Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nat Commun ; 11(1): 5566, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33149141

ABSTRACT

Tuberculosis (TB) is a leading cause of mortality due to infectious disease, but the factors determining disease progression are unclear. Transcriptional signatures associated with type I IFN signalling and neutrophilic inflammation were shown to correlate with disease severity in mouse models of TB. Here we show that similar transcriptional signatures correlate with increased bacterial loads and exacerbate pathology during Mycobacterium tuberculosis infection upon GM-CSF blockade. Loss of GM-CSF signalling or genetic susceptibility to TB (C3HeB/FeJ mice) result in type I IFN-induced neutrophil extracellular trap (NET) formation that promotes bacterial growth and promotes disease severity. Consistently, NETs are present in necrotic lung lesions of TB patients responding poorly to antibiotic therapy, supporting the role of NETs in a late stage of TB pathogenesis. Our findings reveal an important cytokine-based innate immune effector network with a central role in determining the outcome of M. tuberculosis infection.


Subject(s)
Extracellular Traps/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interferon Type I/metabolism , Lung/microbiology , Mycobacterium tuberculosis/immunology , Neutrophils/immunology , Pneumonia/immunology , Tuberculosis, Pulmonary/immunology , Animals , Databases, Genetic , Disease Progression , Gene Expression Profiling , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Interferon Type I/genetics , Interferon-gamma/genetics , Interferon-gamma/metabolism , Lung/immunology , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis/pathogenicity , Pneumonia/genetics , Pneumonia/metabolism , Pneumonia/pathology , RNA-Seq , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Tuberculosis, Pulmonary/blood , Tuberculosis, Pulmonary/genetics , Tuberculosis, Pulmonary/microbiology
2.
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
3.
Nat Commun ; 10(1): 2887, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31253760

ABSTRACT

Understanding how immune challenges elicit different responses is critical for diagnosing and deciphering immune regulation. Using a modular strategy to interpret the complex transcriptional host response in mouse models of infection and inflammation, we show a breadth of immune responses in the lung. Lung immune signatures are dominated by either IFN-γ and IFN-inducible, IL-17-induced neutrophil- or allergy-associated gene expression. Type I IFN and IFN-γ-inducible, but not IL-17- or allergy-associated signatures, are preserved in the blood. While IL-17-associated genes identified in lung are detected in blood, the allergy signature is only detectable in blood CD4+ effector cells. Type I IFN-inducible genes are abrogated in the absence of IFN-γ signaling and decrease in the absence of IFNAR signaling, both independently contributing to the regulation of granulocyte responses and pathology during Toxoplasma gondii infection. Our framework provides an ideal tool for comparative analyses of transcriptional signatures contributing to protection or pathogenesis in disease.


Subject(s)
Candidiasis/metabolism , Interferon Type I/metabolism , Interferon-gamma/metabolism , Melioidosis/metabolism , Orthomyxoviridae Infections/metabolism , Respiratory Syncytial Virus Infections/metabolism , Animals , Burkholderia pseudomallei , Candida albicans , Candidiasis/immunology , Candidiasis/microbiology , Gene Expression Regulation/immunology , Influenza A Virus, H3N2 Subtype , Interferon Type I/blood , Interferon Type I/genetics , Interferon-gamma/blood , Interferon-gamma/genetics , Lung , Melioidosis/immunology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Receptor, Interferon alpha-beta , Receptors, Interferon , Respiratory Syncytial Virus Infections/immunology , Interferon gamma Receptor
4.
J Exp Med ; 215(5): 1273-1285, 2018 05 07.
Article in English | MEDLINE | ID: mdl-29666166

ABSTRACT

Tuberculosis remains one of the leading causes of mortality worldwide, and, despite its clinical significance, there are still significant gaps in our understanding of pathogenic and protective mechanisms triggered by Mycobacterium tuberculosis infection. Type I interferons (IFN) regulate a broad family of genes that either stimulate or inhibit immune function, having both host-protective and detrimental effects, and exhibit well-characterized antiviral activity. Transcriptional studies have uncovered a potential deleterious role for type I IFN in active tuberculosis. Since then, additional studies in human tuberculosis and experimental mouse models of M. tuberculosis infection support the concept that type I IFN promotes both bacterial expansion and disease pathogenesis. More recently, studies in a different setting have suggested a putative protective role for type I IFN. In this study, we discuss the mechanistic and contextual factors that determine the detrimental versus beneficial outcomes of type I IFN induction during M. tuberculosis infection, from human disease to experimental mouse models of tuberculosis.


