Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
Add more filters

Publication year range
1.
Immunity ; 42(4): 756-66, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25902485

ABSTRACT

Staphylococcus aureus skin colonization is universal in atopic dermatitis and common in cancer patients treated with epidermal growth factor receptor inhibitors. However, the causal relationship of dysbiosis and eczema has yet to be clarified. Herein, we demonstrate that Adam17(fl/fl)Sox9-(Cre) mice, generated to model ADAM17-deficiency in human, developed eczematous dermatitis with naturally occurring dysbiosis, similar to that observed in atopic dermatitis. Corynebacterium mastitidis, S. aureus, and Corynebacterium bovis sequentially emerged during the onset of eczematous dermatitis, and antibiotics specific for these bacterial species almost completely reversed dysbiosis and eliminated skin inflammation. Whereas S. aureus prominently drove eczema formation, C. bovis induced robust T helper 2 cell responses. Langerhans cells were required for eliciting immune responses against S. aureus inoculation. These results characterize differential contributions of dysbiotic flora during eczema formation, and highlight the microbiota-host immunity axis as a possible target for future therapeutics in eczematous dermatitis.


Subject(s)
Dermatitis, Atopic/immunology , Dysbiosis/immunology , Eczema/immunology , Langerhans Cells/immunology , Skin/immunology , T-Lymphocytes, Helper-Inducer/immunology , ADAM Proteins/deficiency , ADAM Proteins/genetics , ADAM Proteins/immunology , ADAM17 Protein , Animals , Anti-Bacterial Agents/pharmacology , Corynebacterium/immunology , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/genetics , Dermatitis, Atopic/microbiology , Dysbiosis/drug therapy , Dysbiosis/genetics , Dysbiosis/microbiology , Eczema/drug therapy , Eczema/genetics , Eczema/microbiology , ErbB Receptors/genetics , ErbB Receptors/immunology , Gene Expression Regulation , Humans , Immunity, Innate , Inflammation/drug therapy , Inflammation/genetics , Inflammation/immunology , Inflammation/microbiology , Integrases/genetics , Integrases/immunology , Langerhans Cells/drug effects , Langerhans Cells/microbiology , Langerhans Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/immunology , Signal Transduction , Skin/drug effects , Skin/microbiology , Skin/pathology , Staphylococcus aureus/immunology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/microbiology , T-Lymphocytes, Helper-Inducer/pathology
2.
Semin Immunol ; 39: 30-34, 2018 10.
Article in English | MEDLINE | ID: mdl-30122362

ABSTRACT

The discovery and wide spread use of vaccines have saved millions of lives in the past few decades. Vaccine adjuvants represent an integral part of the modern vaccines. Despite numerous efforts, however, only a handful of vaccine adjuvants is currently available for human use. A comprehensive understanding of the mechanisms of action of adjuvants is pivotal to harness the potential of existing and new adjuvants in mounting desirable immune responses to counter human pathogens. Decomposing the host response to vaccines and its components at systems level has recently been made possible owing to the recent advancements in Omics technology and cutting edge immunological assays powered by systems biology approaches. This approach has begun to shed light on the molecular signatures of several human vaccines and adjuvants. This review is an attempt to provide an overview of the recent efforts in systems analysis of vaccine adjuvants that are currently in clinic.


Subject(s)
Adjuvants, Immunologic/pharmacology , HIV Infections/prevention & control , Immunogenicity, Vaccine , Influenza, Human/prevention & control , Malaria, Falciparum/prevention & control , Systems Analysis , Adjuvants, Immunologic/chemistry , Animals , Drug Combinations , Glucosides/chemistry , Glucosides/pharmacology , HIV Infections/immunology , HIV Infections/virology , Humans , Immunity, Innate/drug effects , Influenza, Human/immunology , Influenza, Human/virology , Lipid A/chemistry , Lipid A/pharmacology , Liposomes/administration & dosage , Liposomes/chemistry , Liposomes/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Polysorbates/chemistry , Polysorbates/pharmacology , Squalene/chemistry , Squalene/pharmacology , Systems Biology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/microbiology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Vaccines/administration & dosage , Vaccines/chemistry , Vaccines/immunology , alpha-Tocopherol/chemistry , alpha-Tocopherol/pharmacology
3.
Semin Immunol ; 39: 22-29, 2018 10.
Article in English | MEDLINE | ID: mdl-30366662

