Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 182
Filter
1.
J Infect Dis ; 227(4): 577-582, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36520641

ABSTRACT

The mechanisms involved in HIV-associated natural killer (NK) cell impairment are still incompletely understood. We observed HIV infection to be associated with increased plasma levels of IFABP, a marker for gut epithelial barrier dysfunction, and LBP, a marker for microbial translocation. Both IFABP and LBP plasma concentrations were inversely correlated with NK cell interferon-γ production, suggesting microbial translocation to modulate NK cell functions. Accordingly, we found lipopolysaccharide to have an indirect inhibitory effect on NK cells via triggering monocytes' transforming growth factor-ß production. Taken together, our data suggest increased microbial translocation to be involved in HIV-associated NK cell dysfunction.


Subject(s)
HIV Infections , Monocytes , Humans , Cytokines , HIV Infections/metabolism , HIV Infections/microbiology , Killer Cells, Natural/metabolism , Killer Cells, Natural/microbiology , Killer Cells, Natural/pathology , CD56 Antigen , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology
2.
Front Immunol ; 12: 767359, 2021.
Article in English | MEDLINE | ID: mdl-34966388

ABSTRACT

Melioidosis is a potentially fatal bacterial disease caused by Burkholderia pseudomallei and is estimated to cause 89,000 deaths per year in endemic areas of Southeast Asia and Northern Australia. People with diabetes mellitus are most at risk of melioidosis, with a 12-fold increased susceptibility for severe disease. Interferon gamma (IFN-γ) responses from CD4 and CD8 T cells, but also from natural killer (NK) and natural killer T (NKT) cells, are necessary to eliminate the pathogen. We previously reported that immunization with B. pseudomallei OmpW (BpOmpW antigen) protected mice from lethal B. pseudomallei challenge for up to 81 days. Elucidating the immune correlates of protection of the protective BpOmpW vaccine is an essential step prior to clinical trials. Thus, we immunized either non-insulin-resistant C57BL/6J mice or an insulin-resistant C57BL/6J mouse model of type 2 diabetes (T2D) with a single dose of BpOmpW. BpOmpW induced strong antibody responses, stimulated effector CD4+ and CD8+ T cells and CD4+ CD25+ Foxp3+ regulatory T cells, and produced higher IFN-γ responses in CD4+, CD8+, NK, and NKT cells in non-insulin-resistant mice. The T-cell responses of insulin-resistant mice to BpOmpW were comparable to those of non-insulin-resistant mice. In addition, as a precursor to its evaluation in human studies, humanized HLA-DR and HLA-DQ (human leukocyte antigen DR and DQ isotypes, respectively) transgenic mice elicited IFN-γ recall responses in an enzyme-linked immune absorbent spot (ELISpot)-based study. Moreover, human donor peripheral blood mononuclear cells (PBMCs) exposed to BpOmpW for 7 days showed T-cell proliferation. Finally, plasma from melioidosis survivors with diabetes recognized our BpOmpW vaccine antigen. Overall, the range of approaches used strongly indicated that BpOmpW elicits the necessary immune responses to combat melioidosis and bring this vaccine closer to clinical trials.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Burkholderia pseudomallei/immunology , Melioidosis/immunology , T-Lymphocytes/immunology , Animals , Bacterial Vaccines/administration & dosage , Burkholderia pseudomallei/metabolism , Burkholderia pseudomallei/physiology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/microbiology , Cells, Cultured , Diabetes Mellitus, Type 2/immunology , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/microbiology , Male , Melioidosis/microbiology , Melioidosis/prevention & control , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes/metabolism , T-Lymphocytes/microbiology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/microbiology
3.
PLoS Pathog ; 17(1): e1008299, 2021 01.
Article in English | MEDLINE | ID: mdl-33465134

ABSTRACT

Host resistance against intracellular pathogens requires a rapid IFN-γ mediated immune response. We reveal that T-bet-dependent production of IFN-γ is essential for the maintenance of inflammatory DCs at the site of infection with a common protozoan parasite, Toxoplasma gondii. A detailed analysis of the cellular sources for T-bet-dependent IFN-γ identified that ILC1s and to a lesser degree NK, but not TH1 cells, were involved in the regulation of inflammatory DCs via IFN-γ. Mechanistically, we established that T-bet dependent innate IFN-γ is critical for the induction of IRF8, an essential transcription factor for cDC1s. Failure to upregulate IRF8 in DCs resulted in acute susceptibility to T. gondii infection. Our data identifies that T-bet dependent production of IFN-γ by ILC1 and NK cells is indispensable for host resistance against intracellular infection via maintaining IRF8+ inflammatory DCs at the site of infection.


