Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1101366, 2023.
Article in English | MEDLINE | ID: mdl-36814927

ABSTRACT

Introduction: The characterization of B. pertussis (Bp) antigen-specific CD4+ T cell cytokine responses should be included in the evaluation of immunogenicity of pertussis vaccines but is often hindered by the lack of standardized robust assays. Methods: To overcome this limitation, we developed a two-step assay comprising a short-term stimulation of fresh whole blood with Bp antigens and cryopreservation of the stimulated cells, followed later on by batch-wise intracellular cytokine analysis by flow cytometry. Blood samples collected from recently acellular (aP) vaccine boosted subjects with a whole-cell- or aP-primed background was incubated for 24 hrs with Pertussis toxin, Filamentous hemagglutinin or a Bp lysate (400µl per stimulation). Antigen-specific IFN-γ-, IL-4/IL-5/IL-13-, IL-17A/IL-17F- and/or IL-22-producing CD4+ T cells were quantified by flow cytometry to reveal Th1, Th2, and Th17-type responses, respectively. The frequencies of IFN-γ-producing CD8+ T cells were also analyzed. Results: We demonstrate high reproducibility of the Bp-specific whole blood intracellular staining assay. The results obtained after cryopreservation of the stimulated and fixed cells were very well correlated to those obtained without cryopreservation, an approach used in our previously published assay. Optimization resulted in high sensitivity thanks to very low non-specific backgrounds, with reliable detection of Bp antigen-specific Th1, Th2 and Th17-type CD4+ T cells, in the lowest range frequency of 0.01-0.03%. Bp antigen-specific IFN-γ+ CD8+ T lymphocytes were also detected. This test is easy to perform, analyse and interpret with the establishment of strict criteria defining Bp antigen responses. Discussion: Thus, this assay appears as a promising test for evaluation of Bp antigen-specific CD4+ T cells induced by current and next generation pertussis vaccines.


Subject(s)
Bordetella pertussis , Whooping Cough , Humans , CD8-Positive T-Lymphocytes , Th1 Cells , Flow Cytometry/methods , Reproducibility of Results , Pertussis Vaccine , Cytokines
2.
Nat Commun ; 11(1): 1055, 2020 02 26.
Article in English | MEDLINE | ID: mdl-32103022

ABSTRACT

Activated caspase-1 and caspase-11 induce inflammatory cell death in a process termed pyroptosis. Here we show that Prostaglandin E2 (PGE2) inhibits caspase-11-dependent pyroptosis in murine and human macrophages. PGE2 suppreses caspase-11 expression in murine and human macrophages and in the airways of mice with allergic inflammation. Remarkably, caspase-11-deficient mice are strongly resistant to developing experimental allergic airway inflammation, where PGE2 is known to be protective. Expression of caspase-11 is elevated in the lung of wild type mice with allergic airway inflammation. Blocking PGE2 production with indomethacin enhances, whereas the prostaglandin E1 analog misoprostol inhibits lung caspase-11 expression. Finally, alveolar macrophages from asthma patients exhibit increased expression of caspase-4, a human homologue of caspase-11. Our findings identify PGE2 as a negative regulator of caspase-11-driven pyroptosis and implicate caspase-4/11 as a critical contributor to allergic airway inflammation, with implications for pathophysiology of asthma.


Subject(s)
Asthma/pathology , Caspases, Initiator/metabolism , Dinoprostone/metabolism , Macrophages/immunology , Pyroptosis/physiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Asthma/immunology , Caspases, Initiator/genetics , Caspases, Initiator/immunology , Cells, Cultured , Drug Synergism , Female , Humans , Indomethacin/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Misoprostol/pharmacology
3.
Article in English | MEDLINE | ID: mdl-31106160

