Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Trends Immunol ; 43(12): 990-1005, 2022 12.
Article in English | MEDLINE | ID: mdl-36347788

ABSTRACT

Metabolites are emerging as essential factors for the immune system that are involved in both metabolic circuits and signaling cascades. Accumulated evidence suggests that altered metabolic programs initiated by the activation and maturation of immune cell types are accompanied by the delivery of various metabolites into the local environment. We propose that, in addition to protein/peptide ligands, secreted immune metabolites (SIMets) are essential components of immune communication networks that fine-tune immune responses under homeostatic and pathological conditions. We summarize recent advances in our understanding of SIMets and discuss the potential mechanisms by which some metabolites engage in immunological responses through receptor-, transporter-, and post-translational-mediated regulation. These insights may contribute to understanding physiology and developing effective therapeutics for inflammatory and immune-mediated diseases.


Subject(s)
Cell Communication , Immune System Diseases , Humans , Signal Transduction , Ligands
2.
Int J Mol Sci ; 23(5)2022 Feb 28.
Article in English | MEDLINE | ID: mdl-35269842

ABSTRACT

Mycobacterium tuberculosis (Mtb) represents a major burden to global health, and refined vaccines are needed. Replication-deficient lymphocytic choriomeningitis virus (rLCMV)-based vaccine vectors against cytomegalovirus have proven safe for human use and elicited robust T cell responses in a large proportion of vaccine recipients. Here, we developed an rLCMV vaccine expressing the Mtb antigens TB10.4 and Ag85B. In mice, rLCMV elicited high frequencies of polyfunctional Mtb-specific CD8 and CD4 T cell responses. CD8 but not CD4 T cells were efficiently boosted upon vector re-vaccination. High-frequency responses were also observed in neonatally vaccinated mice, and co-administration of rLCMV with Expanded Program of Immunization (EPI) vaccines did not result in substantial reciprocal interference. Importantly, rLCMV immunization significantly reduced the lung Mtb burden upon aerosol challenge, resulting in improved lung ventilation. Protection was associated with increased CD8 T cell recruitment but reduced CD4 T cell infiltration upon Mtb challenge. When combining rLCMV with BCG vaccination in a heterologous prime-boost regimen, responses to the rLCMV-encoded Mtb antigens were further augmented, but protection was not significantly different from rLCMV or BCG vaccination alone. This work suggests that rLCMV may show utility for neonatal and/or adult vaccination efforts against pulmonary tuberculosis.


Subject(s)
Mycobacterium tuberculosis , Animals , Antigens, Bacterial , BCG Vaccine , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Lymphocytic choriomeningitis virus/genetics , Mice , Mycobacterium tuberculosis/genetics
3.
Nature ; 599(7885): 471-476, 2021 11.
Article in English | MEDLINE | ID: mdl-34732892

ABSTRACT

Small, soluble metabolites not only are essential intermediates in intracellular biochemical processes, but can also influence neighbouring cells when released into the extracellular milieu1-3. Here we identify the metabolite and neurotransmitter GABA as a candidate signalling molecule synthesized and secreted by activated B cells and plasma cells. We show that B cell-derived GABA promotes monocyte differentiation into anti-inflammatory macrophages that secrete interleukin-10 and inhibit CD8+ T cell killer function. In mice, B cell deficiency or B cell-specific inactivation of the GABA-generating enzyme GAD67 enhances anti-tumour responses. Our study reveals that, in addition to cytokines and membrane proteins, small metabolites derived from B-lineage cells have immunoregulatory functions, which may be pharmaceutical targets allowing fine-tuning of immune responses.


Subject(s)
B-Lymphocytes/metabolism , Interleukin-10/immunology , Macrophages/metabolism , Neoplasms/immunology , gamma-Aminobutyric Acid/metabolism , Animals , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Female , Gene Deletion , Glutamate Decarboxylase/deficiency , Glutamate Decarboxylase/genetics , Humans , Inflammation/immunology , Inflammation/prevention & control , Macrophages/immunology , Male , Mice , Neoplasms/pathology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , gamma-Aminobutyric Acid/biosynthesis
4.
5.
Curr Opin Immunol ; 55: 15-21, 2018 12.
Article in English | MEDLINE | ID: mdl-30248521

ABSTRACT

Many studies describe dysbiosis as a change in the microbiota that accompanies autoimmune illnesses, but little is known about whether these changes are a cause or consequence of an altered immune state. The immune system actively shapes the composition of the microbiota, with divergent outcomes in healthy or autoimmune-prone individuals. The gut microbiota in turn acts as an acquired endocrine organ, influencing the physiology of the host via release of nutrients and chemical messengers. Dysbiosis arising from abnormal immune function can initiate or amplify autoimmunity through multiple mechanisms. We examine how the bidirectional relationship between resident microbes and the immune system contributes to autoimmune diseases.


Subject(s)
Autoimmunity/immunology , Gastrointestinal Microbiome/immunology , Animals , Autoimmune Diseases/immunology , Dysbiosis/immunology , Humans
6.
J Exp Med ; 215(8): 2019-2034, 2018 08 06.
Article in English | MEDLINE | ID: mdl-30042191

ABSTRACT

Immunoglobulin A (IgA) promotes health by regulating the composition and function of gut microbiota, but the molecular requirements for such homeostatic IgA function remain unknown. We found that a heavily glycosylated monoclonal IgA recognizing ovalbumin coats Bacteroides thetaiotaomicron (B. theta), a prominent gut symbiont of the phylum Bacteroidetes. In vivo, IgA alters the expression of polysaccharide utilization loci (PUL), including a functionally uncharacterized molecular family provisionally named Mucus-Associated Functional Factor (MAFF). In both mice and humans, MAFF is detected predominantly in mucus-resident bacteria, and its expression requires the presence of complex microbiota. Expression of the MAFF system facilitates symbiosis with other members of the phylum Firmicutes and promotes protection from a chemically induced model of colitis. Our data reveal a novel mechanism by which IgA promotes symbiosis and colonic homeostasis.


Subject(s)
Bacteria/metabolism , Gastrointestinal Microbiome , Immunoglobulin A/metabolism , Symbiosis , Adult , Aged , Aged, 80 and over , Animals , Antibodies, Monoclonal/metabolism , Bacteria/genetics , Bacteroides/genetics , Bacteroides/physiology , Colon/metabolism , DNA-Binding Proteins , Female , Gene Expression Regulation, Bacterial , Glycosylation , Homeostasis , Humans , Lipopolysaccharides/metabolism , MafF Transcription Factor/metabolism , Male , Mice, Inbred C57BL , Middle Aged , Models, Biological , Mucus/metabolism , Nuclear Proteins/metabolism , Ovalbumin/metabolism , Phenotype
7.
Immunity ; 48(4): 628-631, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29669247

ABSTRACT

Disruption of the gut microbiota is thought to contribute to disease onset in individuals with a genetic predisposition to autoimmunity. In a recent issue of Science, Manfredo Vieira et al. (2018) identify translocation of the gut commensal Enterococcus gallinarum into the liver as a trigger for the autoimmune disease systemic lupus erythematous.


Subject(s)
Autoimmunity , Wolves , Animals , Chickens , Female , Gastrointestinal Microbiome , Humans , Mice
8.
J Infect Dis ; 217(10): 1667-1671, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29471332

ABSTRACT

Persistence of Mycobacterium tuberculosis within human bone marrow stem cells has been identified as a potential bacterial niche during latent tuberculosis. Using a murine model of tuberculosis, we show here that bone marrow stem and progenitor cells containing M. tuberculosis propagated tuberculosis when transferred to naive mice, given that both transferred cells and recipient mice were unable to express inducible nitric oxide synthase, which mediates killing of intracellular bacteria via nitric oxide. Our findings suggest that bone marrow stem and progenitor cells containing M. tuberculosis propagate hallmarks of disease if nitric oxide-mediated killing of bacteria is defective.


Subject(s)
Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/microbiology , Mycobacterium tuberculosis/pathogenicity , Nitric Oxide Synthase Type II/metabolism , Stem Cells/metabolism , Stem Cells/microbiology , Tuberculosis/metabolism , Animals , Disease Models, Animal , Hematopoietic Stem Cell Transplantation/methods , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Tuberculosis/microbiology
9.
PLoS Pathog ; 13(10): e1006676, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29040326

ABSTRACT

Mycobacterium tuberculosis (Mtb) primarily resides in the lung but can also persist in extrapulmonary sites. Macrophages are considered the prime cellular habitat in all tissues. Here we demonstrate that Mtb resides inside adipocytes of fat tissue where it expresses stress-related genes. Moreover, perigonadal fat of Mtb-infected mice disseminated the infection when transferred to uninfected animals. Adipose tissue harbors leukocytes in addition to adipocytes and other cell types and we observed that Mtb infection induces changes in adipose tissue biology depending on stage of infection. Mice infected via aerosol showed infiltration of inducible nitric oxide synthase (iNOS) or arginase 1 (Arg1)-negative F4/80+ cells, despite recruitment of CD3+, CD4+ and CD8+ T cells. Gene expression analysis of adipose tissue of aerosol Mtb-infected mice provided evidence for upregulated expression of genes associated with T cells and NK cells at 28 days post-infection. Strikingly, IFN-γ-producing NK cells and Mtb-specific CD8+ T cells were identified in perigonadal fat, specifically CD8+CD44-CD69+ and CD8+CD44-CD103+ subpopulations. Gene expression analysis of these cells revealed that they expressed IFN-γ and the lectin-like receptor Klrg1 and down-regulated CD27 and CD62L, consistent with an effector phenotype of Mtb-specific CD8+ T cells. Sorted NK cells expressed higher abundance of Klrg1 upon infection, as well. Our results reveal the ability of Mtb to persist in adipose tissue in a stressed state, and that NK cells and Mtb-specific CD8+ T cells infiltrate infected adipose tissue where they produce IFN-γ and assume an effector phenotype. We conclude that adipose tissue is a potential niche for Mtb and that due to infection CD8+ T cells and NK cells are attracted to this tissue.


Subject(s)
Adipose Tissue/immunology , Adipose Tissue/microbiology , Tuberculosis/immunology , Tuberculosis/microbiology , Virus Latency/immunology , Adipocytes/microbiology , Animals , CD8-Positive T-Lymphocytes/immunology , Humans , Killer Cells, Natural/immunology , Mice , Mycobacterium tuberculosis/immunology
10.
Nat Immunol ; 18(12): 1342-1352, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29058703

ABSTRACT

T cells reorganize their metabolic profiles after being activated, but the systemic metabolic effect of sustained activation of the immune system has remained unexplored. Here we report that augmented T cell responses in Pdcd1-/- mice, which lack the inhibitory receptor PD-1, induced a metabolic serum signature characterized by depletion of amino acids. We found that the depletion of amino acids in serum was due to the accumulation of amino acids in activated Pdcd1-/- T cells in the lymph nodes. A systemic decrease in tryptophan and tyrosine led to substantial deficiency in the neurotransmitters serotonin and dopamine in the brain, which resulted in behavioral changes dominated by anxiety-like behavior and exacerbated fear responses. Together these data indicate that excessive activation of T cells causes a systemic metabolomic shift with consequences that extend beyond the immune system.


Subject(s)
Anxiety/physiopathology , Behavior, Animal/physiology , Fear/physiology , Lymphocyte Activation/immunology , Programmed Cell Death 1 Receptor/genetics , T-Lymphocytes/immunology , Amino Acids/blood , Animals , Brain/metabolism , Dopamine/deficiency , Interferon-gamma/blood , Kynurenine/blood , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/deficiency , Serotonin/deficiency , T-Lymphocytes/metabolism , Tryptophan/metabolism , Tyrosine/metabolism
11.
Nat Commun ; 8: 14647, 2017 03 17.
Article in English | MEDLINE | ID: mdl-28303891

ABSTRACT

T follicular regulatory (Tfr) cells control the magnitude and specificity of the germinal centre reaction, but how regulation is contained to ensure generation of high-affinity antibody is unknown. Here we show that this balance is maintained by the reciprocal influence of interleukin (IL)-2 and IL-21. The number of IL-2-dependent FoxP3+ regulatory T cells is increased in the peripheral blood of human patients with loss-of-function mutations in the IL-21 receptor (IL-21R). In mice, IL-21:IL-21R interactions influence the phenotype of T follicular cells, reducing the expression of CXCR4 and inhibiting the expansion of Tfr cells after T-cell-dependent immunization. The negative effect of IL-21 on Tfr cells in mice is cell intrinsic and associated with decreased expression of the high affinity IL-2 receptor (CD25). Bcl-6, expressed in abundance in Tfr cells, inhibits CD25 expression and IL-21-mediated inhibition of CD25 is Bcl-6 dependent. These findings identify a mechanism by which IL-21 reinforces humoral immunity by restricting Tfr cell proliferation.


Subject(s)
Cell Proliferation/genetics , Immunity, Humoral/genetics , Interleukin-21 Receptor alpha Subunit/genetics , Interleukin-2/immunology , Interleukins/genetics , Proto-Oncogene Proteins c-bcl-6/immunology , T-Lymphocytes, Regulatory/immunology , Adolescent , Animals , Child , Female , Humans , Immunity, Humoral/immunology , Immunologic Deficiency Syndromes/genetics , Infant , Interleukin-21 Receptor alpha Subunit/immunology , Interleukins/immunology , Male , Mice , Mice, Knockout , Receptors, CXCR4/immunology
12.
PLoS One ; 12(1): e0169119, 2017.
Article in English | MEDLINE | ID: mdl-28046053

ABSTRACT

An estimated third of the world's population is latently infected with Mycobacterium tuberculosis (Mtb), with no clinical signs of tuberculosis (TB), but lifelong risk of reactivation to active disease. The niches of persisting bacteria during latent TB infection remain unclear. We detect Mtb DNA in peripheral blood selectively in long-term repopulating pluripotent hematopoietic stem cells (LT-pHSCs) as well as in mesenchymal stem cells from latently infected human donors. In mice infected with low numbers of Mtb, that do not develop active disease we, again, find LT-pHSCs selectively infected with Mtb. In human and mouse LT-pHSCs Mtb are stressed or dormant, non-replicating bacteria. Intratracheal injection of Mtb-infected human and mouse LT-pHSCs into immune-deficient mice resuscitates Mtb to replicating bacteria within the lung, accompanied by signs of active infection. We conclude that LT-pHSCs, together with MSCs of Mtb-infected humans and mice serve as a hitherto unappreciated quiescent cellular depot for Mtb during latent TB infection.


Subject(s)
Hematopoietic Stem Cells/microbiology , Latent Tuberculosis/microbiology , Mesenchymal Stem Cells/microbiology , Mycobacterium tuberculosis , Adult , Animals , Antigens, CD34/metabolism , Bone Marrow Cells/metabolism , Cell Separation , Female , Flow Cytometry , Humans , Lung/microbiology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Signaling Lymphocytic Activation Molecule Family Member 1/metabolism , Young Adult
13.
mBio ; 7(6)2016 11 22.
Article in English | MEDLINE | ID: mdl-27879332

ABSTRACT

Mycobacterium bovis Bacille Calmette-Guérin (BCG) is the only licensed vaccine against tuberculosis (TB), yet its moderate efficacy against pulmonary TB calls for improved vaccination strategies. Mucosal BCG vaccination generates superior protection against TB in animal models; however, the mechanisms of protection remain elusive. Tissue-resident memory T (TRM) cells have been implicated in protective immune responses against viral infections, but the role of TRM cells following mycobacterial infection is unknown. Using a mouse model of TB, we compared protection and lung cellular infiltrates of parenteral and mucosal BCG vaccination. Adoptive transfer and gene expression analyses of lung airway cells were performed to determine the protective capacities and phenotypes of different memory T cell subsets. In comparison to subcutaneous vaccination, intratracheal and intranasal BCG vaccination generated T effector memory and TRM cells in the lung, as defined by surface marker phenotype. Adoptive mucosal transfer of these airway-resident memory T cells into naive mice mediated protection against TB. Whereas airway-resident memory CD4+ T cells displayed a mixture of effector and regulatory phenotype, airway-resident memory CD8+ T cells displayed prototypical TRM features. Our data demonstrate a key role for mucosal vaccination-induced airway-resident T cells in the host defense against pulmonary TB. These results have direct implications for the design of refined vaccination strategies. IMPORTANCE: BCG remains the only licensed vaccine against TB. Parenterally administered BCG has variable efficacy against pulmonary TB, and thus, improved prevention strategies and a more refined understanding of correlates of vaccine protection are required. Induction of memory T cells has been shown to be essential for protective TB vaccines. Mimicking the natural infection route by mucosal vaccination has been known to generate superior protection against TB in animal models; however, the mechanisms of protection have remained elusive. Here we performed an in-depth analysis to dissect the immunological mechanisms associated with superior mucosal protection in the mouse model of TB. We found that mucosal, and not subcutaneous, BCG vaccination generates lung-resident memory T cell populations that confer protection against pulmonary TB. We establish a comprehensive phenotypic characterization of these populations, providing a framework for future vaccine development.


Subject(s)
Administration, Intranasal , BCG Vaccine/administration & dosage , Immunologic Memory , Lung/immunology , T-Lymphocytes/immunology , Tuberculosis/prevention & control , Adjuvants, Immunologic , Adoptive Transfer , Animals , Disease Models, Animal , Injections, Subcutaneous , Mice , Treatment Outcome , Tuberculosis/immunology
14.
Infect Immun ; 84(10): 2914-21, 2016 10.
Article in English | MEDLINE | ID: mdl-27481246

ABSTRACT

The neonatal Fc receptor (FcRn) extends the systemic half-life of IgG antibodies by chaperoning bound Fc away from lysosomal degradation inside stromal and hematopoietic cells. FcRn also transports IgG across mucosal barriers into the lumen, and yet little is known about how FcRn modulates immunity in the lung during homeostasis or infection. We infected wild-type (WT) and FcRn-deficient (fcgrt(-/-)) mice with Pseudomonas aeruginosa or Mycobacterium tuberculosis to investigate whether recycling and transport of IgG via FcRn influences innate and adaptive immunity in the lung in response to bacterial infection. We found that FcRn expression maintains homeostatic IgG levels in lung and leads to preferential secretion of low-affinity IgG ligands into the lumen. Fcgrt(-/-) animals exhibited no evidence of developmental impairment of innate immunity in the lung and were able to efficiently recruit neutrophils in a model of acute bacterial pneumonia. Although local humoral immunity in lung increased independently of the presence of FcRn during tuberculosis, there was nonetheless a strong impact of FcRn deficiency on local adaptive immunity. We show that the quantity and quality of IgG in airways, as well as the abundance of dendritic cells in the lung, are maintained by FcRn. FcRn ablation transiently enhanced local T cell immunity and neutrophil recruitment during tuberculosis, leading to a lower bacterial burden in lung. This novel understanding of tissue-specific modulation of mucosal IgG isotypes in the lung by FcRn sheds light on the role of mucosal IgG in immune responses in the lung during homeostasis and bacterial disease.


Subject(s)
Dendritic Cells/immunology , Histocompatibility Antigens Class I/physiology , Immunoglobulin G/metabolism , Lung , Receptors, Fc/physiology , Tuberculosis/immunology , Adaptive Immunity , Animals , Antigens, CD/metabolism , Bacterial Load , Dendritic Cells/cytology , Disease Models, Animal , Histocompatibility Antigens Class I/metabolism , Immunity, Innate , Immunoglobulin G/immunology , Integrin alpha Chains/metabolism , Lung/cytology , Lung/immunology , Lung/metabolism , Mice , Mice, Transgenic , Mucous Membrane/metabolism , Mycobacterium tuberculosis , Myeloid Cells/metabolism , Pseudomonas Infections/immunology , Pseudomonas aeruginosa , Receptors, Fc/metabolism , Tuberculosis/microbiology
15.
mBio ; 7(3)2016 05 24.
Article in English | MEDLINE | ID: mdl-27222470

ABSTRACT

UNLABELLED: The current tuberculosis (TB) vaccine, Mycobacterium bovis Bacillus Calmette-Guérin (BCG), provides insufficient protection against pulmonary TB. Previously, we generated a listeriolysin-expressing recombinant BCG strain, which to date has successfully completed phase I and phase IIa clinical trials. In an attempt to further improve efficacy, we deleted the antiapoptotic virulence gene nuoG, encoding NADH dehydrogenase 1 subunit G, from BCG ΔureC::hly In vitro, deletion of nuoG unexpectedly led to strongly increased recruitment of the autophagosome marker LC3 to the engulfed vaccine, suggesting that nuoG also affects xenophagic pathways. In mice, BCG ΔureC::hly ΔnuoG vaccination was safer than BCG and improved protection over that of parental BCG ΔureC::hly, significantly reducing TB load in murine lungs, ameliorating pulmonary pathology, and enhancing immune responses. Transcriptome analysis of draining lymph nodes after vaccination with either BCG ΔureC::hly or BCG ΔureC::hly ΔnuoG demonstrated earlier and stronger induction of immune responses than that with BCG SSI and suggested upregulation of inflammasome activation and interferon-induced GTPases. In summary, BCG ΔureC::hly ΔnuoG is a promising next-generation TB vaccine candidate with excellent efficacy and safety. IMPORTANCE: Autophagy and apoptosis are fundamental processes allowing cells to degrade their components or kill themselves, respectively. The immune system has adopted these mechanisms to eliminate intracellular pathogens. Residing in host cells, the causative agent of tuberculosis, Mycobacterium tuberculosis, has evolved strategies to set cellular programs of autophagy and apoptosis "on hold." The mycobacterial gene nuoG was found to prevent host cell apoptosis. We have deleted nuoG in the live vaccine candidate BCG ΔureC::hly, which is in phase II clinical development, to leave cellular apoptosis "on go" upon immunization. In preclinical models, this strategy boosted immunity and improved protection from M. tuberculosis infection. Unexpectedly, we obtained compelling evidence that mycobacterial nuoG facilitates inhibition of autophagic pathways, suggesting a new role for this gene in the host-pathogen interplay in tuberculosis.


Subject(s)
BCG Vaccine/genetics , BCG Vaccine/immunology , Electron Transport Complex I/genetics , Gene Deletion , Mycobacterium bovis/genetics , Mycobacterium bovis/immunology , Tuberculosis, Pulmonary/prevention & control , Animals , BCG Vaccine/administration & dosage , BCG Vaccine/adverse effects , Bacterial Load , Bacterial Proteins/genetics , Gene Expression Profiling , Inflammasomes/immunology , Lung/immunology , Lung/microbiology , Lung/pathology , Lymph Nodes/immunology , Lymph Nodes/microbiology , Mice , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , Vaccination
16.
Am J Respir Cell Mol Biol ; 54(3): 370-83, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26222277

ABSTRACT

The cation channel transient receptor potential vanilloid (TRPV) 4 is expressed in endothelial and immune cells; however, its role in acute lung injury (ALI) is unclear. The functional relevance of TRPV4 was assessed in vivo, in isolated murine lungs, and in isolated neutrophils. Genetic deficiency of TRPV4 attenuated the functional, histological, and inflammatory hallmarks of acid-induced ALI. Similar protection was obtained with prophylactic administration of the TRPV4 inhibitor, GSK2193874; however, therapeutic administration of the TRPV4 inhibitor, HC-067047, after ALI induction had no beneficial effect. In isolated lungs, platelet-activating factor (PAF) increased vascular permeability in lungs perfused with trpv4(+/+) more than with trpv4(-/-) blood, independent of lung genotype, suggesting a contribution of TRPV4 on blood cells to lung vascular barrier failure. In neutrophils, TRPV4 inhibition or deficiency attenuated the PAF-induced increase in intracellular calcium. PAF induced formation of epoxyeicosatrienoic acids by neutrophils, which, in turn, stimulated TRPV4-dependent Ca(2+) signaling, whereas inhibition of epoxyeicosatrienoic acid formation inhibited the Ca(2+) response to PAF. TRPV4 deficiency prevented neutrophil responses to proinflammatory stimuli, including the formation of reactive oxygen species, neutrophil adhesion, and chemotaxis, putatively due to reduced activation of Rac. In chimeric mice, however, the majority of protective effects in acid-induced ALI were attributable to genetic deficiency of TRPV4 in parenchymal tissue, whereas TRPV4 deficiency in circulating blood cells primarily reduced lung myeloperoxidase activity. Our findings identify TRPV4 as novel regulator of neutrophil activation and suggest contributions of both parenchymal and neutrophilic TRPV4 in the pathophysiology of ALI.


Subject(s)
Acute Lung Injury/metabolism , Lung/metabolism , Neutrophil Activation , Neutrophils/metabolism , TRPV Cation Channels/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/prevention & control , Animals , Bone Marrow Transplantation , Calcium Signaling , Capillary Permeability , Disease Models, Animal , Humans , Hydrochloric Acid , Lung/blood supply , Lung/drug effects , Male , Mice, Knockout , Morpholines/pharmacology , Neutrophil Activation/drug effects , Neutrophils/drug effects , Pneumonia/metabolism , Pulmonary Edema/metabolism , Pyrroles/pharmacology , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/deficiency , TRPV Cation Channels/genetics
17.
J Immunol ; 195(11): 5123-35, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26491200

ABSTRACT

The selection of affinity-matured Ab-producing B cells is supported by interactions with T follicular helper (Tfh) cells. In addition to cell surface-expressed molecules, cytokines produced by Tfh cells, such as IL-21 and IL-4, provide B cell helper signals. In this study, we analyze how the fitness of Th cells can influence Ab responses. To do this, we used a model in which IL-21R-sufficient (wild-type [WT]) and -deficient (Il21r(-/-)) Ag-specific Tfh cells were used to help immunodeficient Il21r(-/-) B cells following T-dependent immunization. Il21r(-/-) B cells that had received help from WT Tfh cells, but not from Il21r(-/-) Tfh cells, generated affinity-matured Ab upon recall immunization. This effect was dependent on IL-4 produced in the primary response and associated with an increased fraction of memory B cells. Il21r(-/-) Tfh cells were distinguished from WT Tfh cells by a decreased frequency, reduced conjugate formation with B cells, increased expression of programmed cell death 1, and reduced production of IL-4. IL-21 also influenced responsiveness to IL-4 because expression of both membrane IL-4R and the IL-4-neutralizing soluble (s)IL-4R were reduced in Il21r(-/-) mice. Furthermore, the concentration of sIL-4R was found to correlate inversely with the amount of IgE in sera, such that the highest IgE levels were observed in Il21r(-/-) mice with the least sIL-4R. Taken together, these findings underscore the important collaboration between IL-4 and IL-21 in shaping T-dependent Ab responses.


Subject(s)
B-Lymphocytes/immunology , Interleukin-21 Receptor alpha Subunit/genetics , Interleukin-4/immunology , Interleukins/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antibody Formation/immunology , Immunoglobulin E/blood , Interleukin-21 Receptor alpha Subunit/immunology , Interleukin-4/biosynthesis , Interleukins/biosynthesis , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Receptors, Cell Surface/biosynthesis , Signal Transduction/immunology
18.
mBio ; 5(3): e01262-14, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24895310

ABSTRACT

UNLABELLED: The only tuberculosis (TB) vaccine in use today, bacillus Calmette-Guérin (BCG), provides insufficient protection and can cause adverse events in immunocompromised individuals, such as BCGosis in HIV(+) newborns. We previously reported improved preclinical efficacy and safety of the recombinant vaccine candidate BCG ΔureC::hly, which secretes the pore-forming listeriolysin O of Listeria monocytogenes. Here, we evaluate a second-generation construct, BCG ΔureC::hly Δpdx1, which is deficient in pyridoxine synthase, an enzyme that is required for biosynthesis of the essential cofactor vitamin B6. This candidate was auxotrophic for vitamin B6 in a concentration-dependent manner, as was its survival in vivo. BCG ΔureC::hly Δpdx1 showed markedly restricted dissemination in subcutaneously vaccinated mice, which was ameliorated by dietary supplementation with vitamin B6. The construct was safer in severe combined immunodeficiency mice than the parental BCG ΔureC::hly. A prompt innate immune response to vaccination, measured by secretion of interleukin-6, granulocyte colony-stimulating factor, keratinocyte cytokine, and macrophage inflammatory protein-1α, remained independent of vitamin B6 administration, while acquired immunity, notably stimulation of antigen-specific CD4 T cells, B cells, and memory T cells, was contingent on vitamin B6 administration. The early protection provided by BCG ΔureC::hly Δpdx1 in a murine Mycobacterium tuberculosis aerosol challenge model consistently depended on vitamin B6 supplementation. Prime-boost vaccination increased protection against the canonical M. tuberculosis H37Rv laboratory strain and a clinical isolate of the Beijing/W lineage. We demonstrate that the efficacy of a profoundly attenuated recombinant BCG vaccine construct can be modulated by external administration of a small molecule. This principle fosters the development of safer vaccines required for immunocompromised individuals, notably HIV(+) infants. IMPORTANCE: Mycobacterium tuberculosis can synthesize the essential cofactor vitamin B6, while humans depend on dietary supplementation. Unlike the lipophilic vitamins A, D, and E, water-soluble vitamin B6 is well tolerated at high doses. We generated a vitamin B6 auxotroph of the phase II clinical tuberculosis vaccine candidate bacillus Calmette-Guérin ΔureC::hly. The next-generation candidate was profoundly attenuated compared to the parental strain. Adaptive immunity and protection in mice consistently depended on increased dietary vitamin B6 above the daily required dose. Control of vaccine efficacy via food supplements such as vitamin B6 could provide a fast track toward improved safety. Safer vaccines are urgently needed for HIV-infected individuals at high risk of adverse events in response to live vaccines.


Subject(s)
BCG Vaccine/immunology , Dietary Supplements , Mycobacterium bovis/immunology , Mycobacterium tuberculosis/immunology , Pyridoxine/administration & dosage , Tuberculosis/prevention & control , Vitamin B 6/biosynthesis , Animals , BCG Vaccine/genetics , Disease Models, Animal , Female , Genes, Bacterial , Immunity, Innate , Immunization, Secondary , Immunocompromised Host , Mice , Mutation , Mycobacterium bovis/genetics , Mycobacterium bovis/metabolism , Tuberculosis/mortality , Vaccination
19.
J Infect Dis ; 210(12): 1928-37, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-24943726

ABSTRACT

Bacillus Calmette-Guérin (BCG) has been used for vaccination against tuberculosis for nearly a century. Here, we analyze immunity induced by a live tuberculosis vaccine candidate, recombinant BCG ΔureC::hly vaccine (rBCG), with proven preclinical and clinical safety and immunogenicity. We pursue in-depth analysis of the endogenous mycobacteria-specific CD4(+) T-cell population, comparing the more efficacious rBCG with canonical BCG to determine which T-cell memory responses are prerequisites for superior protection against tuberculosis. rBCG induced higher numbers and proportions of antigen-specific memory CD4(+) T cells than BCG, with a CXCR5(+)CCR7(+) phenotype and low expression of the effector transcription factors T-bet and Bcl-6. We found that the superior protection of rBCG, compared with BCG, correlated with higher proportions and numbers of these central memory T cells and of T follicular helper cells associated with specific antibody responses. Adoptive transfer of mycobacteria-specific central memory T cells validated their critical role in protection against pulmonary tuberculosis.


Subject(s)
BCG Vaccine/immunology , CD4-Positive T-Lymphocytes/immunology , Immunologic Memory , Mycobacterium bovis/immunology , Tuberculosis/prevention & control , Animals , BCG Vaccine/administration & dosage , BCG Vaccine/genetics , Female , Gene Deletion , Male , Mice, Inbred C57BL , Mutagenesis, Insertional , Mycobacterium bovis/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
20.
Eur J Immunol ; 44(8): 2380-93, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24782112

ABSTRACT

General interest in the biological functions of IFN type I in Mycobacterium tuberculosis (Mtb) infection increased after the recent identification of a distinct IFN gene expression signature in tuberculosis (TB) patients. Here, we demonstrate that TB-susceptible mice lacking the receptor for IFN I (IFNAR1) were protected from death upon aerogenic infection with Mtb. Using this experimental model to mimic primary progressive pulmonary TB, we dissected the immune processes affected by IFN I. IFNAR1 signaling did not affect T-cell responses, but markedly altered migration of inflammatory monocytes and neutrophils to the lung. This process was orchestrated by IFNAR1 expressed on both immune and tissue-resident radioresistant cells. IFNAR1-driven TB susceptibility was initiated by augmented Mtb replication and in situ death events, along with CXCL5/CXCL1-driven accumulation of neutrophils in alveoli, followed by the discrete compartmentalization of Mtb in lung phagocytes. Early depletion of neutrophils rescued TB-susceptible mice to levels observed in mice lacking IFNAR1. We conclude that IFN I alters early innate events at the site of Mtb invasion leading to fatal immunopathology. These data furnish a mechanistic explanation for the detrimental role of IFN I in pulmonary TB and form a basis for understanding the complex roles of IFN I in chronic inflammation.


Subject(s)
Interferon Type I/immunology , Lung/immunology , Phagocytes/immunology , Signal Transduction/immunology , Tuberculosis, Pulmonary/immunology , Animals , Cells, Cultured , Chemokine CXCL1/immunology , Chemokine CXCL5/immunology , Inflammation/immunology , Mice , Mice, Inbred C57BL , Monocytes/immunology , Mycobacterium tuberculosis/immunology , Neutrophils/immunology , Pulmonary Alveoli/immunology , Receptor, Interferon alpha-beta/immunology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...