Subject(s)
Interferon Type I/metabolism , Tuberculosis/metabolism , Animals , Disease Models, Animal , Humans , Immunity , Models, Biological , Tuberculosis/immunology , Tuberculosis/pathology
5.
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
6.
J Immunol ; 197(12): 4714-4726, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27849167

ABSTRACT

Tuberculosis causes ∼1.5 million deaths every year, thus remaining a leading cause of death from infectious diseases in the world. A growing body of evidence demonstrates that type I IFN plays a detrimental role in tuberculosis pathogenesis, likely by interfering with IFN-γ-dependent immunity. In this article, we reveal a novel mechanism by which type I IFN may confer protection against Mycobacterium tuberculosis infection in the absence of IFN-γ signaling. We show that production of type I IFN by M. tuberculosis-infected macrophages induced NO synthase 2 and inhibited arginase 1 gene expression. In vivo, absence of both type I and type II IFN receptors led to strikingly increased levels of arginase 1 gene expression and protein activity in infected lungs, characteristic of alternatively activated macrophages. This correlated with increased lung bacterial burden and pathology and decreased survival compared with mice deficient in either receptor. Increased expression of other genes associated with alternatively activated macrophages, as well as increased expression of Th2-associated cytokines and decreased TNF expression, were also observed. Thus, in the absence of IFN-γ signaling, type I IFN suppressed the switching of macrophages from a more protective classically activated phenotype to a more permissive alternatively activated phenotype. Together, our data support a model in which suppression of alternative macrophage activation by type I IFN during M. tuberculosis infection, in the absence of IFN-γ signaling, contributes to host protection.


Subject(s)
Interferon Type I/metabolism , Lung/immunology , Macrophages/immunology , Mycobacterium tuberculosis/immunology , Nitric Oxide Synthase Type II/metabolism , Tuberculosis, Pulmonary/immunology , Animals , Arginase/genetics , Arginase/metabolism , Bacterial Load , Cytokines/metabolism , Gene Expression Regulation , Humans , Interferon-gamma/metabolism , Lung/microbiology , Macrophage Activation , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Receptors, Interferon/genetics , Signal Transduction , Th2 Cells/immunology
7.
PLoS One ; 11(2): e0150251, 2016.
Article in English | MEDLINE | ID: mdl-26918359

ABSTRACT

Analysis of the mouse transcriptional response to Listeria monocytogenes infection reveals that a large set of genes are perturbed in both blood and tissue and that these transcriptional responses are enriched for pathways of the immune response. Further we identified enrichment for both type I and type II interferon (IFN) signaling molecules in the blood and tissues upon infection. Since type I IFN signaling has been reported widely to impair bacterial clearance we examined gene expression from blood and tissues of wild type (WT) and type I IFNαß receptor-deficient (Ifnar1-/-) mice at the basal level and upon infection with L. monocytogenes. Measurement of the fold change response upon infection in the absence of type I IFN signaling demonstrated an upregulation of specific genes at day 1 post infection. A less marked reduction of the global gene expression signature in blood or tissues from infected Ifnar1-/- as compared to WT mice was observed at days 2 and 3 after infection, with marked reduction in key genes such as Oasg1 and Stat2. Moreover, on in depth analysis, changes in gene expression in uninfected mice of key IFN regulatory genes including Irf9, Irf7, Stat1 and others were identified, and although induced by an equivalent degree upon infection this resulted in significantly lower final gene expression levels upon infection of Ifnar1-/- mice. These data highlight how dysregulation of this network in the steady state and temporally upon infection may determine the outcome of this bacterial infection and how basal levels of type I IFN-inducible genes may perturb an optimal host immune response to control intracellular bacterial infections such as L. monocytogenes.


Subject(s)
Interferon Type I/physiology , Listeriosis/immunology , Transcription, Genetic/immunology , Transcriptome , Animals , Blood Cells/metabolism , Disease Resistance , Gene Expression Regulation/immunology , Interferon-gamma/physiology , Listeriosis/genetics , Listeriosis/metabolism , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/deficiency , Signal Transduction , Spleen/metabolism , T-Lymphocyte Subsets/immunology
9.
PLoS One ; 10(7): e0131904, 2015.
Article in English | MEDLINE | ID: mdl-26135889