ABSTRACT

Pertussis is a respiratory infectious disease that has been resurged during the last decades. The change from the traditional multi-antigen whole-cell pertussis (wP) vaccines to acellular pertussis (aP) vaccines that consist of a few antigens formulated with alum, appears to be a key factor in the resurgence of pertussis in many countries. Though current aP vaccines have helped to reduce the morbidity and mortality associated with pertussis, they do not provide durable immunity or adequate protection against the disease caused by the current circulating strains of Bordetella pertussis, which have evolved in the face of the selection pressure induced by the vaccines. Based on the hypothesis that a new vaccine containing multiple antigens could overcome deficiencies in the current aP vaccines, we have designed and characterized a vaccine candidate based on outer membrane vesicle (OMVs). Here we show that the OMVs vaccine, but not an aP vaccine, protected mice against lung infection with a circulating pertactin (PRN)-deficient isolate. Using isogenic bacteria that in principle only differ in PRN expression, we found that deficiency in PRN appears to be largely responsible for the failure of the aP vaccine to protect against this circulating clinical isolates. Regarding the durability of induced immunity, we have already reported that the OMV vaccine is able to induce long-lasting immune responses that effectively prevent infection with B. pertussis. Consistent with this, here we found that CD4 T cells with a tissue-resident memory (TRM) cell phenotype (CD44+CD62LlowCD69+ and/or CD103+) accumulated in the lungs of mice 14 days after immunization with 2 doses of the OMVs vaccine. CD4 TRM cells, which have previously been shown to play a critical role sustained protective immunity against B. pertussis, were also detected in mice immunized with wP vaccine, but not in the animals immunized with a commercial aP vaccine. The CD4 TRM cells secreted IFN-γ and IL-17 and were significantly expanded through local proliferation following respiratory challenge of mice with B. pertussis. Our findings that the OMVs vaccine induce respiratory CD4 TRM cells may explain the ability of this vaccine to induce long-term protection and is therefore an ideal candidate for a third generation vaccine against B. pertussis.


Subject(s)
Bordetella pertussis/immunology , CD4-Positive T-Lymphocytes/immunology , Exosomes/immunology , Immunologic Memory , Pertussis Vaccine/immunology , Whooping Cough/prevention & control , Animals , Cytokines/metabolism , Disease Models, Animal , Immunologic Factors/metabolism , Mice , Pertussis Vaccine/administration & dosage , Vaccines, Acellular/administration & dosage , Vaccines, Acellular/immunology
4.
Emerg Microbes Infect ; 8(1): 169-185, 2019.
Article in English | MEDLINE | ID: mdl-30866771

ABSTRACT

Protective immunity wanes rapidly after immunization of children with acellular pertussis (aP) vaccines and these vaccines do not prevent nasal colonization or transmission of Bordetella pertussis in baboons. In this study, we examined the role of tissue-resident memory T (TRM) cells in persistent protective immunity induced by infection or immunization with aP and whole-cell pertussis (wP) vaccines in mice. Immunization of mice with a wP vaccine protected against lung and nasal colonization, whereas an aP vaccine failed to protect in the nose. IL-17 and IFN-γ-secreting CD69+CD4+ TRM cells were expanded in the lung and nasal tissue after B. pertussis challenge of mice immunized with wP, but not aP vaccines. However, previous infection induced the most persistent protection against nasal colonization and this correlated with potent induction of nasal tissue TRM cells, especially IL-17-secreting TRM cells. Blocking T cell migration to respiratory tissue during immunization with a wP vaccine impaired bacterial clearance, whereas transfer of TRM cells from convalescent or wP-immunized mice conferred protection to naïve mice. Our findings reveal that previous infection or wP vaccination are significantly more effective than aP vaccination in conferring persistent protective immunity against B. pertussis and that this is mediated by respiratory TRM cells.


Subject(s)
Bordetella pertussis/immunology , CD4-Positive T-Lymphocytes/immunology , Pertussis Vaccine/administration & dosage , Whooping Cough/prevention & control , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Movement , Immunization , Interleukin-17/metabolism , Lung/microbiology , Mice , Nose/microbiology , Pertussis Vaccine/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Whooping Cough/immunology
5.
EMBO Rep ; 20(5)2019 05.
Article in English | MEDLINE | ID: mdl-30894405

ABSTRACT

Regulatory T (Treg) cells help to maintain tolerance and prevent the development of autoimmune diseases. Retinoic acid (RA) can promote peripheral conversion of naïve T cells into Foxp3+ Treg cells. Here, we show that RA can act as an adjuvant to induce antigen-specific type 1 Treg (Tr1) cells, which is augmented by co-administration of IL-2. Immunization of mice with the model antigen KLH in the presence of RA and IL-2 induces T cells that secrete IL-10, but not IL-17 or IFN-γ, and express LAG-3, CD49b and PD-1 but not Foxp3, a phenotype typical of Tr1 cells. Furthermore, immunization of mice with the autoantigen MOG in the presence of RA and IL-2 induces Tr1 cells, which suppress pathogenic Th1 and Th17 cells that mediate the development of experimental autoimmune encephalomyelitis (EAE), an autoimmune disease of the CNS. Furthermore, immunization with a surrogate autoantigen, RA and IL-2 prevents development of spontaneous autoimmune uveitis. Our findings demonstrate that the induction of autoantigen-specific Tr1 cells can prevent the development of autoimmunity.


Subject(s)
Autoantigens/immunology , Autoimmunity/immunology , T-Lymphocytes, Regulatory/immunology , Tretinoin/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Forkhead Transcription Factors/immunology , Interleukin-10/immunology , Interleukin-17/immunology , Mice , Mice, Inbred C57BL , Th1 Cells/immunology , Th17 Cells/immunology
6.
Mucosal Immunol ; 11(6): 1763-1776, 2018 11.
Article in English | MEDLINE | ID: mdl-30127384

ABSTRACT

Current acellular pertussis (aP) vaccines induce strong antibody and Th2 responses but fail to protect against nasal colonization and transmission of Bordetella pertussis. Furthermore, immunity wanes rapidly after immunization. We have developed a novel adjuvant combination (called LP-GMP), comprising c-di-GMP, an intracellular receptor stimulator of interferon genes (STING) agonist, and LP1569, a TLR2 agonist from B. pertussis, which synergistically induces production of IFN-ß, IL-12 and IL-23, and maturation of dendritic cells. Parenteral immunization of mice with an experimental aP vaccine formulated with LP-GMP promoted Th1 and Th17 responses and conferred protection against lung infection with B. pertussis. Intranasal immunization with the same aP vaccine-induced potent B. pertussis-specific Th17 responses and IL-17-secreting respiratory tissue-resident memory (TRM) CD4 T cells, and conferred a high level of protection against nasal colonization as well as lung infection, which was sustained for at least 10 months. Furthermore, long-term protection against nasal colonization with B. pertussis correlated with the number of IL-17-secreting TRM cells in nasal tissue. Our study has identified an approach for inducing IL-17-secreting TRM cells that sustain sterilizing immunity against nasal colonization of mice with B. pertussis, and could form the basis of a third generation pertussis vaccine for humans.


Subject(s)
Bordetella pertussis/physiology , Interleukin-17/metabolism , Nose/immunology , Pertussis Vaccine/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Whooping Cough/immunology , Adjuvants, Immunologic , Administration, Intranasal , Animals , Cells, Cultured , Humans , Immunity, Cellular , Immunologic Memory , Mice , Mice, Inbred C57BL , Nose/microbiology , Vaccination
7.
Front Immunol ; 9: 1764, 2018.
Article in English | MEDLINE | ID: mdl-30105030

ABSTRACT

Treatment with the macrolide antibiotic azithromycin (AZM) is an important intervention for controlling infection of children with Bordetella pertussis and as a prophylaxis for preventing transmission to family members. However, antibiotics are known to have immunomodulatory effects independent of their antimicrobial activity. Here, we used a mouse model to examine the effects of AZM treatment on clearance of B. pertussis and induction of innate and adaptive immunity. We found that treatment of mice with AZM either 7 or 14 days post challenge effectively cleared the bacteria from the lungs. The numbers of innate immune cells in the lungs were significantly reduced in antibiotic-treated mice. Furthermore, AZM reduced the activation status of macrophages and dendritic cells, but only in mice treated on day 7. Early treatment with antibiotics also reduced the frequency of tissue-resident T cells and IL-17-producing cells in the lungs. To assess the immunomodulatory effects of AZM independent of its antimicrobial activity, mice were antibiotic treated during immunization with a whole cell pertussis (wP) vaccine. Protection against B. pertussis induced by immunization with wP was slightly reduced in AZM-treated mice. Antibiotic-treated wP-immunized mice had reduced numbers of lung-resident memory CD4 T cells and IL-17-production and reduced CD49d expression on splenic CD4 T cells after challenge, suggestive of impaired CD4 T cell memory. Taken together these results suggest that AZM can modulate the induction of memory CD4 T cells during B. pertussis infection, but this may in part be due to the clearance of B. pertussis and resulting loss of components that stimulate innate and adaptive immune response.


Subject(s)
Adaptive Immunity , Azithromycin/pharmacology , Bordetella pertussis/drug effects , Bordetella pertussis/immunology , Immunity, Innate , Immunologic Memory , T-Lymphocytes/immunology , Whooping Cough/immunology , Whooping Cough/microbiology , Animals , Anti-Bacterial Agents/pharmacology , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Cytokines/metabolism , Immunization , Immunomodulation , Lung/drug effects , Lung/immunology , Lung/microbiology , Lung/pathology , Mice , Pertussis Vaccine/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Whooping Cough/metabolism , Whooping Cough/prevention & control
8.
Vaccine ; 35(39): 5256-5263, 2017 09 18.
Article in English | MEDLINE | ID: mdl-28823618

ABSTRACT

A resurgence of whooping cough (pertussis) has been observed in recent years in a number of developed countries, despite widespread vaccine coverage. Although the exact reasons of the recurrence of pertussis are not clear, there are a number of potential causes, like antigenic variation in the circulating strains of Bordetella pertussis, changes in surveillance and diagnostic tools, and potential differences in protection afforded by current acellular pertussis (aP) vaccines compared to more reactogenic whole cell (wP) vaccines, which they replaced. Studies in animal models have shown that induction of cellular as well as humoral immune responses are key to conferring effective and long lasting protection against B. pertussis. wP vaccines induce robust Th1/Th17 responses, which are associated with good protection against lung infection. In contrast, aP vaccines induce mixed Th2/Th17 responses. One research option is to modify current aP vaccines with the intention of inducing protective T cell responses, without compromising on their low reactogenicity profile. Here we found that formulation of an aP vaccine with a novel adjuvant based on a Toll-like receptor 7 agonist (TLR7a) adsorbed to aluminum hydroxide (alum) enhanced B. pertussis-specific Th1 and Th17 responses and serum IgG2a/b antibodies, which had greater functional capacity than those induced by aP formulated with alum alone. Furthermore, addition of a TLR7a enhanced the protective efficacy of the aP vaccine against B. pertussis aerosol challenge; protection was comparable to that of a wP vaccine. These findings suggest that alum-TLR7a is a promising adjuvant for clinical development of next generation pertussis vaccines.


Subject(s)
Membrane Glycoproteins/agonists , Membrane Glycoproteins/metabolism , Pertussis Vaccine/therapeutic use , Th1 Cells/metabolism , Th17 Cells/metabolism , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/metabolism , Adjuvants, Immunologic , Animals , Bordetella pertussis/immunology , Bordetella pertussis/pathogenicity , CHO Cells , Cricetulus , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Immunity, Humoral/immunology , Immunity, Humoral/physiology , Immunoassay , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Th1 Cells/immunology , Th17 Cells/immunology , Vaccination/methods
9.
J Immunol ; 199(1): 233-243, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28533445

ABSTRACT

Th1 and Th17 cells have an established role in protective immunity to Bordetella pertussis, but this evidence is based largely on peripheral T cells. There is emerging evidence that local tissue-resident memory T (TRM) cells that accumulate in tissue following mucosal infection may be crucial for long-term immunity. In this study, we examined the role of respiratory CD4 TRM cells in immunity to B. pertussis Natural immunity to B. pertussis induced by infection is considered long lasting and effective at preventing reinfection. Consistent with this, we found that convalescent mice rapidly cleared the bacteria after reinfection. Furthermore, CD4 T cells with a TRM cell phenotype (CD44+CD62L-CD69+ or CD44+CD62L-CD69+CD103+) accumulated in the lungs of mice during infection with B. pertussis and significantly expanded through local proliferation following reinfection. These CD4 TRM cells were B. pertussis specific and secreted IL-17 or IL-17 and IFN-γ. Treatment of mice with FTY720, which prevented migration of T and B cells from lymph nodes to the circulation, significantly exacerbated B. pertussis infection. This was associated with significantly reduced infiltration of central memory T cells and B cells into the lungs. However, the local expansion of TRM cells and the associated rapid clearance of the secondary infection were not affected by treatment with FTY720 before rechallenge. Moreover, adoptive transfer of lung CD4 TRM cells conferred protection in naive mice. Our findings reveal that Ag-specific CD4 TRM cells play a critical role in adaptive immunity against reinfection and memory induced by natural infection with B. pertussis.


Subject(s)
Adaptive Immunity , Bordetella pertussis/immunology , CD4-Positive T-Lymphocytes/immunology , Immunity, Innate , Immunologic Memory , Lung/immunology , Adoptive Transfer , Animals , B-Lymphocytes/immunology , Cell Proliferation , Fingolimod Hydrochloride/administration & dosage , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Lung/microbiology , Lung/pathology , Mice
10.
J Immunol ; 198(1): 363-374, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27864475

ABSTRACT

γδ T cells play a role in protective immunity to infection at mucosal surface, but also mediate pathology in certain autoimmune diseases through innate IL-17 production. Recent reports have suggested that γδ T cells can have memory analogous to conventional αß T cells. In this study we have examined the role of γδ T cells in immunity to the respiratory pathogen Bordetella pertussis γδ T cells, predominantly Vγ4-γ1- cells, produced IL-17 in the lungs as early as 2 h after infection. The bacterial burden during primary infection was significantly enhanced and the induction of antimicrobial peptides was reduced in the absence of early IL-17. A second peak of γδ T cells is detected in the lungs 7-14 d after challenge and these γδ T cells were pathogen specific. γδ T cells, exclusively Vγ4, from the lungs of infected but not naive mice produced IL-17 in response to heat-killed B. pertussis in the presence of APC. Furthermore, γδ T cells from the lungs of mice reinfected with B. pertussis produced significantly more IL-17 than γδ T cells from infected unprimed mice. γδ T cells with a tissue resident memory T cell phenotype (CD69+CD103+) were expanded in the lungs during infection with B. pertussis and proliferated rapidly after rechallenge of convalescent mice. Our findings demonstrate that lung γδ T cells provide an early source of innate IL-17, which promotes antimicrobial peptide production, whereas pathogen-specific Vγ4 cells function in adaptive immunological memory against B. pertussis.


Subject(s)
Immunologic Memory/immunology , Interleukin-17/biosynthesis , T-Lymphocyte Subsets/immunology , Whooping Cough/immunology , Adaptive Immunity/immunology , Animals , Bordetella pertussis/immunology , Disease Models, Animal , Flow Cytometry , Immunity, Innate/immunology , Interleukin-17/immunology , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, gamma-delta/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocyte Subsets/metabolism , Whooping Cough/metabolism
11.
Cancer Immunol Immunother ; 65(12): 1491-1498, 2016 12.
Article in English | MEDLINE | ID: mdl-27680570

ABSTRACT

The co-inhibitory molecule PD-1 suppresses T cell responses and has been targeted in the treatment of cancer. Here, we examined the role of PD-1 in regulating the balance between regulatory and effector T cells and whether blocking PD-1 could enhance tumour vaccine-induced protective immunity. A significantly higher proportion of tumour-resident T cells expressed PD-1 and Foxp3 compared with T cells in the tumour circulation or draining lymph nodes, and this correlated with a lower frequency of IFN-γ- and TNF-secreting CD8 T cells. Blocking PD-1 with a specific antibody reduced Foxp3+ regulatory T (Treg) cell induction and enhanced proliferation, cytokine production, and tumour killing by CD8 T cells. Treatment of CT26 tumour-bearing mice with anti-PD-1 in combination with a vaccine, comprising heat-shocked irradiated tumour cells and a TLR 7/8 agonist, significantly reduced tumour growth and enhanced survival. Furthermore, surviving mice resisted tumour re-challenge. The rejection of tumours in mice treated with the anti-PD-1 vaccine combination was associated with a reduction in tumour-infiltrating Treg cells and enhancement of IFN-γ-secreting CD8 T cells. Our findings demonstrate that high PD-1 expression correlates with increased tumour-infiltrating Treg cells and reduced effector T cells and that when combined with a potent antigen-adjuvant combination, blocking PD-1 effectively enhances anti-tumour immunity.


Subject(s)
Cancer Vaccines/immunology , Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Disease Models, Animal , Mice , Mice, Inbred BALB C
12.
Immunol Cell Biol ; 94(8): 763-73, 2016 09.
Article in English | MEDLINE | ID: mdl-27089940

ABSTRACT

Retinoic acid (RA) in the steady state enhances induction of Foxp3(+) regulatory T (Treg) cells and inhibits differentiation of Th1 and Th17 cells, thereby maintaining tolerance, but can in inflammatory conditions promote effector Th1 and Th17 cells that mediate inflammation. IL-17-producing γδ T cells have recently been shown to have a major pathogenic role in autoimmune diseases. Here, we examined the immunomodulatory effects of RA on γδ T cells. We found that RA had a dramatic suppressive effect on IL-17A and IL-17F production by γδ T cells stimulated with IL-1ß and IL-23. RA suppressed RORγt, IL-1R and IL-23R expression in γδ T cells. Treatment of mice with RA suppressed IL-17 production by γδ T cells in vivo. Furthermore, treatment of T cells with RA attenuated their ability to induce disease in experimental autoimmune encephalomyelitis (EAE), a murine model for multiple sclerosis. This was associated with a reduction in the number of central nervous system-infiltrating γδ T cells, but also CD4(+) T cells that produced IL-17A, IL-17F or GM-CSF. Interestingly, treatment of γδ T cells with RA or removal of γδ T cells from a bulk population of T cells significantly reduced their capacity to induce EAE, demonstrating a critical role for γδ T cells in promoting pathogenic Th17 cells. Our findings demonstrate that the anti-inflammatory properties of RA are mediated in part by suppressing STAT3-mediated activation of cytokine production and cytokine receptor expression in γδ T cells, which suppresses their ability to activate Th17 cells.


Subject(s)
Central Nervous System/immunology , Interleukin-17/biosynthesis , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Tretinoin/pharmacology , Adoptive Transfer , Animals , Autoimmunity , Central Nervous System/drug effects , Central Nervous System/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Immunologic Factors/pharmacology , Interleukin-1beta/metabolism , Interleukin-23/metabolism , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/immunology
13.
J Exp Med ; 210(6): 1117-24, 2013 Jun 03.
Article in English | MEDLINE | ID: mdl-23690441

ABSTRACT

Retinoic acid (RA), a vitamin A metabolite, modulates mucosal T helper cell responses. Here we examined the role of RA in regulating IL-22 production by γδ T cells and innate lymphoid cells in intestinal inflammation. RA significantly enhanced IL-22 production by γδ T cells stimulated in vitro with IL-1ß or IL-18 and IL-23. In vivo RA attenuated colon inflammation induced by dextran sodium sulfate treatment or Citrobacter rodentium infection. This was associated with a significant increase in IL-22 secretion by γδ T cells and innate lymphoid cells. In addition, RA treatment enhanced production of the IL-22-responsive antimicrobial peptides Reg3ß and Reg3γ in the colon. The attenuating effects of RA on colitis were reversed by treatment with an anti-IL-22 neutralizing antibody, demonstrating that RA mediates protection by enhancing IL-22 production. To define the molecular events involved, we used chromatin immunoprecipitation assays and found that RA promoted binding of RA receptor to the IL-22 promoter in γδ T cells. Our findings provide novel insights into the molecular events controlling IL-22 transcription and suggest that one key outcome of RA signaling may be to shape early intestinal immune responses by promoting IL-22 synthesis by γδ T cells and innate lymphoid cells.


Subject(s)
Colon/immunology , Inflammation/immunology , Interleukins/immunology , Lymphocytes/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes, Helper-Inducer/immunology , Tretinoin/immunology , Animals , Antibodies, Neutralizing/immunology , Citrobacter rodentium/immunology , Colitis/genetics , Colitis/immunology , Colon/metabolism , Dextran Sulfate/adverse effects , Enterobacteriaceae Infections/genetics , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/metabolism , Inflammation/genetics , Inflammation/metabolism , Interleukins/biosynthesis , Interleukins/genetics , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Protein Binding/genetics , Protein Binding/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Retinoic Acid/immunology , Receptors, Retinoic Acid/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Transcription, Genetic/genetics , Transcription, Genetic/immunology , Tretinoin/metabolism , Interleukin-22
14.
PLoS Pathog ; 9(4): e1003264, 2013.
Article in English | MEDLINE | ID: mdl-23592988

ABSTRACT

Whooping cough caused by Bordetella pertussis is a re-emerging infectious disease despite the introduction of safer acellular pertussis vaccines (Pa). One explanation for this is that Pa are less protective than the more reactogenic whole cell pertussis vaccines (Pw) that they replaced. Although Pa induce potent antibody responses, and protection has been found to be associated with high concentrations of circulating IgG against vaccine antigens, it has not been firmly established that host protection induced with this vaccine is mediated solely by humoral immunity. The aim of this study was to examine the relative contribution of Th1 and Th17 cells in host immunity to infection with B. pertussis and in immunity induced by immunization with Pw and Pa and to use this information to help rationally design a more effective Pa. Our findings demonstrate that Th1 and Th17 both function in protective immunity induced by infection with B. pertussis or immunization with Pw. In contrast, a current licensed Pa, administered with alum as the adjuvant, induced Th2 and Th17 cells, but weak Th1 responses. We found that IL-1 signalling played a central role in protective immunity induced with alum-adsorbed Pa and this was associated with the induction of Th17 cells. Pa generated strong antibody and Th2 responses, but was fully protective in IL-4-defective mice, suggesting that Th2 cells were dispensable. In contrast, Pa failed to confer protective immunity in IL-17A-defective mice. Bacterial clearance mediated by Pa-induced Th17 cells was associated with cell recruitment to the lungs after challenge. Finally, protective immunity induced by an experimental Pa could be enhanced by substituting alum with a TLR agonist that induces Th1 cells. Our findings demonstrate that alum promotes protective immunity through IL-1ß-induced IL-17A production, but also reveal that optimum protection against B. pertussis requires induction of Th1, but not Th2 cells.


Subject(s)
Adaptive Immunity , Interleukin-17/metabolism , Interleukin-1beta/metabolism , Th1 Cells/immunology , Th17 Cells/immunology , Whooping Cough/immunology , Alum Compounds/pharmacology , Animals , Bordetella pertussis/immunology , Immunity, Cellular , Interleukin-17/deficiency , Interleukin-17/genetics , Interleukin-4/deficiency , Interleukin-4/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Pertussis Vaccine/immunology , Vaccination , Whooping Cough/microbiology , Whooping Cough/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...