ABSTRACT

Lipopolysaccharide (LPS) is a well-defined agonist of Toll-like receptor (TLR) 4 that activates innate immune responses and influences the development of the adaptive response during infection with Gram-negative bacteria. Many years ago, Dr. Edgar Ribi separated the adjuvant activity of LPS from its toxic effects, an effort that led to the development of monophosphoryl lipid A (MPL). MPL, derived from Salmonella minnesota R595, has progressed through clinical development and is now used in various product-enabling formulations to support the generation of antigen-specific responses in several commercial and preclinical vaccines. We have generated several synthetic lipid A molecules, foremost glucopyranosyl lipid adjuvant (GLA) and second-generation lipid adjuvant (SLA), and have advanced these to clinical trial for various indications. In this review we summarize the potential and current positioning of TLR4-based adjuvant formulations in approved and emerging vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Alum Compounds/pharmacology , Glucosides/pharmacology , Immunogenicity, Vaccine , Lipid A/analogs & derivatives , Tuberculosis/prevention & control , Adjuvants, Immunologic/chemistry , Alum Compounds/chemistry , Animals , Glucosides/chemistry , HIV Infections/immunology , HIV Infections/prevention & control , HIV Infections/virology , Humans , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Leishmaniasis/immunology , Leishmaniasis/parasitology , Leishmaniasis/prevention & control , Leprosy/immunology , Leprosy/parasitology , Leprosy/prevention & control , Lipid A/chemistry , Lipid A/pharmacology , Liposomes/administration & dosage , Liposomes/chemistry , Liposomes/immunology , Malaria/immunology , Malaria/parasitology , Malaria/prevention & control , Mice , Schistosomiasis/immunology , Schistosomiasis/parasitology , Schistosomiasis/prevention & control , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/microbiology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Tuberculosis/immunology , Tuberculosis/microbiology , Vaccines/administration & dosage , Vaccines/chemistry , Vaccines/immunology
4.
Infect Immun ; 88(8)2020 07 21.
Article in English | MEDLINE | ID: mdl-32393507

ABSTRACT

The intracellular bacterial pathogen Salmonella is able to evade the immune system and persist within the host. In some cases, these persistent infections are asymptomatic for long periods and represent a significant public health hazard because the hosts are potential chronic carriers, yet the mechanisms that control persistence are incompletely understood. Using a mouse model of chronic typhoid fever combined with major histocompatibility complex (MHC) class II tetramers to interrogate endogenous, Salmonella-specific CD4+ helper T cells, we show that certain host microenvironments may favorably contribute to a pathogen's ability to persist in vivo We demonstrate that the environment in the hepatobiliary system may contribute to the persistence of Salmonella enterica subsp. enterica serovar Typhimurium through liver-resident immunoregulatory CD4+ helper T cells, alternatively activated macrophages, and impaired bactericidal activity. This contrasts with lymphoid organs, such as the spleen and mesenteric lymph nodes, where these same cells appear to have a greater capacity for bacterial killing, which may contribute to control of bacteria in these organs. We also found that, following an extended period of infection of more than 2 years, the liver appeared to be the only site that harbored Salmonella bacteria. This work establishes a potential role for nonlymphoid organ immunity in regulating chronic bacterial infections and provides further evidence for the hepatobiliary system as the site of chronic Salmonella infection.


Subject(s)
Host-Pathogen Interactions/immunology , Liver/immunology , Salmonella Infections, Animal/immunology , Salmonella typhimurium/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Chronic Disease , Coculture Techniques , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/immunology , Gallbladder/immunology , Gallbladder/microbiology , Gene Expression Regulation/immunology , Host-Pathogen Interactions/genetics , Immunity, Innate , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Liver/microbiology , Lymph Nodes/immunology , Lymph Nodes/microbiology , Macrophage Activation , Mice , Mice, Inbred C57BL , Organ Specificity , RAW 264.7 Cells , Salmonella Infections, Animal/genetics , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/pathology , Salmonella typhimurium/growth & development , Salmonella typhimurium/pathogenicity , Single-Cell Analysis , Spleen/immunology , Spleen/microbiology , T-Lymphocytes, Helper-Inducer/microbiology
5.
Scand J Immunol ; 91(4): e12857, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31811655

ABSTRACT

Since their discovery about 10 years ago, Th9 cells have been increasingly linked to allergic pathologies. Within this review, we summarize the current knowledge on associations between Th9 cells and allergic diseases and acknowledge Th9 cells as important targets in future treatment of allergic diseases. However, until today, it is not fully understood how these Th9 cell responses are modulated. We describe current literature suggesting that these Th9 cell responses might be stimulated by microbial species such as Staphylococcus aureus and Candida albicans, while on the other hand, microbial and dietary compounds such as retinoic acid (RA), butyrate and vitamin D show suppressive capacity on allergy-related Th9 responses. By reviewing this recent research, we provide new insights into the modulating capacity of the microbiota on Th9 cell responses. Consequently, microbial and dietary factors may be used as innovative tools to target Th9 cells in the treatment of allergic diseases. However, further research is needed to elucidate the mechanisms behind these interactions in order to translate this knowledge into clinical allergy settings.


Subject(s)
Hypersensitivity/immunology , Hypersensitivity/microbiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/microbiology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/microbiology , Animals , Humans
6.
Curr Opin Lipidol ; 30(2): 125-133, 2019 04.
Article in English | MEDLINE | ID: mdl-30664014

ABSTRACT

PURPOSE OF REVIEW: Not all of the risk of cardiovascular disease can be explained by diet and genetics, and the human microbiome, which lies at the interface of these two factors, may help explain some of the unaccounted risk. This review examines some of the well established links between the microbiome and cardiovascular health, and proposes relatively unexplored associations. RECENT FINDINGS: Byproducts of microbial metabolism are associated with health and disease: Trimethylamine N oxide is associated with atherosclerosis; whereas short-chain fatty acids are associated with decreased inflammation and increased energy expenditure. More broadly, a large number of association studies have been conducted to explore the connections between bacterial taxa and metabolic syndrome. In contrast, the relationship between the microbiome and triglycerides levels remains poorly understood. SUMMARY: We suggest that deeper understanding of the molecular mechanisms that drive linkages between the microbiome and disease can be determined by replacing 16S rRNA gene sequencing with shotgun metagenomic sequencing or other functional approaches. Furthermore, to ensure translatability and reproducibility of research findings, a combination of multiple different complementary '-omic' approaches should be employed.


Subject(s)
Atherosclerosis/microbiology , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/immunology , Lipid Metabolism/immunology , Metabolic Syndrome/microbiology , Methylamines/metabolism , Animals , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Bile Acids and Salts/immunology , Bile Acids and Salts/metabolism , Carnitine/immunology , Carnitine/metabolism , Choline/immunology , Choline/metabolism , Energy Metabolism/genetics , Energy Metabolism/immunology , Fatty Acids, Volatile/immunology , Gastrointestinal Microbiome/genetics , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Humans , Lipid Metabolism/genetics , Metabolic Syndrome/genetics , Metabolic Syndrome/immunology , Metabolic Syndrome/pathology , Methylamines/immunology , Methylamines/pharmacology , Phosphatidylcholines/immunology , Phosphatidylcholines/metabolism , RNA, Ribosomal, 16S/genetics , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/microbiology , Triglycerides/immunology , Triglycerides/metabolism
7.
J Med Virol ; 91(3): 411-418, 2019 03.
Article in English | MEDLINE | ID: mdl-30192392

ABSTRACT

Altered immune parameters associated with hepatitis C virus (HCV) genotype 1b infection and their correlation with virus eradication in direct-acting antivirals (DAA)-treated patients were examined. Thirty-one HCV-infected patients were treated with DAAs for 12 weeks. Pre-DAA-treatment and post-DAA-treatment sera were analyzed for cytokines/chemokines using MILLIPLEX MAP. Serum complement level and antibody neutralization activity were measured separately. Sera from 11 spontaneously cleared HCV subjects were included for comparison. Rapid virological responders (RVR) or end-of-treatment responders (EOTR) were defined as patients with HCV RNA negative at week 4 or positive at week 4 and negative at week 12, respectively. HCV RNA eradication and a decrease in liver fibrosis-related cytokines after treatment were observed when compared with pretreatment sera from RVR and EOTR. In pretreatment sera, interferons and T-helper 1 or 2 cell-associated cytokines/chemokines were significantly higher among RVR as compared with EOTR. Furthermore, serum complement and virus neutralizing antibody levels were higher in pretreatment RVR sera. Eradication of HCV RNA by DAA decreased liver fibrosis-related cytokines. Pretreatment sera from RVR displayed an enhanced cytokine/chemokine, complement and virus neutralizing antibody response as compared with EOTR sera. Our results suggested that enhanced host immune status may play an additive role on HCV RNA clearance by DAA.


Subject(s)
Antiviral Agents/therapeutic use , Hepatitis C, Chronic/immunology , Hepatitis C/drug therapy , Hepatitis C/immunology , Immunity, Innate , Aged , Chemokines/blood , Chemokines/immunology , Cytokines/blood , Cytokines/immunology , Female , Genotype , Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Humans , Interferon-alpha/therapeutic use , Male , Middle Aged , Neutralization Tests , Polyethylene Glycols , RNA, Viral/blood , Ribavirin/therapeutic use , T-Lymphocytes, Helper-Inducer/microbiology , Treatment Outcome , Viral Load
8.
Immunity ; 31(4): 536-8, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19833084

ABSTRACT

Microbes appear to modulate homeostatic plasticity of T helper and T regulatory cells. In this issue of Immunity, Gaboriau-Routhiau et al. (2009) now reveal that segmented filamentous bacteria uniquely coordinate the intestinal T cell profile. The potential implications of this process to various immune functions are discussed.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Intestines/immunology , Intestines/microbiology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Bacteria/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/microbiology , Cytokines/immunology , Cytokines/metabolism , Humans , Intestinal Mucosa/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/microbiology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/microbiology
9.
Immunity ; 31(6): 974-85, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-20064452

ABSTRACT

Immunity to Mycobacterium tuberculosis in humans and in mice requires interferon gamma (IFN-gamma). Whereas IFN-gamma has been studied extensively for its effects on macrophages in tuberculosis, we determined that protective immunity to tuberculosis also requires IFN-gamma-responsive nonhematopoietic cells. Bone marrow chimeric mice with IFN-gamma-unresponsive lung epithelial and endothelial cells exhibited earlier mortality and higher bacterial burdens than control mice, underexpressed indoleamine-2,3-dioxygenase (Ido1) in lung endothelium and epithelium, and overexpressed interleukin-17 (IL-17) with massive neutrophilic inflammation in the lungs. We also found that the products of IDO catabolism of tryptophan selectively inhibit IL-17 production by Th17 cells, by inhibiting the action of IL-23. These results reveal a previously unsuspected role for IFN-gamma responsiveness in nonhematopoietic cells in regulation of immunity to M. tuberculosis and illustrate the role of IDO in the inhibition of Th17 cell responses.


Subject(s)
Endothelial Cells/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interferon-gamma/metabolism , Mycobacterium tuberculosis/immunology , Respiratory Mucosa/immunology , Tuberculosis, Pulmonary/immunology , Animals , Bacteremia/immunology , Bacteremia/microbiology , Cells, Cultured , Endothelial Cells/microbiology , Endothelial Cells/pathology , Female , Gene Expression Profiling , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interleukin-17/metabolism , Interleukin-23/immunology , Interleukin-23/metabolism , Kynurenine/immunology , Kynurenine/metabolism , Lung/immunology , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/immunology , Oligonucleotide Array Sequence Analysis , Pneumonia, Bacterial/enzymology , Pneumonia, Bacterial/immunology , Receptors, Interferon/genetics , Respiratory Mucosa/microbiology , Respiratory Mucosa/pathology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/microbiology , Tuberculosis, Pulmonary/enzymology , Interferon gamma Receptor
10.
Immunity ; 31(2): 321-30, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19682928

ABSTRACT

Gammadelta T cells are an innate source of interleukin-17 (IL-17), preceding the development of the adaptive T helper 17 (Th17) cell response. Here we show that IL-17-producing T cell receptor gammadelta (TCRgammadelta) T cells share characteristic features with Th17 cells, such as expression of chemokine receptor 6 (CCR6), retinoid orphan receptor (RORgammat), aryl hydrocarbon receptor (AhR), and IL-23 receptor. AhR expression in gammadelta T cells was essential for the production of IL-22 but not for optimal IL-17 production. In contrast to Th17 cells, CCR6(+)IL-17-producing gammadelta T cells, but not other gammadelta T cells, express Toll-like receptors TLR1 and TLR2, as well as dectin-1, but not TLR4 and could directly interact with certain pathogens. This process was amplified by IL-23 and resulted in expansion, increased IL-17 production, and recruitment of neutrophils. Thus, innate receptor expression linked with IL-17 production characterizes TCRgammadelta T cells as an efficient first line of defense that can orchestrate an inflammatory response to pathogen-derived as well as environmental signals long before Th17 cells have sensed bacterial invasion.


Subject(s)
Bacterial Infections/immunology , Interleukin-17/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors , Interleukin-17/metabolism , Interleukin-23/genetics , Interleukin-23/immunology , Interleukin-23/metabolism , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-6/metabolism , Interleukins/immunology , Interleukins/metabolism , Lectins, C-Type , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Nuclear Receptor Subfamily 1, Group F, Member 3 , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/immunology , Receptors, Aryl Hydrocarbon/metabolism , Receptors, CCR6/immunology , Receptors, CCR6/metabolism , Receptors, Retinoic Acid/immunology , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/immunology , Receptors, Thyroid Hormone/metabolism , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/microbiology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/microbiology , Toll-Like Receptor 1/immunology , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism , Interleukin-22
11.
Infect Immun ; 83(12): 4594-603, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26371121

ABSTRACT

Pneumocystis is a respiratory fungal pathogen that causes pneumonia (Pneumocystis pneumonia [PcP]) in immunocompromised patients. Alveolar macrophages are critical effectors for CD4(+) T cell-dependent clearance of Pneumocystis, and previous studies found that alternative macrophage activation accelerates fungal clearance during PcP-related immune reconstitution inflammatory syndrome (IRIS). However, the requirement for either classically or alternatively activated macrophages for Pneumocystis clearance has not been determined. Therefore, RAG2(-/-) mice lacking either the interferon gamma (IFN-γ) receptor (IFN-γR) or interleukin 4 receptor alpha (IL-4Rα) were infected with Pneumocystis. These mice were then immune reconstituted with wild-type lymphocytes to preserve the normal T helper response while preventing downstream effects of Th1 or Th2 effector cytokines on macrophage polarization. As expected, RAG2(-/-) mice developed severe disease but effectively cleared Pneumocystis and resolved IRIS. Neither RAG/IFN-γR(-/-) nor RAG/IL-4Rα(-/-) mice displayed impaired Pneumocystis clearance. However, RAG/IFN-γR(-/-) mice developed a dysregulated immune response, with exacerbated IRIS and greater pulmonary function deficits than those in RAG2 and RAG/IL-4Rα(-/-) mice. RAG/IFN-γR(-/-) mice had elevated numbers of lung CD4(+) T cells, neutrophils, eosinophils, and NK cells but severely depressed numbers of lung CD8(+) T suppressor cells. Impaired lung CD8(+) T cell responses in RAG/IFN-γR(-/-) mice were associated with elevated lung IFN-γ levels, and neutralization of IFN-γ restored the CD8 response. These data demonstrate that restricting the ability of macrophages to polarize in response to Th1 or Th2 cytokines does not impair Pneumocystis clearance. However, a cell type-specific IFN-γ/IFN-γR-dependent mechanism regulates CD8(+) T suppressor cell recruitment, limits immunopathogenesis, preserves lung function, and enhances the resolution of PcP-related IRIS.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immune Reconstitution Inflammatory Syndrome/immunology , Macrophages, Alveolar/immunology , Pneumocystis/immunology , Pneumonia, Pneumocystis/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , CD8-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/pathology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Eosinophils/immunology , Eosinophils/microbiology , Eosinophils/pathology , Gene Expression Regulation , Host-Pathogen Interactions , Immune Reconstitution Inflammatory Syndrome/genetics , Immune Reconstitution Inflammatory Syndrome/microbiology , Immune Reconstitution Inflammatory Syndrome/pathology , Killer Cells, Natural/immunology , Killer Cells, Natural/microbiology , Killer Cells, Natural/pathology , Lung/immunology , Lung/microbiology , Lung/pathology , Macrophage Activation , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/pathology , Mice , Mice, Knockout , Mice, SCID , Neutrophils/immunology , Neutrophils/microbiology , Neutrophils/pathology , Pneumocystis/pathogenicity , Pneumonia, Pneumocystis/genetics , Pneumonia, Pneumocystis/microbiology , Pneumonia, Pneumocystis/pathology , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Signal Transduction , T-Lymphocytes, Helper-Inducer/microbiology , T-Lymphocytes, Helper-Inducer/pathology , Th1-Th2 Balance , Interferon gamma Receptor
12.
J Immunol ; 190(6): 2692-701, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23401589

ABSTRACT

Streptococcus pyogenes (group A Streptococcus [GAS]) is a leading human pathogen associated with a diverse array of mucosal and systemic infections. Vaccination with J8, a conserved region synthetic peptide derived from the M-protein of GAS and containing only 12 aa from GAS, when conjugated to diphtheria toxoid, has been shown to protect mice against a lethal GAS challenge. Protection has been previously shown to be Ab-mediated. J8 does not contain a dominant GAS-specific T cell epitope. The current study examined long-term Ab memory and dissected the role of B and T cells. Our results demonstrated that vaccination generates specific memory B cells (MBC) and long-lasting Ab responses. The MBC response can be activated following boost with Ag or limiting numbers of whole bacteria. We further show that these memory responses protect against systemic infection with GAS. T cell help is required for activation of MBC but can be provided by naive T cells responding directly to GAS at the time of infection. Thus, individuals whose T cells do not recognize the short synthetic peptide in the vaccine will be able to generate a protective and rapid memory Ab response at the time of infection. These studies significantly strengthen previous findings, which showed that protection by the J8-diphtheria toxoid vaccine is Ab-mediated and suggest that in vaccine design for other organisms the source of T cell help for Ab responses need not be limited to sequences from the organism itself.


Subject(s)
Antibodies, Bacterial/biosynthesis , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/microbiology , Immunologic Memory/immunology , Streptococcal Infections/prevention & control , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/microbiology , Vaccines, Subunit/administration & dosage , Animals , Antibodies, Bacterial/administration & dosage , B-Lymphocyte Subsets/metabolism , Immunization, Secondary/methods , Mice , Mice, Inbred BALB C , Mice, SCID , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Time Factors , Vaccines, Subunit/immunology , Vaccines, Subunit/therapeutic use
13.
Immunology ; 143(2): 277-86, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24773322

ABSTRACT

To prevent the global spread of tuberculosis (TB), more effective vaccines and vaccination strategies are urgently needed. As a result of the success of bacillus Calmette-Guérin (BCG) in protecting children against miliary and meningeal TB, the majority of individuals will have been vaccinated with BCG; hence, boosting BCG-primed immunity will probably be a key component of future vaccine strategies. In this study, we compared the ability of DNA-, protein- and lentiviral vector-based vaccines that express the antigens Ag85B and Rv3425 to boost the effects of BCG in the context of immunity and protection against Mycobacterium tuberculosis in C57BL/6 mice. Our results demonstrated that prime-boost BCG vaccination with a lentiviral vector expressing the antigens Ag85B and Rv3425 significantly enhanced immune responses, including T helper type 1 and CD8(+) cytotoxic T lymphocyte responses, compared with DNA- and protein-based vaccines. However, lentivirus-vectored and DNA-based vaccines greatly improved the protective efficacy of BCG against M. tuberculosis, as indicated by a lack of weight loss and significantly reduced bacterial loads and histological damage in the lung. Our study suggests that the use of lentiviral or DNA vaccines containing the antigens Ag85B and Rv3425 to boost BCG is a good choice for the rational design of an efficient vaccination strategy against TB.


Subject(s)
Acyltransferases/immunology , Antigens, Bacterial/immunology , BCG Vaccine/immunology , Bacterial Proteins/immunology , Genetic Vectors , Lentivirus/genetics , Mycobacterium tuberculosis/immunology , Tuberculosis/prevention & control , Vaccination , Acyltransferases/administration & dosage , Acyltransferases/genetics , Animals , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/genetics , BCG Vaccine/administration & dosage , Bacterial Load , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , Cytokines/metabolism , Disease Models, Animal , Female , Lung/immunology , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/microbiology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/microbiology , Time Factors , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/pathology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
14.
Immunology ; 141(4): 514-30, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24138116

ABSTRACT

Pneumococcal surface adhesin A (PsaA) is a multifunctional lipoprotein known to bind nasopharyngeal epithelial cells, and is significantly involved in bacterial adherence and virulence. Identification of PsaA peptides that optimally bind human leucocyte antigen (HLA) and elicit a potent immune response would be of great importance to vaccine development. However, this is hindered by the multitude of HLA polymorphisms in humans. To identify the conserved immunodominant epitopes, we used an experimental dataset of 28 PsaA synthetic peptides and in silico methods to predict specific peptide-binding to HLA and murine MHC class II molecules. We also characterized spleen and cervical lymph node (CLN) -derived T helper (Th) lymphocyte cytokine responses to these peptides after Streptococcus pneumoniae strain EF3030 challenge in mice. Individual, yet overlapping, peptides 15 amino acids in length revealed residues of PsaA that consistently caused the highest interferon-γ, interleukin-2 (IL-2), IL-5 and IL-17 responses and proliferation as well as moderate IL-10 and IL-4 responses by ex vivo re-stimulated splenic and CLN CD4⁺ T cells isolated from S. pneumoniae strain EF3030-challenged F1 (B6 × BALB/c) mice. In silico analysis revealed that peptides from PsaA may interact with a broad range of HLA-DP, -DQ and -DR alleles, due in part to regions lacking ß-turns and asparagine endopeptidase sites. These data suggest that Th cell peptides (7, 19, 20, 22, 23 and 24) screened for secondary structures and MHC class II peptide-binding affinities can elicit T helper cytokine and proliferative responses to PsaA peptides.


Subject(s)
Bacterial Proteins/immunology , Epitope Mapping , Epitopes, T-Lymphocyte , Immunodominant Epitopes , Pneumococcal Infections/immunology , Streptococcus pneumoniae/immunology , T-Lymphocytes, Helper-Inducer/immunology , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cell Proliferation , Coculture Techniques , Feeder Cells , Female , HLA Antigens/immunology , HLA Antigens/metabolism , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Interferon-gamma/metabolism , Interleukins/metabolism , Lymph Nodes/immunology , Lymph Nodes/microbiology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Pneumococcal Infections/metabolism , Pneumococcal Infections/microbiology , Protein Binding , Protein Structure, Secondary , Spleen/immunology , Spleen/microbiology , Streptococcus pneumoniae/metabolism , T-Lymphocytes, Helper-Inducer/microbiology
15.
J Immunol ; 188(11): 5636-43, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22529294

ABSTRACT

Fungal pathogens are a frequent cause of opportunistic infections. They live as commensals in healthy individuals but can cause disease when the immune status of the host is altered. T lymphocytes play a critical role in pathogen control. However, specific Ags determining the activation and function of antifungal T cells remain largely unknown. By using an immunoproteomic approach, we have identified for the first time, to our knowledge, a natural T cell epitope from Candida albicans. Isolation and sequencing of MHC class II-bound ligands from infected dendritic cells revealed a peptide that was recognized by a major population of all Candida-specific Th cells isolated from infected mice. Importantly, human Th cells also responded to stimulation with the peptide in an HLA-dependent manner but without restriction to any particular HLA class II allele. Immunization of mice with the peptide resulted in a population of epitope-specific Th cells that reacted not only with C. albicans but also with other clinically highly relevant species of Candida including the distantly related Candida glabrata. The extent of the reaction to different Candida species correlated with their degree of phylogenetic relationship to C. albicans. Finally, we show that the newly identified peptide acts as an efficient vaccine when used in combination with an adjuvant inducing IL-17A secretion from peptide-specific T cells. Immunized mice were protected from fatal candidiasis. Together, these results uncover a new immune determinant of the host response against Candida ssp. that could be exploited for the development of antifungal vaccines and immunotherapies.


Subject(s)
Candida albicans/immunology , Candidiasis/prevention & control , Epitopes, T-Lymphocyte/immunology , Fungal Vaccines/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Candidiasis/immunology , Cell Line , Fungal Vaccines/administration & dosage , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes, Helper-Inducer/microbiology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
16.
J Immunol ; 186(3): 1531-7, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21178008

ABSTRACT

Retinoic acid-related orphan receptor (ROR)γt(+) TCRαß(+) cells expressing IL-17, termed Th17 cells, are most abundant in the intestinal lamina propria. Symbiotic microbiota are required for the generation of Th17 cells, but the requirement for microbiota-derived Ag is not documented. In this study, we show that normal numbers of Th17 cells develop in the intestine of mice that express a single TCR in the absence of cognate Ag, whereas the microbiota remains essential for their development. However, such mice, or mice monocolonized with the Th17-inducing segmented filamentous bacteria, fail to induce normal numbers of Foxp3(+) RORγt(+) T cells, the regulatory counterpart of IL-17(+)RORγt(+) T cells. These results demonstrate that a complex microbiota and cognate Ag are required to generate a properly regulated set of RORγt(+) T cells and Th17 cells.


Subject(s)
Cell Proliferation , Interleukin-17/biosynthesis , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Nuclear Receptor Subfamily 1, Group F, Member 3/deficiency , Receptors, Antigen, T-Cell/deficiency , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/microbiology , Amino Acid Sequence , Animals , CD4 Lymphocyte Count , Cell Differentiation/genetics , Cell Differentiation/immunology , Female , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Germ-Free Life/genetics , Germ-Free Life/immunology , Gram-Positive Bacterial Infections/immunology , Gram-Positive Bacterial Infections/pathology , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Interleukin-17/genetics , Intestinal Mucosa/pathology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Mice , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes, Helper-Inducer/pathology
17.
J Immunol ; 187(3): 1393-402, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21705623

ABSTRACT

MHC class I-restricted CD8(+) T cells play an important role in protective immunity against mycobacteria. Previously, we showed that p113-121, derived from Mycobacterium leprae protein ML1419c, induced significant IFN-γ production by CD8(+) T cells in 90% of paucibacillary leprosy patients and in 80% of multibacillary patients' contacts, demonstrating induction of M. leprae-specific CD8(+) T cell immunity. In this work, we studied the in vivo role and functional profile of ML1419c p113-121-induced T cells in HLA-A*0201 transgenic mice. Immunization with 9mer or 30mer covering the p113-121 sequence combined with TLR9 agonist CpG induced HLA-A*0201-restricted, M. leprae-specific CD8(+) T cells as visualized by p113-121/HLA-A*0201 tetramers. Most CD8(+) T cells produced IFN-γ, but distinct IFN-γ(+)/TNF-α(+) populations were detected simultaneously with significant secretion of CXCL10/IFN-γ-induced protein 10, CXCL9/MIG, and VEGF. Strikingly, peptide immunization also induced high ML1419c-specific IgG levels, strongly suggesting that peptide-specific CD8(+) T cells provide help to B cells in vivo, as CD4(+) T cells were undetectable. An additional important characteristic of p113-121-specific CD8(+) T cells was their capacity for in vivo killing of p113-121-labeled, HLA-A*0201(+) splenocytes. The cytotoxic function of p113-121/HLA-A*0201-specific CD8(+) T cells extended into direct killing of splenocytes infected with live Mycobacterium smegmatis expressing ML1419c: both 9mer and 30mer induced CD8(+) T cells that reduced the number of ML1419c-expressing mycobacteria by 95%, whereas no reduction occurred using wild-type M. smegmatis. These data, combined with previous observations in Brazilian cohorts, show that ML1419c p113-121 induces potent CD8(+) T cells that provide protective immunity against M. leprae and B cell help for induction of specific IgG, suggesting its potential use in diagnostics and as a subunit (vaccine) for M. leprae infection.


Subject(s)
Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Cytotoxicity Tests, Immunologic , Epitopes, T-Lymphocyte/immunology , HLA-A Antigens/immunology , Mycobacterium leprae/immunology , Peptide Fragments/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/microbiology , Amino Acid Sequence , Animals , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/microbiology , B-Lymphocyte Subsets/pathology , Bacterial Proteins/administration & dosage , Bacterial Vaccines/administration & dosage , Cells, Cultured , Cytotoxicity Tests, Immunologic/methods , Epitopes, T-Lymphocyte/administration & dosage , HLA-A Antigens/biosynthesis , HLA-A Antigens/genetics , HLA-A2 Antigen , Humans , Leprosy/immunology , Leprosy/microbiology , Leprosy/prevention & control , Mice , Mice, Transgenic , Molecular Sequence Data , Mycobacterium leprae/pathogenicity , Peptide Fragments/administration & dosage , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/microbiology , T-Lymphocytes, Helper-Inducer/pathology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
18.
Clin Dev Immunol ; 2013: 503754, 2013.
Article in English | MEDLINE | ID: mdl-23762091

ABSTRACT

Periodontal disease (PD), or periodontitis, is defined as a bacterially induced disease of the tooth-supporting (periodontal) tissues. It is characterized by inflammation and bone loss; therefore understanding how they are linked would help to address the most efficacious therapeutic approach. Bacterial infection is the primary etiology but is not sufficient to induce the disease initiation or progression. Indeed, bacteria-derived factors stimulate a local inflammatory reaction and activation of the innate immune system. The innate response involves the recognition of microbial components by host cells, and this event is mediated by toll-like receptors (TLRs) expressed by resident cells and leukocytes. Activation of these cells leads to the release of proinflammatory cytokines and recruitment of phagocytes and lymphocytes. Activation of T and B cells initiates the adaptive immunity with Th1 Th2 Th17 Treg response and antibodies production respectively. In this inflammatory scenario, cytokines involved in bone regulation and maintenance have considerable relevance because tissue destruction is believed to be the consequence of host inflammatory response to the bacterial challenge. In the present review, we summarize host factors including cell populations, cytokines, and mechanisms involved in the destruction of the supporting tissues of the tooth and discuss treatment perspectives based on this knowledge.


Subject(s)
Alveolar Bone Loss/immunology , B-Lymphocytes/immunology , Bacterial Infections/immunology , Periodontitis/immunology , Phagocytes/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adaptive Immunity , Alveolar Bone Loss/microbiology , Alveolar Bone Loss/pathology , B-Lymphocytes/microbiology , B-Lymphocytes/pathology , Bacterial Infections/microbiology , Bacterial Infections/pathology , Cytokines/genetics , Cytokines/immunology , Gene Expression , Humans , Immunity, Innate , Inflammation , Periodontitis/microbiology , Periodontitis/pathology , Phagocytes/microbiology , Phagocytes/pathology , T-Lymphocytes, Helper-Inducer/microbiology , T-Lymphocytes, Helper-Inducer/pathology , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology
19.
Gastroenterology ; 141(1): 186-96, 196.e1, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21569773

ABSTRACT

BACKGROUND & AIMS: Immunization against the gastric bacterium Helicobacter pylori could prevent many gastric cancers and other disorders. Most vaccination protocols used in preclinical models are not suitable for humans. New adjuvants and a better understanding of the correlates and requirements for vaccine-induced protection are needed to accelerate development of vaccines for H pylori. METHODS: Vaccine-induced protection against H pylori infection and its local and systemic immunological correlates were assessed in animal models, using cholera toxin or CAF01 as adjuvants. The contribution of B cells, T-helper (Th)-cell subsets, and dendritic cells to H pylori-specific protection were analyzed in mice. RESULTS: Parenteral administration of a whole-cell sonicate, combined with the mycobacterial cell-wall-derived adjuvant CAF01, protected against infection with H pylori and required cell-mediated, but not humoral, immunity. The vaccine-induced control of H pylori was accompanied by Th1 and Th17 responses in the gastric mucosa and in the gut-draining mesenteric lymph nodes; both Th subsets were required for protective immunity against H pylori. The numbers of memory CD4+ T cells and neutrophils in gastric tissue were identified as the best correlates of protection. Systemic depletion of dendritic cells or regulatory T cells during challenge infection significantly increased protection by overriding immunological tolerance mechanisms activated by live H pylori. CONCLUSIONS: Parenteral immunization with a Helicobacter vaccine using a novel mycobacterial adjuvant induces protective immunity against H pylori that is mediated by Th1 and Th17 cells. Tolerance mechanisms mediated by dendritic cells and regulatory T cells impair H pylori clearance and must be overcome to improve immunity.


Subject(s)
Adjuvants, Immunologic/pharmacology , Bacterial Vaccines/pharmacology , Cancer Vaccines/pharmacology , Dendritic Cells/drug effects , Helicobacter Infections/prevention & control , Helicobacter pylori/immunology , Stomach Neoplasms/prevention & control , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Administration, Oral , Animals , Antibodies, Bacterial/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/microbiology , Bacterial Vaccines/administration & dosage , Cancer Vaccines/administration & dosage , Cell Wall/immunology , Chemotaxis/drug effects , Cholera Toxin/immunology , Dendritic Cells/immunology , Dendritic Cells/microbiology , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Immune Tolerance/drug effects , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Injections, Intraperitoneal , Injections, Subcutaneous , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mycobacterium/immunology , Stomach/drug effects , Stomach/immunology , Stomach/microbiology , Stomach Neoplasms/immunology , Stomach Neoplasms/microbiology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/microbiology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/microbiology , Time Factors
20.
J Immunol ; 184(4): 2076-85, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20089698

ABSTRACT

The T cell response to Shigella, the causative agent of bacillary dysentery, remains poorly understood. Using a murine model of infection, we report that Shigella flexneri primes predominately IL-17A- and IL-22-producing Th17 cells. Shigella-specific Th1 cells are only significantly induced on secondary infection, whereas specific Th2 and CD8(+) T cells are undetectable. Apart from Th17 cells that are primed in a MHC class II- and IL-6-dependent, but IL12/23p40-independent manner, we identified gammadelta T cells as an additional but minor source of IL-17A. Priming of IL-17A(+) gammadelta T cells is dependent on IL12/23p40, but independent of MHC-class II and IL-6. Th17 cells have emerged as important players in inflammatory, autoimmune, and infectious diseases. Among the yet unresolved questions is their role in long-term immunity to pathogens. In this study, we show that the elicited S. flexneri-specific Th17 pool gives rise to an enhanced recall response up to 12 mo after priming, suggesting the presence of a long-term memory state. The clearance of primary infection is impaired in the absence of T cells, but independently of IL-17A. However, after reinfection, IL-17A produced by S. flexneri-specific Th17 cells becomes important to ultimately restrict bacterial growth. These findings bring new insights into the adaptive immune response to Shigella infection and highlight the importance of pathogen-specific Th17 cell immunity for secondary immune protection.


Subject(s)
Dysentery, Bacillary/immunology , Interleukin-17/biosynthesis , Shigella flexneri/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/microbiology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/microbiology , Animals , Cells, Cultured , Dysentery, Bacillary/microbiology , Dysentery, Bacillary/prevention & control , Immunodominant Epitopes/immunology , Immunologic Memory , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/microbiology , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL