre mice infected with Helicobacter hepaticus developed severe colitis with an increase in TH1/NK/ILC1 effector genes in LPLs, while Prdm1fl/flCd4Cre and Maffl/flCd4Cre mice exhibited moderate pathology and a less-marked type 1 effector response. LPLs from infected Maffl/flCd4Cre mice had increased type 17 responses with increased Il17a and Il22 expression and an increase in granulocytes and myeloid cell numbers, resulting in increased T cell-myeloid-neutrophil interactions. Genes over-expressed in human inflammatory bowel disease showed differential expression in LPLs from infected mice in the absence of Prdm1 or Maf, revealing potential mechanisms of human disease.


Subject(s)
Colitis , Helicobacter hepaticus , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins c-maf , Animals , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Mice , Proto-Oncogene Proteins c-maf/genetics , Colitis/immunology , Colitis/genetics , Humans , Helicobacter hepaticus/immunology , Helicobacter Infections/immunology , Mice, Inbred C57BL , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/microbiology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/genetics , Gene Expression Regulation , Disease Models, Animal
2.
Annu Rev Immunol ; 31: 475-527, 2013.
Article in English | MEDLINE | ID: mdl-23516984

ABSTRACT

There are 9 million cases of active tuberculosis reported annually; however, an estimated one-third of the world's population is infected with Mycobacterium tuberculosis and remains asymptomatic. Of these latent individuals, only 5-10% will develop active tuberculosis disease in their lifetime. CD4(+) T cells, as well as the cytokines IL-12, IFN-γ, and TNF, are critical in the control of Mycobacterium tuberculosis infection, but the host factors that determine why some individuals are protected from infection while others go on to develop disease are unclear. Genetic factors of the host and of the pathogen itself may be associated with an increased risk of patients developing active tuberculosis. This review aims to summarize what we know about the immune response in tuberculosis, in human disease, and in a range of experimental models, all of which are essential to advancing our mechanistic knowledge base of the host-pathogen interactions that influence disease outcome.


Subject(s)
Tuberculosis, Pulmonary/immunology , Animals , Disease Models, Animal , Disease Progression , Genetic Predisposition to Disease , Histocompatibility Antigens Class II/immunology , Humans , Mycobacterium tuberculosis/immunology , Treatment Outcome , Tuberculosis, Pulmonary/diagnosis , Tuberculosis, Pulmonary/epidemiology
3.
Nat Immunol ; 21(4): 464-476, 2020 04.
Article in English | MEDLINE | ID: mdl-32205882

ABSTRACT

Although mouse infection models have been extensively used to study the host response to Mycobacterium tuberculosis, their validity in revealing determinants of human tuberculosis (TB) resistance and disease progression has been heavily debated. Here, we show that the modular transcriptional signature in the blood of susceptible mice infected with a clinical isolate of M. tuberculosis resembles that of active human TB disease, with dominance of a type I interferon response and neutrophil activation and recruitment, together with a loss in B lymphocyte, natural killer and T cell effector responses. In addition, resistant but not susceptible strains of mice show increased lung B cell, natural killer and T cell effector responses in the lung upon infection. Notably, the blood signature of active disease shared by mice and humans is also evident in latent TB progressors before diagnosis, suggesting that these responses both predict and contribute to the pathogenesis of progressive M. tuberculosis infection.


Subject(s)
Transcriptome/immunology , Tuberculosis/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/microbiology , Humans , Interferon Type I/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/microbiology , Lung/immunology , Lung/microbiology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mycobacterium tuberculosis/immunology , T-Lymphocytes/immunology , T-Lymphocytes/microbiology , Tuberculosis/microbiology
4.
Nat Immunol ; 20(3): 373, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30728492

ABSTRACT

In the version of this article initially published, a source of funding was not included in the Acknowledgements section. That section should include the following: P.J.M.O. was supported by EU FP7 PREPARE project 602525. The error has been corrected in the HTML and PDF version of the article.

5.
Nat Immunol ; 20(3): 374, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30733606

ABSTRACT

In the version of this article initially published, the Supplementary Data file was an incorrect version. The correct version is now provided. The error has been corrected in the HTML and PDF version of the article.

6.
Nat Immunol ; 19(11): 1159-1168, 2018 11.
Article in English | MEDLINE | ID: mdl-30333612

ABSTRACT

Blood transcriptomics analysis of tuberculosis has revealed an interferon-inducible gene signature that diminishes in expression after successful treatment; this promises improved diagnostics and treatment monitoring, which are essential for the eradication of tuberculosis. Sensitive radiography revealing lung abnormalities and blood transcriptomics have demonstrated heterogeneity in patients with active tuberculosis and exposed asymptomatic people with latent tuberculosis, suggestive of a continuum of infection and immune states. Here we describe the immune response to infection with Mycobacterium tuberculosis revealed through the use of transcriptomics, as well as differences among clinical phenotypes of infection that might provide information on temporal changes in host immunity associated with evolving infection. We also review the diverse blood transcriptional signatures, composed of small sets of genes, that have been proposed for the diagnosis of tuberculosis and the identification of at-risk asymptomatic people and suggest novel approaches for the development of such biomarkers for clinical use.


Subject(s)
Biomarkers/blood , Gene Expression Profiling/methods , Tuberculosis/immunology , Humans , Transcriptome/immunology , Tuberculosis/blood , Tuberculosis/diagnosis
7.
Nat Immunol ; 19(6): 625-635, 2018 06.
Article in English | MEDLINE | ID: mdl-29777224

ABSTRACT

Transcriptional profiles and host-response biomarkers are used increasingly to investigate the severity, subtype and pathogenesis of disease. We now describe whole-blood mRNA signatures and concentrations of local and systemic immunological mediators in 131 adults hospitalized with influenza, from whom extensive clinical and investigational data were obtained by MOSAIC investigators. Signatures reflective of interferon-related antiviral pathways were common up to day 4 of symptoms in patients who did not require mechanical ventilator support; in those who needed mechanical ventilation, an inflammatory, activated-neutrophil and cell-stress or death ('bacterial') pattern was seen, even early in disease. Identifiable bacterial co-infection was not necessary for this 'bacterial' signature but was able to enhance its development while attenuating the early 'viral' signature. Our findings emphasize the importance of timing and severity in the interpretation of host responses to acute viral infection and identify specific patterns of immune-system activation that might enable the development of novel diagnostic and therapeutic tools for severe influenza.


Subject(s)
Biomarkers/blood , Influenza, Human/blood , Influenza, Human/immunology , Transcriptome , Adolescent , Adult , Aged , Disease Progression , Female , Humans , Influenza, Human/genetics , Interferons/blood , Interferons/immunology , Male , Middle Aged , Neutrophils/immunology , RNA, Messenger/blood , Young Adult
8.
Nat Immunol ; 19(5): 497-507, 2018 05.
Article in English | MEDLINE | ID: mdl-29662170

ABSTRACT

The transcription factor c-Maf induces the anti-inflammatory cytokine IL-10 in CD4+ T cells in vitro. However, the global effects of c-Maf on diverse immune responses in vivo are unknown. Here we found that c-Maf regulated IL-10 production in CD4+ T cells in disease models involving the TH1 subset of helper T cells (malaria), TH2 cells (allergy) and TH17 cells (autoimmunity) in vivo. Although mice with c-Maf deficiency targeted to T cells showed greater pathology in TH1 and TH2 responses, TH17 cell-mediated pathology was reduced in this context, with an accompanying decrease in TH17 cells and increase in Foxp3+ regulatory T cells. Bivariate genomic footprinting elucidated the c-Maf transcription-factor network, including enhanced activity of NFAT; this led to the identification and validation of c-Maf as a negative regulator of IL-2. The decreased expression of the gene encoding the transcription factor RORγt (Rorc) that resulted from c-Maf deficiency was dependent on IL-2, which explained the in vivo observations. Thus, c-Maf is a positive and negative regulator of the expression of cytokine-encoding genes, with context-specific effects that allow each immune response to occur in a controlled yet effective manner.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Gene Expression Regulation/immunology , Gene Regulatory Networks/immunology , Interleukin-2/biosynthesis , Proto-Oncogene Proteins c-maf/immunology , Animals , Interleukin-2/immunology , Mice
9.
Immunity ; 50(4): 871-891, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995504

ABSTRACT

Cytokines are among the most important effector and messenger molecules in the immune system. They profoundly participate in immune responses during infection and inflammation, protecting against or contributing to diseases such as allergy, autoimmunity, and cancer. Manipulating cytokine pathways, therefore, is one of the most effective strategies to treat various diseases. IL-10 family cytokines exert essential functions to maintain tissue homeostasis during infection and inflammation through restriction of excessive inflammatory responses, upregulation of innate immunity, and promotion of tissue repairing mechanisms. Their important functions in diseases are supported by data from many preclinical models, human genetic studies, and clinical interventions. Despite significant efforts, however, there is still no clinically approved therapy through manipulating IL-10 family cytokines. Here, we summarize the recent progress in understanding the biology of this family of cytokines, suggesting more specific strategies to maneuver these cytokines for the effective treatment of inflammatory diseases and cancers.