ABSTRACT

Sirtuins (Sirts) regulate several cellular mechanisms through deacetylation of several transcription factors and enzymes. Recently, Sirt2 was shown to prevent the development of inflammatory processes and its expression favors acute Listeria monocytogenes infection. The impact of this molecule in the context of chronic infections remains unknown. We found that specific Sirt2 deletion in the myeloid lineage transiently increased Mycobacterium tuberculosis load in the lungs and liver of conditional mice. Sirt2 did not affect long-term infection since no significant differences were observed in the bacterial burden at days 60 and 120 post-infection. The initial increase in M. tuberculosis growth was not due to differences in inflammatory cell infiltrates in the lung, myeloid or CD4+ T cells. The transcription levels of IFN-γ, IL-17, TNF, IL-6 and NOS2 were also not affected in the lungs by Sirt2-myeloid specific deletion. Overall, our results demonstrate that Sirt2 expression has a transitory effect in M. tuberculosis infection. Thus, modulation of Sirt2 activity in vivo is not expected to affect chronic infection with M. tuberculosis.


Subject(s)
Gene Expression Regulation , Mycobacterium tuberculosis/metabolism , Myeloid Cells/metabolism , Sirtuin 2/metabolism , Tuberculosis, Pulmonary/metabolism , Animals , CD4-Positive T-Lymphocytes/microbiology , Disease Models, Animal , Female , Flow Cytometry , Gene Deletion , Inflammation , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukin-6/metabolism , Liver/microbiology , Lung/microbiology , Macrophages/metabolism , Macrophages/microbiology , Male , Mice , Nitric Oxide Synthase Type II/metabolism , Real-Time Polymerase Chain Reaction , Tuberculosis, Pulmonary/microbiology , Tumor Necrosis Factor-alpha/metabolism
10.
J Immunol ; 193(7): 3600-12, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25187652

ABSTRACT

Tuberculosis, caused by the intracellular bacterium Mycobacterium tuberculosis, currently causes ∼1.4 million deaths per year, and it therefore remains a leading global health problem. The immune response during tuberculosis remains incompletely understood, particularly regarding immune factors that are harmful rather than protective to the host. Overproduction of the type I IFN family of cytokines is associated with exacerbated tuberculosis in both mouse models and in humans, although the mechanisms by which type I IFN promotes disease are not well understood. We have investigated the effect of type I IFN on M. tuberculosis-infected macrophages and found that production of host-protective cytokines such as TNF-α, IL-12, and IL-1ß is inhibited by exogenous type I IFN, whereas production of immunosuppressive IL-10 is promoted in an IL-27-independent manner. Furthermore, much of the ability of type I IFN to inhibit cytokine production was mediated by IL-10. Additionally, type I IFN compromised macrophage activation by the lymphoid immune response through severely disrupting responsiveness to IFN-γ, including M. tuberculosis killing. These findings describe important mechanisms by which type I IFN inhibits the immune response during tuberculosis.


Subject(s)
Interferon Type I/immunology , Interferon-gamma/immunology , Interleukin-10/immunology , Interleukin-12/immunology , Interleukins/immunology , Macrophages/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Animals , Interferon Type I/genetics , Interferon-gamma/genetics , Interleukin-10/genetics , Interleukin-12/genetics , Interleukin-1beta/immunology , Interleukins/genetics , Macrophage Activation/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Knockout , Tuberculosis/genetics , Tuberculosis/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
11.
PLoS One ; 9(8): e105422, 2014.
Article in English | MEDLINE | ID: mdl-25144742

ABSTRACT

Clinicians are well aware of existing pharmacologically-induced immune deficient status in kidney-transplanted patients that will favor their susceptibility to bacterial or viral infections. Previous studies indicated that advanced Stage 4-5 Chronic Kidney Disease might also be regarded as an immune deficiency-like status as well, even though the mechanisms are not fully understood. Here, we analyzed the ex vivo frequency and the functional properties of both conventional and innate-like T (ILT) lymphocyte subsets in the peripheral blood of 35 patients on hemodialysis, 29 kidney transplanted patients and 38 healthy donors. We found that peripheral blood cell count of ILT cells, as iNKT (invariant Natural Killer T) and MAIT (mucosal-associated invariant T), were significantly decreased in hemodialyzed patients compared to healthy controls. This deficiency was also observed regarding conventional T cells, including the IL-17-producing CD4(+) Th17 cells. Pertaining to regulatory T cells, we also noticed major modifications in the global frequency of CD4(+)CD25(+)Foxp3(+) T lymphocytes, including the resting suppressive CD45RA(+)Foxp3lo and activated suppressive CD45RA-Foxp3hi T cell subpopulations. We found no significant differences between the immune status of hemodialyzed and kidney-transplanted subjects. In conclusion, we demonstrated that both ILT and conventional T cell numbers are equally impaired in hemodialyzed and kidney-transplanted patients.


Subject(s)
Immunity, Innate , Kidney Transplantation , Renal Dialysis , T-Lymphocyte Subsets/immunology , Adult , Cytokines/metabolism , Female , Humans , Immunocompromised Host , Immunophenotyping , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/immunology , Kidney Failure, Chronic/therapy , Lymphocyte Count , Male , Middle Aged , Phenotype , T-Lymphocyte Subsets/metabolism , Young Adult
12.
PLoS One ; 8(6): e67277, 2013.
Article in English | MEDLINE | ID: mdl-23840651

ABSTRACT

Tuberculosis associates with a wide spectrum of disease outcomes. The Beijing (Bj) lineage of Mycobacterium tuberculosis (Mtb) is suggested to be more virulent than other Mtb lineages and prone to elicit non-protective immune responses. However, highly heterogeneous immune responses were reported upon infection of innate immune cells with Bj strains or stimulation with their glycolipids. Using both in vitro and in vivo mouse models of infection, we here report that the molecular mechanism for this heterogeneity may be related to distinct TLR activations. Among this Mtb lineage, we found strains that preferentially activate TLR2, and others that also activate TLR4. Recognition of Mtb strains by TLR4 resulted in a distinct cytokine profile in vitro and in vivo, with specific production of type I IFN. We also uncover a novel protective role for TLR4 activation in vivo. Thus, our findings contribute to the knowledge of the molecular basis underlying how host innate immune cells handle different Mtb strains, in particular the intricate host-pathogen interaction with strains of the Mtb Bj lineage.


Subject(s)
Immunity, Innate , Mycobacterium tuberculosis/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Animals , Female , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/physiology , Protein Binding , Species Specificity
13.
Blood ; 120(10): 2144-54, 2012 Sep 06.
Article in English | MEDLINE | ID: mdl-22730537

ABSTRACT

Invariant natural killer T (iNKT) cells can experimentally dissociate GVL from graft-versus-host-disease (GVHD). Their role in human conventional allogeneic hematopoietic stem cell transplantation (HSCT) is unknown. Here, we analyzed the post-HSCT recovery of iNKT cells in 71 adult allografted patients. Results were compared with conventional T- and NK-cell recovery and correlated to the occurrence of GVHD, relapse, and survival. We observed that posttransplantation iNKT cells, likely of donor origin, recovered independently of T and NK cells in the first 90 days after HSCT and reached greater levels in recipient younger than 45 years (P = .003) and after a reduced-intensity conditioning regimen (P = .03). Low posttransplantation iNKT/T ratios (ie, < 10(-3)) were an independent factor associated with the occurrence of acute GVHD (aGVHD; P = .001). Inversely, reaching iNKT/T ratios > 10(-3) before day 90 was associated with reduced nonrelapse mortality (P = .009) without increased risk of relapse and appeared as an independent predictive factor of an improved overall survival (P = .028). Furthermore, an iNKT/T ratio on day 15 > 0.58 × 10(-3) was associated with a 94% risk reduction of aGVHD. These findings provide a proof of concept that early postallogeneic HSCT iNKT cell recovery can predict the occurrence of aGVHD and an improved overall survival.


Subject(s)
Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , Natural Killer T-Cells/immunology , Transplantation Conditioning/methods , Acute Disease , Adolescent , Adult , Age Factors , Female , Graft vs Host Disease/pathology , Graft vs Host Disease/therapy , Graft vs Leukemia Effect/immunology , Hematologic Neoplasms/immunology , Hematologic Neoplasms/pathology , Hematologic Neoplasms/therapy , Humans , Lymphocyte Count , Male , Middle Aged , Natural Killer T-Cells/pathology , Recurrence , Risk Factors , Survival Analysis , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Time Factors , Tissue Donors , Transplantation, Homologous
14.
J Immunol ; 188(2): 624-31, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22156591

ABSTRACT

Invariant NKT (iNKT) cells constitute a versatile T cell subset with important regulatory functions, which are thought to result essentially from their capacity to promptly produce cytokines that influence the Th1/Th2 balance. In this study, we report that these cells can also express Foxp3, an important transcriptional regulator associated with suppressive activity, once they have been exposed to TGF-ß. Foxp3 was expressed by iNKT cells from both peripheral and cord blood. CD4(+) iNKT cells acquired Foxp3 expression preferentially, although a lower proportion of their CD4(-) counterpart also became positive. All Foxp3(+) iNKT cells displayed CD25 but not necessarily CTLA4 or GITR, regardless of the upregulation of these markers in the presence of TGF-ß. Exposure to TGF-ß decreased IL-4 and IFN-γ production while increasing IL-10, independently from Foxp3 expression. IL-17 was not detected. TGF-ß induced high levels of Foxp3, but no suppressor activity, which emerged only in the presence of rapamycin. Peripheral and cord blood Foxp3(+) iNKT cells suppressed the proliferation of conventional autologous and heterologous CD4(+) T cells equally, in a cell contact-dependent and Ag-independent manner. Our findings demonstrate that human iNKT cells become suppressive in the presence of TGF-ß plus rapamycin, thus adding a new facet to their complex functional properties.


Subject(s)
Cell Differentiation/immunology , Forkhead Transcription Factors/biosynthesis , Immunosuppressive Agents/pharmacology , Natural Killer T-Cells/cytology , Natural Killer T-Cells/immunology , Sirolimus/pharmacology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/physiology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Fetal Blood/cytology , Fetal Blood/drug effects , Fetal Blood/immunology , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Natural Killer T-Cells/drug effects , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects
15.
Mycopathologia ; 172(5): 357-63, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21805204

ABSTRACT

Invariant natural killer T (iNKT) cells are capable of recognizing lipid antigens and secreting Th1/Th2 cytokines. Deficiency in iNKT cell number or function has been partially implicated in susceptibility to some infectious diseases, such as tuberculosis. We evaluated iNKT cells in paracoccidioidomycosis, another chronic granulomatous disease endemic in Latin America. iNKT cells were detected using PBS57-loaded tetramer staining and flow cytometry. Circulating iNKT cell numbers were similar among healthy individuals who had previously been cured of paracoccidioidomycosis (susceptible individuals, n = 7) and healthy Paracoccidioides brasiliensis-infected (n = 5) and non-infected individuals (n = 5). iNKT from all three groups expanded similarly upon α-GalCer and a synthetic analog (OCH) stimulation. IFN-γ was the dominant cytokine produced both by ex vivo and by expanded iNKT cells, followed by IL-4 and IL-10, in the three groups. No deficit in the monocyte expression of CD1d was detected. In conclusion, individuals who had developed paracoccidioidomycosis in the past have no impairment in iNKT number, expansion capacity, and cytokine secretion.


Subject(s)
Natural Killer T-Cells/immunology , Paracoccidioides/immunology , Paracoccidioidomycosis/immunology , Adult , Antigens, CD1d/biosynthesis , Female , Humans , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Interleukin-4/biosynthesis , Male , Middle Aged , Monocytes/metabolism , Natural Killer T-Cells/metabolism
16.
J Immunol ; 186(10): 5758-65, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21478400

ABSTRACT

CD1d-reactive invariant NKT (iNKT) cells have been implicated in a number of experimental models of human pathologies. Given the scope of their immunoregulatory activities mediated through distinct cytokine patterns, it has been proposed that this functional diversity originates from distinct iNKT subpopulations. In this study, we report that human CD161(+) iNKT cells are intrinsically endowed with the capacity to generate IL-17, but require TGF-ß, IL-1ß, and IL-23 to carry out this potential. IL-17-producing iNKT cells are already present in cord blood but, in contrast to peripheral blood iNKT cells, they cannot generate IFN-γ. These IL-17 producers respond to aryl hydrocarbon receptor stimulation and express IL-23 receptor and retinoic acid-related orphan receptor C, similar to conventional T helper 17 cells, from which they differ by their restricted ability to coproduce IL-22. In conclusion, IL-17 production by human iNKT cells depends on two critical parameters, namely an intrinsic program and a proinflammatory environment.


Subject(s)
Inflammation/immunology , Interleukin-17/biosynthesis , Natural Killer T-Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Interferon-gamma , Interleukin-1beta/biosynthesis , Interleukin-23/biosynthesis , Interleukins/biosynthesis , NK Cell Lectin-Like Receptor Subfamily B/immunology , Natural Killer T-Cells/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Polymerase Chain Reaction , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Interleukin/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Transforming Growth Factor beta/biosynthesis , Interleukin-22
SELECTION OF CITATIONS
SEARCH DETAIL
...