Subject(s)
Dendritic Cells/immunology , Immunity, Innate/immunology , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Lymphocytes/immunology , T-Box Domain Proteins/metabolism , Toxoplasma/immunology , Toxoplasmosis/immunology , Animals , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Female , Interferon Regulatory Factors/physiology , Killer Cells, Natural/metabolism , Killer Cells, Natural/microbiology , Lymphocytes/metabolism , Lymphocytes/microbiology , Male , Mice, Inbred C57BL , Mice, Knockout , T-Box Domain Proteins/genetics , Toxoplasma/metabolism , Toxoplasmosis/metabolism , Toxoplasmosis/microbiology
4.
Front Cell Infect Microbiol ; 11: 799276, 2021.
Article in English | MEDLINE | ID: mdl-35071048

ABSTRACT

Aim: Smoker COPD patients with chest radiological signs of prior tuberculosis (TB) showed more severe lung damage, but the mechanisms remain unclear. Emerging evidence has implicated NK cells in the pathogenesis of both COPD and TB. The purpose of this study was to delineate the profile and cytokine production of NK-cell subpopulations and their immunometabolic changes after exposure to both cigarette smoke (CS) and Mycobacterium tuberculosis(MTB). Methods: We profiled NK-cell subpopulations in terms of percentage and cytokine production by flow cytometry in smoker patients with pulmonary TB (PTB). In an in vitro coexposure model, we investigated proinflammatory cytokine production, glycolytic influx, and oxidative phosphorylation of NK cells under CS extract (CSE) and PPD costimulation. Results: Peripheral blood NK cells in smoker patients with active PTB (CS+PTB group) showed altered proportion of subpopulations and excessive proinflammatory cytokine expressions. In vitro, CSE- and PPD-coexposed NK-92 cells displayed enhanced proinflammatory cytokine production, concurrent with decreased glycolytic influx and oxidative phosphorylation. Conclusion: Smoker patients with active PTB showed enhanced proinflammatory cytokine expression within altered NK cell subpopulations. CSE and PPD coexposure induced heightened cytokine production concurrent with impaired cell metabolism in NK cells. These novel data suggest a potential role of NK cells in the pathogenesis of lung injury in subjects with coexposure to CS and TB.


Subject(s)
Cytokines/immunology , Killer Cells, Natural/immunology , Mycobacterium tuberculosis , Smoke/adverse effects , Cells, Cultured , Humans , Killer Cells, Natural/microbiology , Lung , Pulmonary Disease, Chronic Obstructive
5.
Infect Immun ; 88(11)2020 10 19.
Article in English | MEDLINE | ID: mdl-32817330

ABSTRACT

Natural killer (NK) cells are critically involved in the early immune response against various intracellular pathogens, including Coxiella burnetii and Chlamydia psittaciChlamydia-infected NK cells functionally mature, induce cellular immunity, and protect themselves by killing the bacteria in secreted granules. Here, we report that infected NK cells do not allow intracellular multiday growth of Coxiella, as is usually observed in other host cell types. C. burnetii-infected NK cells display maturation and gamma interferon (IFN-γ) secretion, as well as the release of Coxiella-containing lytic granules. Thus, NK cells possess a potent program to restrain and expel different types of invading bacteria via degranulation. Strikingly, though, in contrast to Chlamydia, expulsed Coxiella organisms largely retain their infectivity and, hence, escape the cell-autonomous self-defense mechanism in NK cells.


Subject(s)
Cell Degranulation/immunology , Immunity, Cellular/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/microbiology , Q Fever/immunology , Animals , Coxiella burnetii , Mice
6.
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
7.
Cell Rep ; 29(12): 3933-3945.e3, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31851924

ABSTRACT

Natural killer (NK) cells are unique players in innate immunity and, as such, an attractive target for immunotherapy. NK cells display immune memory properties in certain models, but the long-term status of NK cells following systemic inflammation is unknown. Here we show that following LPS-induced endotoxemia in mice, NK cells acquire cell-intrinsic memory-like properties, showing increased production of IFNγ upon specific secondary stimulation. The NK cell memory response is detectable for at least 9 weeks and contributes to protection from E. coli infection upon adoptive transfer. Importantly, we reveal a mechanism essential for NK cell memory, whereby an H3K4me1-marked latent enhancer is uncovered at the ifng locus. Chemical inhibition of histone methyltransferase activity erases the enhancer and abolishes NK cell memory. Thus, NK cell memory develops after endotoxemia in a histone methylation-dependent manner, ensuring a heightened response to secondary stimulation.


Subject(s)
Endotoxemia/immunology , Escherichia coli Infections/immunology , Histones/metabolism , Immunity, Innate/immunology , Immunologic Memory/immunology , Inflammation/immunology , Killer Cells, Natural/immunology , Animals , Endotoxemia/metabolism , Endotoxemia/microbiology , Endotoxemia/pathology , Enhancer Elements, Genetic , Escherichia coli/immunology , Escherichia coli Infections/microbiology , Histones/genetics , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Killer Cells, Natural/microbiology , Killer Cells, Natural/pathology , Male , Mice
8.
Sci Rep ; 9(1): 4799, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30886314

ABSTRACT

Natural killer (NK) cells are innate immune cells critically involved in the early immune response against various pathogens including chlamydia. Here, we demonstrate that chlamydia-infected NK cells prevent the intracellular establishment and growth of the bacteria. Upon infection, they display functional maturation characterized by enhanced IFN-γ secretion, CD146 induction, PKCϴ activation, and granule secretion. Eventually, chlamydia are released in a non-infectious, highly immunogenic form driving a potent Th1 immune response. Further, anti-chlamydial antibodies generated during immunization neutralize the infection of epithelial cells. The release of chlamydia from NK cells requires PKCϴ function and active degranulation, while granule-associated granzyme B drives the loss of chlamydial infectivity. Cellular infection and bacterial release can be undergone repeatedly and do not affect NK cell function. Strikingly, NK cells passing through such an infection cycle significantly improve their cytotoxicity. Thus, NK cells not only protect themselves against productive chlamydial infections but also actively trigger potent anti-bacterial responses.


Subject(s)
Chlamydophila psittaci/immunology , Immunity, Cellular , Killer Cells, Natural/immunology , Psittacosis/immunology , Th1 Cells/immunology , Animals , CD146 Antigen/metabolism , Cell Communication/immunology , Cells, Cultured , Disease Models, Animal , Female , Humans , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Killer Cells, Natural/microbiology , Mice , Primary Cell Culture , Protein Kinase C-theta/metabolism , Psittacosis/blood , Psittacosis/microbiology , Spleen/cytology
9.
Mycoses ; 62(7): 588-596, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30908750

ABSTRACT

The prevalence of atopic dermatitis (AD) has been increasing. Whereas AD symptoms are obvious and easy to recognise, the etiopathogenesis remains not fully elucidated. Recently, the role of microorganisms and their impact on the immunology of AD have been discussed. In this review, we summarise a possible role of Malassezia in the development and persistence of eczema in patients with atopic eczema/dermatitis syndrome. A high proportion of AD patients present with a positive reaction to Malassezia allergens. Several possible pathogenic mechanisms enable Malassezia to trigger the development of AD. Malassezia spp. may release more allergens in a less acidic (pH <6), typical for AD, environment. The similarity between fungal thioredoxin and human proteins causes T-cell cross-reactivity. TLR-mediated mechanisms are involved in host response against Malassezia spp. An interaction between Malassezia spp. and keratinocytes alters the profile of cytokine release, and what is more, yeast cells can survive when absorbed by keratinocytes. Dendritic cells of AD patients induced by Malassezia are less susceptible to lysis mediated by NK cells which exerts a pro-inflammatory effect. Despite the evidence that Malassezia spp. contribute to the development of AD, the pathogenetic mechanisms and relationship between Malassezia and immune defense remain partly unexplained and require further research.


Subject(s)
Dermatitis, Atopic/pathology , Dermatitis, Atopic/physiopathology , Dermatomycoses/complications , Dermatomycoses/pathology , Host-Pathogen Interactions , Malassezia/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/microbiology , Dermatitis, Atopic/epidemiology , Dermatomycoses/epidemiology , Humans , Keratinocytes/immunology , Keratinocytes/microbiology , Killer Cells, Natural/immunology , Killer Cells, Natural/microbiology , Prevalence
10.
Cell Death Differ ; 26(4): 703-714, 2019 03.
Article in English | MEDLINE | ID: mdl-30737478

ABSTRACT

Autophagy is an evolutionally conserved, highly regulated catabolic process that combines cellular functions required for the regulation of metabolic balance under conditions of stress with those needed for the degradation of damaged cell organelles via the lysosomal machinery. The importance of autophagy for cell homeostasis and survival has long been appreciated. Recent data suggest that autophagy is also involved in non-metabolic functions that impact the immune system. Here, we reflect in two review articles the recent literature pointing to an important role for autophagy in innate immune cells. In this article, we focus on neutrophils, eosinophils, mast cells, and natural killer cells. We mainly discuss the influence of autophagy on functional cellular responses and its importance for overall host defense. In the companion review, we present the role of autophagy in the functions performed by monocytes/macrophages and dendritic cells.


Subject(s)
Autophagy/immunology , Eosinophils/immunology , Immunity, Innate , Killer Cells, Natural/immunology , Mast Cells/immunology , Neutrophils/metabolism , Animals , Autophagy/genetics , Autophagy/physiology , Cell Death/genetics , Cell Death/immunology , Eosinophils/metabolism , Extracellular Traps/immunology , Extracellular Traps/metabolism , Extracellular Traps/microbiology , Humans , Immunity, Innate/drug effects , Immunity, Innate/genetics , Killer Cells, Natural/metabolism , Killer Cells, Natural/microbiology , Killer Cells, Natural/virology , Mast Cells/metabolism , Mast Cells/microbiology , Neutrophils/immunology , Neutrophils/microbiology
11.
Inflamm Bowel Dis ; 25(3): 510-523, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30462201

ABSTRACT

BACKGROUND: The tumor necrosis factor alpha (TNFα)-homologous cytokine TL1A is emerging as a major player in intestinal inflammation. From in vitro experiments on human lymphocytes, TNF-like molecule 1A (TL1A) is known to activate a highly inflammatory lymphoid response in synergy with interleukin (IL)-12 and IL-18. Carriers of specific genetic polymorphisms associated with IL-12, IL-18, or TL1A signaling have increased Crohn's disease risk, and all 3 cytokines are upregulated during active disease. The study aim was to investigate whether the type 1-polarizing cytokines IL-12 and IL-18 could directly initiate intestinal pathology in mice and how TL1A would influence the resulting inflammatory response. METHODS: Conventional barrier-bred and germ-free mice were randomly allocated to different groups and injected twice with different combinations of IL-12, IL-18, and TL1A, and killed 3 days after the first injection. All treatment groups were co-housed and fed a piroxicam-supplemented chow diet. RESULTS: Intestinal pathology was evident in IL-12- and IL-18-treated mice and highly exacerbated by TL1A in both the colon and ileum. The cytokine-induced intestinal inflammation was characterized by epithelial damage, increased colonic levels of TNFα, IL-1ß, IFN-γ, and IL-6, and various chemokines along with gut microbiota alterations exhibiting high abundance of Enterobacteriaceae. Furthermore, the inflamed ileum and colon exhibited a TL1A-specific increased infiltration of intraepithelial natural killer cells co-expressing NKG2D and IL-18Ra and a higher frequency of unconventional T cells in the colonic epithelium. Upon cytokine injection, germ-free mice exhibited similar intraepithelial lymphoid infiltration and increased colonic levels of IFNγ and TNFα. CONCLUSIONS: This study demonstrates that TL1A aggravates IL-12- and IL-18-induced intestinal inflammation in the presence and absence of microbiota.


Subject(s)
Epithelial Cells/immunology , Gastrointestinal Tract/immunology , Inflammation/etiology , Interleukin-12 Subunit p35/administration & dosage , Interleukin-18/administration & dosage , Killer Cells, Natural/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/physiology , Animals , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Female , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/microbiology , Inflammation/metabolism , Inflammation/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Killer Cells, Natural/drug effects , Killer Cells, Natural/microbiology , Mice , Mice, Inbred BALB C , Mice, Knockout
12.
FASEB J ; 33(2): 2719-2731, 2019 02.
Article in English | MEDLINE | ID: mdl-30303739

ABSTRACT

Remodeling of the gut microbiota is implicated in various metabolic and inflammatory diseases of the gastrointestinal tract. We hypothesized that the gut microbiota affects the DNA methylation profile of intestinal epithelial cells (IECs) which could, in turn, alter intestinal function. In this study, we used mass spectrometry and methylated DNA capture to respectively investigate global and genome-wide DNA methylation of intestinal epithelial cells from germ-free (GF) and conventionally raised mice. In colonic IECs from GF mice, DNA was markedly hypermethylated. This was associated with a dramatic loss of ten-eleven-translocation activity, a lower DNA methyltransferase activity and lower circulating levels of the 1-carbon metabolite, folate. At the gene level, we found an enrichment for differentially methylated regions proximal to genes regulating the cytotoxicity of NK cells (false-discovery rate < 8.9E-6), notably genes regulating the cross-talk between NK cells and target cells, such as members of the NK group 2 member D ligand superfamily Raet. This distinct epigenetic signature was associated with a marked decrease in Raet1 expression and a loss of CD56+/CD45+ cells in the intestine of GF mice. Thus, our results indicate that altered activity of methylation-modifying enzymes in GF mice influences the IEC epigenome and modulates the crosstalk between IECs and NK cells. Epigenetic reprogramming of IECs may modulate intestinal function in diseases associated with altered gut microbiota.-Poupeau, A., Garde, C., Sulek, K., Citirikkaya, K., Treebak, J. T., Arumugam, M., Simar, D., Olofsson, L. E., Bäckhed, F., Barrès, R. Genes controlling the activation of natural killer lymphocytes are epigenetically remodeled in intestinal cells from germ-free mice.


Subject(s)
Biomarkers/analysis , Epigenesis, Genetic , Epithelial Cells/immunology , Gastrointestinal Microbiome , Gene Expression Regulation , Germ-Free Life , Killer Cells, Natural/immunology , Animals , DNA Methylation , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Female , Intestines/cytology , Intestines/microbiology , Intestines/physiology , Killer Cells, Natural/metabolism , Killer Cells, Natural/microbiology , Male , Mice
13.
Nat Commun ; 9(1): 4117, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30297690

ABSTRACT

Infection of specific pathogen-free mice with lymphocytic choriomeningitis virus (LCMV) is a widely used model to study antiviral T-cell immunity. Infections in the real world, however, are often accompanied by coinfections with unrelated pathogens. Here we show that in mice, systemic coinfection with E. coli suppresses the LCMV-specific cytotoxic T-lymphocyte (CTL) response and virus elimination in a NK cell- and TLR2/4-dependent manner. Soluble TLR4 ligand LPS also induces NK cell-mediated negative CTL regulation during LCMV infection. NK cells in LPS-treated mice suppress clonal expansion of LCMV-specific CTLs by a NKG2D- or NCR1-independent but perforin-dependent mechanism. These results suggest a TLR4-mediated immunoregulatory role of NK cells during viral-bacterial coinfections.


Subject(s)
Arenaviridae Infections/immunology , CD8-Positive T-Lymphocytes/immunology , Coinfection/immunology , Escherichia coli Infections/immunology , Killer Cells, Natural/immunology , Lipopolysaccharides/immunology , Animals , Arenaviridae Infections/virology , CD8-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/virology , Coinfection/microbiology , Coinfection/virology , Escherichia coli/immunology , Escherichia coli/physiology , Escherichia coli Infections/microbiology , Host-Pathogen Interactions/immunology , Killer Cells, Natural/microbiology , Killer Cells, Natural/virology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/physiology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Perforin/immunology , Perforin/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/microbiology , T-Lymphocytes, Cytotoxic/virology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism
14.
Proc Natl Acad Sci U S A ; 115(45): 11585-11590, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30348790

ABSTRACT

The cytokine IFN-γ is a critical regulator of immune system development and function. Almost all leukocytes express the receptor for IFN-γ, yet each cell type elicits a different response to this cytokine. Cell type-specific effects of IFN-γ make it difficult to predict the outcomes of the systemic IFN-γ blockade and limit its clinical application, despite many years of research. To better understand the cell-cell interactions and cofactors that specify IFN-γ functions, we focused on the function of IFN-γ on CD8 T cell differentiation. We demonstrated that during bacterial infection, IFN-γ is a dominant paracrine trigger that skews CD8 T cell differentiation toward memory. This skewing is preferentially driven by contact-dependent T cell-T cell (T-T) interactions and the localized IFN-γ secretion among activated CD8 T cells in a unique splenic microenvironment, and is less sensitive to concurrent IFN-γ production by other immune cell populations such as natural killer (NK) cells. Modulation of CD8 T cell differentiation by IFN-γ relies on a nonconventional IFN-γ outcome that occurs specifically within 24 hours following infection. This is driven by IFN-γ costimulation by integrins at T-T synapses, and leads to synergistic phosphorylation of the proximal STAT1 molecule and accelerated IL-2 receptor down-regulation. This study provides evidence of the importance of context-dependent cytokine signaling and gives another example of how cell clusters and the microenvironment drive unique biology.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Cell Differentiation/drug effects , Integrins/immunology , Interferon-gamma/pharmacology , Paracrine Communication/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/microbiology , Cell Differentiation/immunology , Cellular Microenvironment , Immunologic Memory , Immunological Synapses , Interferon-gamma/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/microbiology , Listeria monocytogenes/growth & development , Listeria monocytogenes/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Primary Cell Culture , Signal Transduction , Spleen/cytology , Spleen/immunology , Tetradecanoylphorbol Acetate/pharmacology
15.
Tuberculosis (Edinb) ; 108: 114-117, 2018 01.
Article in English | MEDLINE | ID: mdl-29523310

ABSTRACT

Regulatory B cells (Bregs) have been shown to be present during several disease states. The phenotype of the cells is not completely defined and the function of these cells differ between disease. The presence of FASL expressing (killer) B cells during latent and successfully treated TB disease have been shown but whether these cells are similar to regulatory B cells remain unclear. We assessed the receptor expression of FASL/IL5 (killer B cells), CD24/CD38 (regulatory B cells) on whole peripheral blood of participants with untreated active TB and healthy controls. We then isolated B cells from a second cohort of M.tb exposed (Quantiferon (QFN) positive) and unexposed (Quantiferon negative) HIV negative participants, and evaluated the frequency of killer B cells induced following stimulation with BCG and/or CD40 and IL5. Our data reveal no difference in the expression on CD24 and CD38 between participants with active TB and the controls. There was also no difference in the frequency of regulatory B cells measured in the peripheral blood mononuclear cells (PBMC) fraction between latent TB and uninfected controls. We did however notice that regulatory B cells (CD24hiCD38hi) population express the FASL receptor. The expression of killer B cell phenotype (CD178+IL5RA+) was significantly higher in controls compared to those with active TB disease (1,06% vs 0,455%). Furthermore, we found that BCG restimulation significantly induced the FASL/IL5RA B cells but this was only evident in the QFN positive group. Our data suggest that both regulatory and killer B cells are present during latent and active TB disease but that the frequency of these populations are increased during latent disease. We also show that the FASL+IL5RA+ B killer B cells are induced in latent TB infection following BCG restimulation but whether these cells are indicative of protection remains unclear.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Fas Ligand Protein/immunology , Killer Cells, Natural/immunology , Latent Tuberculosis/immunology , Lymphocyte Activation , Mycobacterium bovis/immunology , Mycobacterium tuberculosis/immunology , ADP-ribosyl Cyclase 1/blood , ADP-ribosyl Cyclase 1/immunology , B-Lymphocytes, Regulatory/metabolism , B-Lymphocytes, Regulatory/microbiology , CD24 Antigen/blood , CD24 Antigen/immunology , Case-Control Studies , Cell Proliferation , Fas Ligand Protein/blood , Host-Pathogen Interactions , Humans , Interleukin-5 Receptor alpha Subunit/blood , Interleukin-5 Receptor alpha Subunit/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/microbiology , Latent Tuberculosis/blood , Latent Tuberculosis/microbiology , Lymphocyte Count , Membrane Glycoproteins/blood , Membrane Glycoproteins/immunology , Phenotype
16.
BMC Immunol ; 19(1): 8, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29433450

ABSTRACT

BACKGROUND: Besides their prominent role in the elimination of infected or malignantly transformed cells, natural killer (NK) cells serve as modulators of adaptive immune responses. Enhancing bidirectional crosstalk between NK cells and dendritic cells (DC) is considered a promising tool to potentiate cancer vaccines. We investigated to what extent direct sensing of viral and bacterial motifs by NK cells contributes to the response of inflammatory DC against the same pathogenic stimulus. RESULTS: We demonstrated that sensing of bacterial and viral PAMPs by NK cells contributes to DC cytokine production via NK cell-derived soluble factors. This enhancement of DC cytokine production was dependent on the pattern recognition receptor (PRR) agonist but also on the cytokine environment in which NK cells recognized the pathogen, indicating the importance of accessory cell activation for this mechanism. We showed in blocking experiments that NK cell-mediated amplification of DC cytokine secretion is dependent on NK cell-derived IFN-γ irrespective of the PRR that is sensed by the NK cell. CONCLUSIONS: These findings illustrate the importance of bidirectional interaction between different PRR-expressing immune cells, which can have implications on the selection of adjuvants for vaccination strategies.


Subject(s)
Cytokines/immunology , Dendritic Cells/immunology , Inflammation Mediators/immunology , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Monocytes/immunology , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/metabolism , Humans , Inflammation Mediators/metabolism , Interferon-gamma/metabolism , Killer Cells, Natural/microbiology , Killer Cells, Natural/virology , Lymphocyte Activation/immunology , Monocytes/metabolism , Pathogen-Associated Molecular Pattern Molecules/immunology , Pathogen-Associated Molecular Pattern Molecules/metabolism , Receptors, Pattern Recognition/immunology , Receptors, Pattern Recognition/metabolism
17.
J Leukoc Biol ; 103(3): 591-599, 2018 03.
Article in English | MEDLINE | ID: mdl-27106671

ABSTRACT

The pivotal role of NK cells in viral infection is extensively studied, whereas the role of NK cells in bacterial infection has been poorly investigated. Here, we have examined how Listeria monocytogenes (LM) affects expression of ligands for NK cell receptors and subsequent NK cell responses, depending on the type of cell infected. LM infected rat cell lines derived from different tissues were coincubated with splenic NK cells, and NK cell proliferation and IFN-γ production were measured. In addition, expression of ligands for the NK cell receptors Ly49 and NK cell receptor protein 1 (NKR-P1), MHC class I and C-type lectin-related molecules, respectively, was assessed. Infected pleural R2 cells, but not epithelium-derived colon carcinoma cell line CC531 cells, induced proliferation of NK cells. Reporter cells expressing the inhibitory NKR-P1G receptor or the activating NKR-P1F receptor were less stimulated under incubation with infected CC531 cells versus uninfected CC531 controls, suggesting that the ligand(s) in question were down-regulated by infection. Conversely, LM infection of R2 cells did not affect reporter cell stimulation compared with uninfected R2 controls. We characterized a rat monocyte cell line, termed RmW cells. In contrast to LM infected R2 cells that up-regulate MHC class I molecules, RmW cells displayed unchanged MHC class I expression following infection. In line with MHC class I expression, more NK cells produced a higher amount of IFN-γ against infected R2 cells compared with RmW cells. Together, L. monocytogenes infection may variously regulate cellular ligands for NK cells, depending on the cell type infected, affecting the outcome of NK cell responses.


Subject(s)
Colonic Neoplasms/metabolism , Killer Cells, Natural/metabolism , Listeria monocytogenes/immunology , Listeriosis/immunology , Macrophages/metabolism , Receptors, Natural Killer Cell/metabolism , Animals , Cells, Cultured , Colonic Neoplasms/immunology , Colonic Neoplasms/microbiology , Colonic Neoplasms/pathology , Killer Cells, Natural/immunology , Killer Cells, Natural/microbiology , Killer Cells, Natural/pathology , Ligands , Listeria monocytogenes/pathogenicity , Listeriosis/metabolism , Listeriosis/microbiology , Listeriosis/pathology , Lymphocyte Activation , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Male , Mice , Rats
18.
J Immunol ; 200(1): 177-185, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29150567

ABSTRACT

Although prophylactic vaccines provide protective humoral immunity against infectious agents, vaccines that elicit potent CD8 T cell responses are valuable tools to shape and drive cellular immunity against cancer and intracellular infection. In particular, IFN-γ-polarized cytotoxic CD8 T cell immunity is considered optimal for protective immunity against intracellular Ags. Suppressor of cytokine signaling (SOCS)1 is a cross-functional negative regulator of TLR and cytokine receptor signaling via degradation of the receptor-signaling complex. We hypothesized that loss of SOCS1 in dendritic cells (DCs) would improve T cell responses by accentuating IFN-γ-directed immune responses. We tested this hypothesis using a recombinant Listeria monocytogenes vaccine platform that targets CD11c+ DCs in mice in which SOCS1 is selectively deleted in all CD11c+ cells. Unexpectedly, in mice lacking SOCS1 expression in CD11c+ cells, we observed a decrease in CD8+ T cell response to the L. monocytogenes vaccine. NK cell responses were also decreased in mice lacking SOCS1 expression in CD11c+ cells but did not explain the defect in CD8+ T cell immunity. We found that DCs lacking SOCS1 expression were functional in driving Ag-specific CD8+ T cell expansion in vitro but that this process was defective following infection in vivo. Instead, monocyte-derived innate TNF-α and inducible NO synthase-producing DCs dominated the antibacterial response. Thus, loss of SOCS1 in CD11c+ cells skewed the balance of immune response to infection by increasing innate responses while decreasing Ag-specific adaptive responses to infectious Ags.


Subject(s)
Bacterial Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Killer Cells, Natural/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Suppressor of Cytokine Signaling 1 Protein/metabolism , Adaptive Immunity , Animals , CD11c Antigen/metabolism , CD8-Positive T-Lymphocytes/microbiology , Cells, Cultured , Cytotoxicity, Immunologic , Humans , Immunity, Innate , Interferon-gamma/metabolism , Killer Cells, Natural/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Suppressor of Cytokine Signaling 1 Protein/genetics
19.
J Mol Biol ; 430(2): 193-206, 2018 01 19.
Article in English | MEDLINE | ID: mdl-28454742

ABSTRACT

Inflammasome signaling impinges on the activation of inflammatory caspases (i.e., caspase-1 and caspase-4/5/11) and endows host cells with a sentinel system to sense microbial intrusion and thereby initiate appropriate immune responses. Lately, it has become evident that mammalian inflammasome-dependent responses to infection are not confined solely to cells of hematopoietic origin. Epithelial cells that line the body's mucosal surfaces use inflammasome signaling to sense and counteract pathogenic microorganisms that compromise barrier integrity. Many of the molecular mechanisms of epithelial inflammasome signaling remain unexplored. However, it now seems clear that epithelial inflammasome activation has a profound impact both on the infected cell itself and on its ability to communicate with other cell types of the mucosa. Here, we summarize current knowledge regarding the output of epithelial inflammasome activation during bacterial infection. Well-established downstream effects include epithelial cell death, release of soluble mediators, and subsequent recruitment of effector cell types, including NK cells, mast cells, and neutrophils, to sites of mucosal infection. We discuss the implications of recent findings for antibacterial defense in the mucosa and sketch out areas for future exploration.


Subject(s)
Bacteria/immunology , Bacterial Infections/immunology , Epithelium/immunology , Inflammasomes/immunology , Inflammation/immunology , Animals , Bacterial Infections/microbiology , Bacterial Infections/pathology , Caspases/immunology , Cell Death , Epithelium/microbiology , Epithelium/pathology , Humans , Inflammation/microbiology , Inflammation/pathology , Interleukin-18/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/microbiology , Killer Cells, Natural/pathology , Mast Cells/immunology , Mast Cells/microbiology , Mast Cells/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Neutrophils/immunology , Neutrophils/microbiology , Neutrophils/pathology
20.
Immunol Res ; 65(6): 1139-1149, 2017 12.
Article in English | MEDLINE | ID: mdl-29052125

ABSTRACT

Cell-mediated immune responses characterized by the secretion of IFNγ and IL-17 play an important role in the immune response to Bordetella pertussis (B. pertussis). We investigated innate sources of IFNγ and IL-17 upon stimulation of spleen cells from BALB/c (B/c) and C57BL/6 (B6) mice with heat-killed B. pertussis (hkBp). Spleen cells from B/c mice secreted less IFNγ and more IL-17 than those from B6 mice. Innate IFNγ was produced predominantly by NK cells in B/c mice and by CD8 T cells and NK cells in B6 mice. Innate IL-17 was produced primarily by γδT cells in both mouse strains. The secretion of IFNγ was abrogated by anti-IL-12, and the production of IL-17 was abolished by anti-IL-1ß- and anti-IL23-neutralizing antibodies. B/c dendritic cells (DCs) stimulated with hkBp secreted significantly more IL-1ß and less IL-12 than B6 DCs. Differences in JNK phosphorylation in DCs suggest that this pathway plays a role in the differences between B/c and B6 strains. Mixed cultures of DCs and γδT cells from B/c and B6 showed that cytokines from DCs as well as γδT cell-intrinsic factors contributed to the robust innate IL-17 response in B/c strain. Stimulation of γδT cells with IL-1ß and IL-23 was sufficient for IL-17 secretion whereas IL-12 inhibited the secretion of IL-17. A larger fraction of γδT cells were γδT-17 cells in B/c mice than B6 mice. Our data indicate important roles for genetically determined factors in the innate IFNγ and IL-17 responses to B. pertussis.


Subject(s)
Bordetella pertussis/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Interleukin-17/metabolism , Killer Cells, Natural/immunology , Whooping Cough/immunology , Animals , CD8-Positive T-Lymphocytes/microbiology , Cells, Cultured , Coculture Techniques , Dendritic Cells/microbiology , Genetic Background , Humans , Immunity, Cellular , Immunity, Innate , Interferon-gamma/metabolism , Killer Cells, Natural/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, gamma-delta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...