Subject(s)
Immunity, Innate , Interleukin-10/immunology , Interleukins/immunology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Cytokines/classification , Cytokines/genetics , Gene Expression Regulation , Humans , Infections/immunology , Infections/therapy , Inflammation/immunology , Inflammation/therapy , Interleukin-10/genetics , Interleukins/genetics , Lymphocyte Subsets/immunology , Mice , Multigene Family , Myeloid Cells/immunology , Neoplasms/immunology , Neoplasms/therapy , Signal Transduction , Transcription Factors/physiology , Interleukin-22
10.
Nat Immunol ; 13(10): 925-31, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22990890

ABSTRACT

Feedback regulatory circuits provided by regulatory T cells (T(reg) cells) and suppressive cytokines are an intrinsic part of the immune system, along with effector functions. Here we discuss some of the regulatory cytokines that have evolved to permit tolerance to components of self as well as the eradication of pathogens with minimal collateral damage to the host. Interleukin 2 (IL-2), IL-10 and transforming growth factor-ß (TGF-ß) are well characterized, whereas IL-27, IL-35 and IL-37 represent newcomers to the spectrum of anti-inflammatory cytokines. We also emphasize how information accumulated through in vitro as well as in vivo studies of genetically engineered mice can help in the understanding and treatment of human diseases.


Subject(s)
Anti-Inflammatory Agents/immunology , Cytokines/immunology , Inflammation/immunology , Interleukins/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Anti-Inflammatory Agents/metabolism , Cytokines/metabolism , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Interleukin-1/immunology , Interleukin-10/immunology , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukin-2/immunology , Interleukins/metabolism , Mice , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism
11.
Nat Immunol ; 12(4): 288-94, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21423225

ABSTRACT

In recent years, numerous qualitative discoveries have been made in immunology research. However, the effect of quantitative events, long recognized as the driving factors for determinism in developmental biology, that dictate the quality of the immune response elicited to an antigen in concert with microbial products still requires serious attention. Here we discuss how the often-neglected issue of quantification affects the specification, differentiation and commitment of helper T cells. As reductionist in vitro approaches have been instrumental in the elucidation of the factors determining the development of helper T cells, in this perspective we highlight the need for the standardization of protocols, also fundamental for the comparison of immune responses in mice and humans. Improving understanding of how these in vitro quantitative events translate to immune responses in vivo, which can be studied in mouse models, is of importance in obtaining information on immune responses in humans, thus empowering translational research.


Subject(s)
Cell Differentiation/immunology , Models, Animal , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cytokines/immunology , Cytokines/metabolism , Humans , Mice , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Translational Research, Biomedical/methods , Translational Research, Biomedical/standards
12.
J Allergy Clin Immunol ; 147(5): 1892-1906, 2021 05.
Article in English | MEDLINE | ID: mdl-33571538

ABSTRACT

BACKGROUND: Early life represents a major risk window for asthma development. However, the mechanisms controlling the threshold for establishment of allergic airway inflammation in early life are incompletely understood. Airway macrophages (AMs) regulate pulmonary allergic responses and undergo TGF-ß-dependent postnatal development, but the role of AM maturation factors such as TGF-ß in controlling the threshold for pathogenic immune responses to inhaled allergens remains unclear. OBJECTIVE: Our aim was to test the hypothesis that AM-derived TGF-ß1 regulates pathogenic immunity to inhaled allergen in early life. METHODS: Conditional knockout (Tgfb1ΔCD11c) mice, with TGF-ß1 deficiency in AMs and other CD11c+ cells, were analyzed throughout early life and following neonatal house dust mite (HDM) inhalation. The roles of specific chemokine receptors were determined by using in vivo blocking antibodies. RESULTS: AM-intrinsic TGF-ß1 was redundant for initial population of the neonatal lung with AMs, but AMs from Tgfb1ΔCD11c mice failed to adopt a mature homeostatic AM phenotype in the first weeks of life. Evidence of constitutive TGF-ß1 signaling was also observed in pediatric human AMs. TGF-ß1-deficient AMs expressed enhanced levels of monocyte-attractant chemokines, and accordingly, Tgfb1ΔCD11c mice exposed to HDM throughout early life accumulated CCR2-dependent inflammatory CD11c+ mononuclear phagocytes into the airway niche that expressed the proallergic chemokine CCL8. Tgfb1ΔCD11c mice displayed augmented TH2, group 2 innate lymphoid cell, and airway remodeling responses to HDM, which were ameliorated by blockade of the CCL8 receptor CCR8. CONCLUSION: Our results highlight a causal relationship between AM maturity, chemokines, and pathogenic immunity to environmental stimuli in early life and identify TGF-ß1 as a key regulator of this.


Subject(s)
Allergens/immunology , Macrophages, Alveolar/immunology , Transforming Growth Factor beta1/immunology , Animals , Chemokines/immunology , Hypersensitivity/immunology , Lung/immunology , Mice, Inbred C57BL , Mice, Knockout , Pyroglyphidae/immunology , Transforming Growth Factor beta1/genetics
13.
Nat Immunol ; 10(9): 929-32, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19692989

ABSTRACT

The authors recount their discovery of how pathogen-induced interleukin 12 production leads to T(H)1 T cell polarization. Simultaneously they discovered the suppressive cytokine interleukin 10 inhibits antigen-presenting cells, thus regulating development of T(H)1 cells.


Subject(s)
Antigen-Presenting Cells/physiology , Interleukin-10/physiology , Interleukin-12/physiology , Th1 Cells/physiology , Animals , Cell Differentiation , Cell Polarity , Humans , Interferon-gamma/biosynthesis , Macrophages/physiology , Th2 Cells/physiology
14.
J Immunol ; 202(3): 684-693, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30598515

ABSTRACT

Glucocorticoids are known to increase production of the anti-inflammatory cytokine IL-10, and this action is associated with their clinical efficacy in asthmatics. However, glucocorticoids also enhance the synthesis of IL-17A by PBMCs, which, in excess, is associated with increased asthma severity and glucocorticoid-refractory disease. In this study, we show that the glucocorticoid dexamethasone significantly increased IL-10 production by human memory CD4+ T cells from healthy donors, as assessed by intracellular cytokine staining. In addition, dexamethasone increased production of IL-17A, IL-17F, and IL-22, with the most striking enhancement in cells coproducing Th17-associated cytokines together with IL-10. Of note, an increase in IFN-γ+IL-10+ cells was also observed despite overall downregulation of IFN-γ production. These dexamethasone-driven IL-10+ cells, and predominantly the IL-17+IL-10+ double-producing cells, were markedly refractory to the inhibitory effect of dexamethasone on proliferation and IL-2Rα expression, which facilitated their preferential IL-2-dependent expansion. Although lower concentrations of exogenous IL-2 promoted IL-10+ cells coproducing proinflammatory cytokines, higher IL-2 doses, both alone and in combination with dexamethasone, increased the proportion of single IL-10+ T cells. Thus, glucocorticoid-induced IL-10 is only accompanied by an increase of IL-17 in a low IL-2 setting, which is, nevertheless, likely to be protective owing to the induction of regulatory IL-17+IL-10+-coproducing cells. These findings open new avenues of investigation with respect to the role of IL-2 in glucocorticoid responsiveness that have potential implications for optimizing the benefit/risk ratio of glucocorticoids in the clinic.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Immunologic Memory , Interleukin-10/immunology , Interleukin-17/immunology , Interleukin-2/pharmacology , Cell Proliferation , Female , Humans , Male , Phenotype
15.
Proc Natl Acad Sci U S A ; 115(5): E964-E973, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29339504

ABSTRACT

The transition between latent and active tuberculosis (TB) occurs before symptom onset. Better understanding of the early events in subclinical disease will facilitate the development of diagnostics and interventions that improve TB control. This is particularly relevant in the context of HIV-1 coinfection where progression of TB is more likely. In a recent study using [18F]-fluoro-2-deoxy-d-glucose positron emission/computed tomography (FDG-PET/CT) on 35 asymptomatic, HIV-1-infected adults, we identified 10 participants with radiographic evidence of subclinical disease, significantly more likely to progress than the 25 participants without. To gain insight into the biological events in early disease, we performed blood-based whole genome transcriptomic analysis on these participants and 15 active patients with TB. We found transcripts representing the classical complement pathway and Fcγ receptor 1 overabundant from subclinical stages of disease. Levels of circulating immune (antibody/antigen) complexes also increased in subclinical disease and were highly correlated with C1q transcript abundance. To validate our findings, we analyzed transcriptomic data from a publicly available dataset where samples were available in the 2 y before TB disease presentation. Transcripts representing the classical complement pathway and Fcγ receptor 1 were also differentially expressed in the 12 mo before disease presentation. Our results indicate that levels of antibody/antigen complexes increase early in disease, associated with increased gene expression of C1q and Fcγ receptors that bind them. Understanding the role this plays in disease progression may facilitate development of interventions that prevent this, leading to a more favorable outcome and may also be important to diagnostic development.


Subject(s)
Antigen-Antibody Complex/blood , Complement System Proteins/genetics , HIV Infections/immunology , Tuberculosis/immunology , Antibodies/blood , Cluster Analysis , Coinfection , Comorbidity , Disease Progression , Fluorodeoxyglucose F18 , HIV Infections/complications , Humans , Interferons/immunology , Oligonucleotide Array Sequence Analysis , Positron Emission Tomography Computed Tomography , Signal Transduction , Transcription, Genetic , Transcriptional Activation , Transcriptome , Tuberculosis/complications
16.
J Allergy Clin Immunol ; 145(2): 666-678.e9, 2020 02.
Article in English | MEDLINE | ID: mdl-31445933

ABSTRACT

BACKGROUND: Although originally defined as a type 2 (T2) immune-mediated condition, non-T2 cytokines, such as IFN-γ and IL-17A, have been implicated in asthma pathogenesis, particularly in patients with severe disease. IL-10 regulates TH cell phenotypes and can dampen T2 immunity to allergens, but its functions in controlling non-T2 cytokine responses in asthmatic patients are unclear. OBJECTIVE: We sought to determine how IL-10 regulates the balance of TH cell responses to inhaled allergen. METHODS: Allergic airway disease was induced in wild-type, IL-10 reporter, and conditional IL-10 or IL-10 receptor α (IL-10Rα) knockout mice by means of repeated intranasal administration of house dust mite (HDM). IL-10 and IFN-γ signaling were disrupted by using blocking antibodies. RESULTS: Repeated HDM inhalation induced a mixed IL-13/IL-17A response and accumulation of IL-10-producing forkhead box P3-negative effector CD4+ T cells in the lungs. Ablation of T cell-derived IL-10 increased the IFN-γ and IL-17A response to HDM, reducing IL-13 levels and airway eosinophilia without affecting IgE levels or airway hyperresponsiveness. The increased IFN-γ response could be recapitulated by IL-10Rα deletion in CD11c+ myeloid cells or local IL-10Rα blockade. Disruption of the T cell-myeloid IL-10 axis resulted in increased pulmonary monocyte-derived dendritic cell numbers and increased IFN-γ-dependent expression of CXCR3 ligands by airway macrophages, which is suggestive of a feedforward loop of TH1 cell recruitment. Augmented IFN-γ responses in the HDM allergic airway disease model were accompanied by increased disruption of airway epithelium, which was reversed by therapeutic blockade of IFN-γ. CONCLUSIONS: IL-10 from effector T cells signals to CD11c+ myeloid cells to suppress an atypical and pathogenic IFN-γ response to inhaled HDM.


Subject(s)
Asthma/immunology , Interferon-gamma/immunology , Interleukin-10/immunology , Myeloid Cells/immunology , T-Lymphocytes, Helper-Inducer/immunology , Allergens/immunology , Animals , Disease Models, Animal , Female , Hypersensitivity/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyroglyphidae/immunology
18.
J Immunol ; 199(2): 613-623, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28584007

ABSTRACT

Tuberculosis (TB), caused by Mycobacterium tuberculosis infection, is a leading cause of mortality and morbidity, causing ∼1.5 million deaths annually. CD4+ T cells and several cytokines, such as the Th1 cytokine IFN-γ, are critical in the control of this infection. Conversely, the immunosuppressive cytokine IL-10 has been shown to dampen Th1 cell responses to M. tuberculosis infection impairing bacterial clearance. However, the critical cellular source of IL-10 during M. tuberculosis infection is still unknown. Using IL-10 reporter mice, we show in this article that during the first 14 d of M. tuberculosis infection, the predominant cells expressing IL-10 in the lung were Ly6C+ monocytes. However, after day 21 postinfection, IL-10-expressing T cells were also highly represented. Notably, mice deficient in T cell-derived IL-10, but not mice deficient in monocyte-derived IL-10, showed a significant reduction in lung bacterial loads during chronic M. tuberculosis infection compared with fully IL-10-competent mice, indicating a major role for T cell-derived IL-10 in TB susceptibility. IL-10-expressing cells were detected among both CD4+ and CD8+ T cells, expressed high levels of CD44 and Tbet, and were able to coproduce IFN-γ and IL-10 upon ex vivo stimulation. Furthermore, during M. tuberculosis infection, Il10 expression in CD4+ T cells was partially regulated by both IL-27 and type I IFN signaling. Together, our data reveal that, despite the multiple immune sources of IL-10 during M. tuberculosis infection, activated effector T cells are the major source accounting for IL-10-induced TB susceptibility.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Interleukin-10/immunology , Tuberculosis/immunology , Animals , Antigens, Ly/immunology , Hyaluronan Receptors/genetics , Hyaluronan Receptors/immunology , Interferon Type I/immunology , Interferon Type I/metabolism , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-10/biosynthesis , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukins/immunology , Interleukins/metabolism , Mice , Monocytes/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/microbiology
19.
Am J Respir Crit Care Med ; 197(6): 801-813, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29161093

ABSTRACT

RATIONALE: In addition to their well-known function as antibody-producing cells, B lymphocytes can markedly influence the course of infectious or noninfectious diseases via antibody-independent mechanisms. In tuberculosis (TB), B cells accumulate in lungs, yet their functional contribution to the host response remains poorly understood. OBJECTIVES: To document the role of B cells in TB in an unbiased manner. METHODS: We generated the transcriptome of B cells isolated from Mycobacterium tuberculosis (Mtb)-infected mice and validated the identified key pathways using in vitro and in vivo assays. The obtained data were substantiated using B cells from pleural effusion of patients with TB. MEASUREMENTS AND MAIN RESULTS: B cells isolated from Mtb-infected mice displayed a STAT1 (signal transducer and activator of transcription 1)-centered signature, suggesting a role for IFNs in B-cell response to infection. B cells stimulated in vitro with Mtb produced type I IFN, via a mechanism involving the innate sensor STING (stimulator of interferon genes), and antagonized by MyD88 (myeloid differentiation primary response 88) signaling. In vivo, B cells expressed type I IFN in the lungs of Mtb-infected mice and, of clinical relevance, in pleural fluid from patients with TB. Type I IFN expression by B cells induced an altered polarization of macrophages toward a regulatory/antiinflammatory profile in vitro. In vivo, increased provision of type I IFN by B cells in a murine model of B cell-restricted Myd88 deficiency correlated with an enhanced accumulation of regulatory/antiinflammatory macrophages in Mtb-infected lungs. CONCLUSIONS: Type I IFN produced by Mtb-stimulated B cells favors macrophage polarization toward a regulatory/antiinflammatory phenotype during Mtb infection.


Subject(s)
B-Lymphocytes/metabolism , Interferon Type I/metabolism , Macrophages/metabolism , Tuberculosis/metabolism , Animals , Disease Models, Animal , Humans , Lung/metabolism , Lung/microbiology , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis , Signal Transduction , Spleen/metabolism , Spleen/microbiology
20.
Immunol Rev ; 264(1): 88-102, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25703554

ABSTRACT

The immune response upon infection with the pathogen Mycobacterium tuberculosis is poorly understood, hampering the discovery of new treatments and the improvements in diagnosis. In the last years, a blood transcriptional signature in tuberculosis has provided knowledge on the immune response occurring during active tuberculosis disease. This signature was absent in the majority of asymptomatic individuals who are latently infected with M. tuberculosis (referred to as latent). Using modular and pathway analyses of the complex data has shown, now in multiple studies, that the signature of active tuberculosis is dominated by overexpression of interferon-inducible genes (consisting of both type I and type II interferon signaling), myeloid genes, and inflammatory genes. There is also downregulation of genes encoding B and T-cell function. The blood signature of tuberculosis correlates with the extent of radiographic disease and is diminished upon effective treatment suggesting the possibility of new improved strategies to support diagnostic assays and methods for drug treatment monitoring. The signature suggested a previously under-appreciated role for type I interferons in development of active tuberculosis disease, and numerous mechanisms have now been uncovered to explain how type I interferon impedes the protective response to M. tuberculosis infection.


Subject(s)
Host-Pathogen Interactions/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Tuberculosis/therapy , Animals , Gene Expression Profiling , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Humans , Prognosis , Transcriptome , Treatment Outcome , Tuberculosis/blood , Tuberculosis/diagnosis , Tuberculosis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL