Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
PLoS One ; 12(7): e0180870, 2017.
Article in English | MEDLINE | ID: mdl-28719615

ABSTRACT

While the immune system is essential for the maintenance of the homeostasis, health and survival of humans, aberrant immune responses can lead to chronic inflammatory and autoimmune disorders. Pharmacological modulation of drug targets in the immune system to ameliorate disease also carry a risk of immunosuppression that could lead to adverse outcomes. Therefore, it is important to understand the 'immune fingerprint' of novel therapeutics as they relate to current and, clinically used immunological therapies to better understand their potential therapeutic benefit as well as immunosuppressive ability that might lead to adverse events such as infection risks and cancer. Since the mechanistic investigation of pharmacological modulators in a drug discovery setting is largely compound- and mechanism-centric but not comprehensive in terms of immune system impact, we developed a human tissue based functional assay platform to evaluate the impact of pharmacological modulators on a range of innate and adaptive immune functions. Here, we demonstrate that it is possible to generate a qualitative and quantitative immune system impact of pharmacological modulators, which might help better understand and predict the benefit-risk profiles of these compounds in the treatment of immune disorders.


Subject(s)
Drug Evaluation, Preclinical/methods , Immune System/drug effects , Small Molecule Libraries/pharmacology , Chemokines/biosynthesis , Humans , Immune System/cytology , Immune System/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Phagocytes/drug effects , Phagocytes/immunology , Phagocytes/metabolism , Reactive Oxygen Species/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Toll-Like Receptors/metabolism , Transcriptome/drug effects
2.
J Leukoc Biol ; 100(2): 339-49, 2016 08.
Article in English | MEDLINE | ID: mdl-26908827

ABSTRACT

TLRs facilitate the recognition of pathogens by immune cells and the initiation of the immune response, leading to the production of proinflammatory cytokines and chemokines. Production of proinflammatory mediators by innate immune cells, such as macrophages, is tightly regulated to facilitate pathogen clearance while limiting an adverse impact on host tissue. Exposure of innate immune cells to TLR ligands induces a state of temporary refractoriness to a subsequent exposure of a TLR ligand, a phenomenon referred to as "tolerance." This study sought to evaluate the mechanistic regulation of TLR4 and TLR7/8 ligand-induced tolerance to other TLRs by microRNA-146a. With the use of THP-1 macrophages, as well as human classic and alternative macrophages, we demonstrate that priming with a TLR4 agonist (LPS) or a TLR7/8 agonist (R848) induces homologous and heterologous tolerance to various TLR ligands in macrophages, leading to the impaired production of cytokines and chemokines. We also demonstrate that overexpression of microRNA-146a is sufficient to mimic LPS or R848-induced hyporesponsiveness. Conversely, inhibition of microRNA-146a activity leads to LPS- or R848-induced TLR hyper-responsiveness in TLR signaling tolerance. Furthermore, we demonstrate that microRNA-146a dampens cytokine production following a primary stimulus with MyD88-dependent but not MyD88-independent TLR pathways. Collectively, these data provide comprehensive evidence of the central role of microRNA-146a in TLR signaling tolerance to plasma membrane, as well as endosomal TLR ligands in human macrophages.


Subject(s)
Gene Expression Regulation/drug effects , Immune Tolerance/drug effects , Macrophages/immunology , MicroRNAs/genetics , Myeloid Differentiation Factor 88/metabolism , Toll-Like Receptor 4/agonists , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Cells, Cultured , Cytokines/metabolism , Humans , Imidazoles/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism
3.
J Immunol ; 196(1): 34-8, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26590314

ABSTRACT

IL-1 family members are central mediators of host defense. In this article, we show that the novel IL-1 family member IL-36γ was expressed during experimental colitis and human inflammatory bowel disease. Germ-free mice failed to induce IL-36γ in response to dextran sodium sulfate (DSS)-induced damage, suggesting that gut microbiota are involved in its induction. Surprisingly, IL-36R-deficient (Il1rl2(-/-)) mice exhibited defective recovery following DSS-induced damage and impaired closure of colonic mucosal biopsy wounds, which coincided with impaired neutrophil accumulation in the wound bed. Failure of Il1rl2(-/-) mice to recover from DSS-induced damage was associated with a profound reduction in IL-22 expression, particularly by colonic neutrophils. Defective recovery of Il1rl2(-/-) mice could be rescued by an aryl hydrocarbon receptor agonist, which was sufficient to restore IL-22 expression and promote full recovery from DSS-induced damage. These findings implicate the IL-36/IL-36R axis in the resolution of intestinal mucosal wounds.


Subject(s)
Colitis/immunology , Interleukin-1/biosynthesis , Interleukins/biosynthesis , Receptors, Interleukin/immunology , Wound Healing/immunology , Animals , Colitis/chemically induced , Colitis/microbiology , Colon/immunology , Colon/injuries , Dextran Sulfate , Helicobacter hepaticus/pathogenicity , Humans , Inflammation/immunology , Inflammation/pathology , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/immunology , Neutrophils/immunology , Neutrophils/metabolism , Receptors, Aryl Hydrocarbon/agonists , Receptors, Interleukin/genetics , Wound Healing/genetics , Interleukin-22
4.
J Immunol ; 195(2): 683-94, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26041536

ABSTRACT

Innate immune responses to allergens by airway epithelial cells (AECs) help initiate and propagate the adaptive immune response associated with allergic airway inflammation in asthma. Activation of the transcription factor NF-κB in AECs by allergens or secondary mediators via G protein-coupled receptors (GPCRs) is an important component of this multifaceted inflammatory cascade. Members of the caspase recruitment domain family of proteins display tissue-specific expression and help mediate NF-κB activity in response to numerous stimuli. We have previously shown that caspase recruitment domain-containing membrane-associated guanylate kinase protein (CARMA)3 is specifically expressed in AECs and mediates NF-κB activation in these cells in response to stimulation with the GPCR agonist lysophosphatidic acid. In this study, we demonstrate that reduced levels of CARMA3 in normal human bronchial epithelial cells decreases the production of proasthmatic mediators in response to a panel of asthma-relevant GPCR ligands such as lysophosphatidic acid, adenosine triphosphate, and allergens that activate GPCRs such as Alternaria alternata and house dust mite. We then show that genetically modified mice with CARMA3-deficient AECs have reduced airway eosinophilia and proinflammatory cytokine production in a murine model of allergic airway inflammation. Additionally, we demonstrate that these mice have impaired dendritic cell maturation in the lung and that dendritic cells from mice with CARMA3-deficient AECs have impaired Ag processing. In conclusion, we show that AEC CARMA3 helps mediate allergic airway inflammation, and that CARMA3 is a critical signaling molecule bridging the innate and adaptive immune responses in the lung.


Subject(s)
Asthma/immunology , CARD Signaling Adaptor Proteins/immunology , Dendritic Cells/immunology , Epithelial Cells/immunology , Lung/immunology , Adaptive Immunity , Adenosine Triphosphate/pharmacology , Allergens/immunology , Alternaria/immunology , Animals , Asthma/chemically induced , Asthma/genetics , Asthma/pathology , CARD Signaling Adaptor Proteins/deficiency , CARD Signaling Adaptor Proteins/genetics , Cells, Cultured , Cytokines/biosynthesis , Cytokines/immunology , Dendritic Cells/drug effects , Dendritic Cells/pathology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Gene Expression Regulation , Immunity, Innate , Lung/drug effects , Lung/pathology , Lysophospholipids/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/immunology , Ovalbumin/immunology , Pyroglyphidae/immunology , Signal Transduction
5.
Crit Rev Immunol ; 33(3): 219-43, 2013.
Article in English | MEDLINE | ID: mdl-23756245

ABSTRACT

Caspase recruitment domain-containing membrane-associated guanylate kinase protein-1 (CARMA1), a member of the membrane associated guanylate kinase (MAGUK) family of kinases, is essential for T lymphocyte activation and proliferation via T-cell receptor (TCR) mediated NF-κB activation. Recent studies suggest a broader role for CARMA1 regulating other T-cell functions as well as a role in non-TCR-mediated signaling pathways important for lymphocyte development and functions. In addition, CARMA1 has been shown to be an important component in the pathogenesis of several human diseases. Thus, comprehensively defining its mechanisms of action and regulation could reveal novel therapeutic targets for T-cell-mediated diseases and lymphoproliferative disorders.


Subject(s)
CARD Signaling Adaptor Proteins/immunology , Guanylate Cyclase/immunology , T-Lymphocytes/immunology , Animals , CARD Signaling Adaptor Proteins/chemistry , Guanylate Cyclase/chemistry , Humans , Receptors, Antigen, T-Cell/immunology , Signal Transduction
6.
PLoS One ; 7(9): e45784, 2012.
Article in English | MEDLINE | ID: mdl-23029241

ABSTRACT

Interleukin (IL-) 36 cytokines (previously designated as novel IL-1 family member cytokines; IL-1F5- IL-1F10) constitute a novel cluster of cytokines structurally and functionally similar to members of the IL-1 cytokine cluster. The effects of IL-36 cytokines in inflammatory lung disorders remains poorly understood. The current study sought to investigate the effects of IL-36α (IL-1F6) and test the hypothesis that IL-36α acts as a pro-inflammatory cytokine in the lung in vivo. Intratracheal instillation of recombinant mouse IL-36α induced neutrophil influx in the lungs of wild-type C57BL/6 mice and IL-1αß(-/-) mice in vivo. IL-36α induced neutrophil influx was also associated with increased mRNA expression of neutrophil-specific chemokines CXCL1 and CXCL2 in the lungs of C57BL/6 and IL-1αß(-/-) mice in vivo. In addition, intratracheal instillation of IL-36α enhanced mRNA expression of its receptor IL-36R in the lungs of C57BL/6 as well as IL-1αß(-/-) mice in vivo. Furthermore, in vitro incubation of CD11c(+) cells with IL-36α resulted in the generation of neutrophil-specific chemokines CXCL1, CXCL2 as well as TNFα. IL-36α increased the expression of the co-stimulatory molecule CD40 and enhanced the ability of CD11c(+) cells to induce CD4(+) T cell proliferation in vitro. Furthermore, stimulation with IL-36α activated NF-κB in a mouse macrophage cell line. These results demonstrate that IL-36α acts as a pro-inflammatory cytokine in the lung without the contribution of IL-1α and IL-1ß. The current study describes the pro-inflammatory effects of IL-36α in the lung, demonstrates the functional redundancy of IL-36α with other agonist cytokines in the IL-1 and IL-36 cytokine cluster, and suggests that therapeutic targeting of IL-36 cytokines could be beneficial in inflammatory lung diseases.


Subject(s)
Inflammation Mediators/physiology , Interleukin-1/physiology , Pneumonia/metabolism , Animals , CD11 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/physiology , Cell Proliferation , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Cytokines/physiology , Gene Expression Regulation , Inflammation Mediators/metabolism , Interleukin-1/genetics , Interleukin-1/metabolism , Interleukin-1alpha/deficiency , Interleukin-1alpha/genetics , Interleukin-1beta/deficiency , Interleukin-1beta/genetics , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Neutrophil Infiltration , Pneumonia/immunology , Pneumonia/pathology , Spleen/immunology , Spleen/pathology
7.
J Immunol ; 187(12): 6197-207, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22075698

ABSTRACT

CARMA1 is a lymphocyte-specific scaffold protein necessary for T cell activation. Deletion of CARMA1 prevents the development of allergic airway inflammation in a mouse model of asthma due to a defect in naive T cell activation. However, it is unknown if CARMA1 is important for effector and memory T cell responses after the initial establishment of inflammation, findings that would be more relevant to asthma therapies targeted to CARMA1. In the current study, we sought to elucidate the role of CARMA1 in T cells that have been previously activated. Using mice in which floxed CARMA1 exons can be selectively deleted in T cells by OX40-driven Cre recombinase (OX40(+/Cre)CARMA1(F/F)), we report that CD4(+) T cells from these mice have impaired T cell reactivation responses and NF-κB signaling in vitro. Furthermore, in an in vivo recall model of allergic airway inflammation that is dependent on memory T cell function, OX40(+/Cre)CARMA1(F/F) mice have attenuated eosinophilic airway inflammation, T cell activation, and Th2 cytokine production. Using MHC class II tetramers, we demonstrate that the development and maintenance of Ag-specific memory T cells is not affected in OX40(+/Cre)CARMA1(F/F) mice. In addition, adoptive transfer of Th2-polarized OX40(+/Cre)CARMA1(F/F) Ag-specific CD4(+) T cells into wild-type mice induces markedly less airway inflammation in response to Ag challenge than transfer of wild-type Th2 cells. These data demonstrate a novel role for CARMA1 in effector and memory T cell responses and suggest that therapeutic strategies targeting CARMA1 could help treat chronic inflammatory disorders such as asthma.


Subject(s)
Asthma/immunology , CARD Signaling Adaptor Proteins/physiology , Th2 Cells/immunology , Acute Disease , Adoptive Transfer , Animals , Asthma/genetics , Asthma/pathology , CARD Signaling Adaptor Proteins/deficiency , CARD Signaling Adaptor Proteins/genetics , Cell Polarity/genetics , Cell Polarity/immunology , Cells, Cultured , Chronic Disease , Disease Models, Animal , Immunologic Memory/genetics , Immunologic Memory/immunology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/physiology , Integrases/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/physiology , Receptors, OX40/biosynthesis , Receptors, OX40/genetics , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/prevention & control , Th2 Cells/cytology , Th2 Cells/transplantation
8.
Am J Respir Cell Mol Biol ; 44(2): 134-45, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20299540

ABSTRACT

Interleukin-1 (IL-1) is a proinflammatory cytokine that signals through the Type I IL-1 receptor (IL-1RI). Novel IL-1-like cytokines were recently identified. Their functions in lung disease remain unclear. Interleukin-1 family member-9 (IL-1F9) is one such IL-1-like cytokine, expressed in the lungs of humans and mice. IL-1F9 signals through IL-1 receptor-related protein 2 (IL-1Rrp2/IL-1RL2), which is distinct from IL-1RI. We sought to determine if IL-1F9 acts as a proinflammatory cytokine in lung disease. IL-1F9 protein was increased in lung homogenates of house dust mite-challenged A/J mice compared with controls, and expression was seen in airway epithelial cells. The intratracheal administration of recombinant mouse IL-1F9 increased airway hyperresponsiveness and induced neutrophil influx and mucus production, but not eosinophilic infiltration in the lungs of mice. In addition, IL-1α protein levels in bronchoalveolar lavage fluid, chemokines, and chemokine-receptor mRNA expression in the lungs were increased after the instillation of intratracheal IL-1F9. Consistent with these changes, NF-κB transcription factor activity was increased in the lungs of mice challenged with IL-1F9 and in a macrophage cell line treated with IL-1F9. These data suggest that IL-1F9 is upregulated during inflammation, and acts as a proinflammatory cytokine in the lungs.


Subject(s)
Chemokines/biosynthesis , Interleukin-1/pharmacology , Lung/drug effects , Lung/immunology , Neutrophil Infiltration/drug effects , Allergens/administration & dosage , Animals , Base Sequence , Bronchial Hyperreactivity/etiology , Bronchial Hyperreactivity/immunology , Cell Line , Chemokines/genetics , DNA Primers/genetics , Interleukin-1/genetics , Interleukin-1/immunology , Interleukin-1/metabolism , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred A , Mice, Inbred C3H , Mucus/metabolism , NF-kappa B/metabolism , Neutrophil Infiltration/immunology , Pyroglyphidae/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
9.
J Immunol ; 182(3): 1560-7, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19155504

ABSTRACT

Mice lacking surfactant protein A (SP-A) are susceptible to bacterial infection associated with an excessive inflammatory response in the lung. To determine mechanisms by which SP-A is antiinflammatory in the lung during bacterial infection, SP-A regulation of secretory leukoprotease inhibitor (SLPI), an inhibitor of serine proteases, was assessed. SLPI protein expression and antineutrophil elastase activity were reduced in bronchoalveolar fluid of SP-A(-/-) compared with SP-A(+/+) mice. Intratracheal administration of SP-A to SP-A(-/-) mice enhanced SLPI protein expression and antineutrophil elastase activity in the lung. SLPI mRNA was similar in whole lung and alveolar type II cells; however, it was significantly reduced in alveolar macrophages from SP-A(-/-) compared with SP-A(+/+) mice. In vitro, SP-A enhanced SLPI production by macrophage THP-1 cells but not respiratory epithelial A549 cells. SP-A inhibited LPS induced IkappaB-alpha degradation in THP-1 cells, which was partially reversed with knockdown of SLPI. Matrix metalloproteinase (MMP)-12 cleaved SLPI and incubation with SP-A reduced MMP-12-mediated SLPI cleavage. The collagen-like region of SP-A conferred protection of SLPI against MMP mediated cleavage. SP-A plays an important role in the lung during bacterial infection regulating protease and antiprotease activity.


Subject(s)
Matrix Metalloproteinase 12/physiology , Matrix Metalloproteinase Inhibitors , Pulmonary Surfactant-Associated Protein A/physiology , Secretory Leukocyte Peptidase Inhibitor/biosynthesis , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/microbiology , Cell Line, Tumor , Female , Haemophilus Infections/enzymology , Haemophilus Infections/immunology , Haemophilus Infections/metabolism , Haemophilus influenzae/immunology , Humans , Hydrolysis , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Male , Matrix Metalloproteinase 12/biosynthesis , Mice , Mice, Knockout , Pneumonia/enzymology , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/pathology , Pulmonary Surfactant-Associated Protein A/deficiency , Pulmonary Surfactant-Associated Protein A/genetics , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , Secretory Leukocyte Peptidase Inhibitor/antagonists & inhibitors , Secretory Leukocyte Peptidase Inhibitor/genetics , Secretory Leukocyte Peptidase Inhibitor/metabolism , Secretory Leukocyte Peptidase Inhibitor/physiology , Up-Regulation/immunology , alpha 1-Antitrypsin/metabolism
10.
DNA Seq ; 18(3): 190-5, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17454003

ABSTRACT

Asthma is a common respiratory disease that is driven by both genetic and environmental factors. Identification of the genes underlying asthma will provide insight into the mechanisms and treatment of this important disease. Previous studies in this laboratory identified two distinct quantitative trait loci for the asthma-related parameter, allergen-induced airway hyperresponsiveness, in a murine model by means of a genome-wide linkage analysis. The present study focuses and refines the map location of these two loci. Additionally, we explore prostaglandin-endoperoxide synthase-1 (Ptgs1) and mannose receptor C-type 1 (Mrc1) genes as two new positional candidate genes for allergen-induced airway hyperresponsiveness through comparative sequence analysis and mRNA expression studies of mouse strains with genetically mediated airway responsiveness.


Subject(s)
Asthma/genetics , Cyclooxygenase 1/genetics , Genes/genetics , Membrane Proteins/genetics , Quantitative Trait Loci , Animals , Chromosome Mapping , Chromosomes, Mammalian , Humans , Mice , Mice, Inbred C3H , Mice, Inbred Strains
11.
Immunogenetics ; 58(10): 851-5, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17021861

ABSTRACT

Interleukin-1 receptor antagonist (IL-1ra) is an inhibitor of the proinflammatory IL-1. The IL-1ra gene (Il1rn) maps near the allergen-induced bronchial hyper-responsiveness-1 locus, Abhr1, which we previously mapped to murine chromosome 2 using A/J (asthma susceptible) and C3H/HeJ (asthma resistant) mice. We evaluated the role of Il1rn in our mouse model by comparing its genomic sequence between A/J and C3H/HeJ mice as well as assessing strain-specific RNA and protein production in response to allergen. We identified no functional sequence variations in the Il1rn gene between A/J and C3H/HeJ mice. Il1rn mRNA and protein were induced by ovalbumin (OVA) exposure in both strains, but to a greater extent in A/J mice at the earlier time points. We examined other IL-1 family members (Il1a, Il1b, Il1f9, and Il1r2) and found OVA-induced expression increases at 6 h, yet only Il1b and Il1f9 had strain-specific differences. Of these, only Il1f9 is located within Abhr1, and we found several non-coding polymorphisms in the Il1f9 gene between A/J and C3H/HeJ mice. Our results exclude Il1rn as the gene for Abhr1 and indicate that Il1f9 warrants further investigation based on genetic and expression differences observed in our mouse model of allergic asthma.


Subject(s)
Asthma/genetics , Asthma/immunology , Genetic Predisposition to Disease , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin 1 Receptor Antagonist Protein/immunology , Animals , Asthma/metabolism , Asthma/pathology , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1/immunology , Interleukin-1/metabolism , Lung/chemistry , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred Strains , Ovalbumin/immunology , Phenotype , RNA/isolation & purification , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...