Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
ALTEX ; 40(2): 248­270, 2023.
Article in English | MEDLINE | ID: mdl-36129398

ABSTRACT

A structurally diverse set of 147 per- and polyfluoroalkyl substances (PFAS) was screened in a panel of 12 human primary cell systems by measuring 148 biomarkers relevant to (patho)physiological pathways to inform hypotheses about potential mechanistic effects of data-poor PFAS in human model systems. This analysis focused on immunosuppressive activity, which was previously reported as an in vivo effect of perfluorooctanoic acid (PFOA) and perfluorooctanesulfonic acid (PFOS), by comparing PFAS responses to four pharmacological immunosuppressants. The PFOS response profile had little correlation with reference immunosuppressants, suggesting in vivo activity does not occur by similar mechanisms. The PFOA response profile did share features with the profile of dexamethasone, although some distinct features were lacking. Other PFAS, including 2,2,3,3-tetrafluoropropyl acrylate, demonstrated more similarity to the reference immunosuppressants but with additional activities not found in the reference immunosuppressive drugs. Correlation of PFAS profiles with a database of environmental chemical responses and pharmacological probes identified potential mechanisms of bioactivity for some PFAS, including responses similar to ubiquitin ligase inhibitors, deubiquitylating enzyme (DUB) inhibitors, and thioredoxin reductase inhibitors. Approximately 21% of the 147 PFAS with confirmed sample quality were bioactive at nominal testing concentrations in the 1-60 micromolar range in these human primary cell systems. These data provide new hypotheses for mechanisms of action for a subset of PFAS and may further aid in development of a PFAS categorization strategy useful in safety assessment.


Subject(s)
Alkanesulfonic Acids , Environmental Pollutants , Fluorocarbons , Humans , Alkanesulfonic Acids/toxicity , Caprylates , Fluorocarbons/toxicity , Fluorocarbons/analysis
2.
PLoS One ; 14(9): e0222944, 2019.
Article in English | MEDLINE | ID: mdl-31560729

ABSTRACT

Janus kinase-signal transducers and activators of transcription (JAK-STAT) signaling is critical to multiple cellular processes, including survival, differentiation, and proliferation. JAK-STAT signaling dysregulation has been noted in inflammatory disorders, and aberrant JAK2 pathway activation has been implicated in myelofibrosis and polycythemia vera. Moreover, 4 therapeutic JAK2 inhibitors (ruxolitinib, fedratinib, momelotinib, and pacritinib) have either been approved or are in advanced clinical development for myelofibrosis. Although all inhibit JAK2, reports indicate that they also inhibit other kinases. Profiling based solely on in vitro potencies is insufficient to predict the observed clinical effects. To provide further translational insights into clinical outcomes, we compared phenotypic biomarker profiles of ruxolitinib, fedratinib, momelotinib, and pacritinib in the BioMAP® Diversity PLUS panel of 12 human primary cell systems designed to recapitulate key aspects of tissue and disease states. Biomarker activity profiles that represent mechanistic signatures for each agent were compared with each other and a database of reference benchmark profiles. At clinically relevant concentrations, these agents had distinct biomarker impacts indicating diverse mechanistic signatures, suggesting divergent clinical effects for each agent. They disparately modulated inflammatory cytokine production and immune function. At clinically relevant concentrations, ruxolitinib had the broadest scope of activities across all 12 cellular systems, whereas pacritinib was more specific for the BT system (modelling T cell-dependent B cell activation) and exhibited the strongest inhibition of sIL-17A, sIL-2, and sIL-6. All 4 agents were antiproliferative to B cells, but ruxolitinib and momelotinib were also antiproliferative to T cells. These differential activities likely reflect distinct secondary pharmacology for these agents known primarily as JAK2 inhibitors. The phenotypic analysis reported herein represents key data on distinct modes-of-action that may provide insights on clinical outcomes reported for these agents. Such translational findings may also inform the development of next-generation molecules with improved efficacy and safety.


Subject(s)
Janus Kinase 2/antagonists & inhibitors , Primary Myelofibrosis/drug therapy , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Benzamides/pharmacology , Bridged-Ring Compounds/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Healthy Volunteers , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Janus Kinase 2/immunology , Janus Kinase 2/metabolism , Nitriles , Primary Cell Culture , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Pyrrolidines/pharmacology , Sulfonamides/pharmacology , Toxicity Tests
3.
Immunology ; 154(3): 500-509, 2018 07.
Article in English | MEDLINE | ID: mdl-29377102

ABSTRACT

Cholera toxin (CT) is a bacterial component that increases intracellular cAMP levels in host cells and suppresses T-cell activation. Recently, CT was reported to induce T helper type 17-skewing dendritic cells and activate interleukin-17A (IL-17A) production in CD4+ T cells through a cAMP-dependent pathway. However, the underlying mechanism by which cAMP regulates IL-17A production in T cells is not completely defined. In this study, we took advantage of a small molecule protein kinase A (PKA) inhibitor (H89) and different cAMP analogues: a PKA-specific activator (N6-benzoyl-adenosine-cAMP), an exchange protein activated by cAMP-specific activator (Rp-8-chlorophenylthio-2'-O-methyl cAMP), and a PKA inhibitor (Rp-8-bromo-cAMP), to elucidate the signalling cascade of cAMP in IL-17A regulation in T cells. We found that CT induced IL-17A production and IL-17A promoter activity in activated CD4+ T cells through a cAMP/PKA pathway. Moreover, this regulation was via cAMP-response element binding protein (CREB) -mediated transcriptional activation by using the transfection of an IL-17A promoter-luciferase reporter construct and CREB small interfering RNA in Jurkat cells. Also, we showed that CREB bound to the CRE motif located at -183 of the IL-17A promoter in vitro. Most interestingly, not only in CD4+ T cells, CT also enhanced cAMP/PKA-dependent IL-17A production and CREB phosphorylation in CD8+ T cells. In conclusion, our data suggest that CT induces an IL-17A-dominated immune microenvironment through the cAMP/PKA/CREB signalling pathway. Our study also highlights the potentials of CT and cAMP in modulating T helper type 17 responses in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cholera Toxin/immunology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Interleukin-17/biosynthesis , Interleukin-17/genetics , Binding Sites , Cell Line , Gene Expression Regulation , Humans , Promoter Regions, Genetic , RNA Interference , Signal Transduction , Transcriptional Activation
4.
Cell Chem Biol ; 24(7): 858-869.e5, 2017 Jul 20.
Article in English | MEDLINE | ID: mdl-28669525

ABSTRACT

Cutaneous reactions represent one of the most common adverse drug effects observed in clinical trials leading to substantial compound attrition. Three negative allosteric modulators (NAMs) of metabotropic glutamate receptors (mGluRs), which represent an important target for neurological diseases, developed by Pfizer, were recently failed in preclinical development due to delayed type IV skin hypersensitivity observed in non-human primates (NHPs). Here we employed large-scale phenotypic profiling in standardized panels of human primary cell/co-culture systems to characterize the skin toxicity mechanism(s) of mGluR5 NAMs from two different series. Investigation of a database of chemicals tested in these systems and transcriptional profiling suggested that the mechanism of toxicity may involve modulation of nuclear receptor targets RAR/RXR, and/or VDR with AhR antagonism. The studies reported here demonstrate how phenotypic profiling of preclinical drug candidates using human primary cells can provide insights into the mechanisms of toxicity and inform early drug discovery and development campaigns.


Subject(s)
Fibroblasts/drug effects , Receptor, Metabotropic Glutamate 5/metabolism , Skin Diseases/chemically induced , Allosteric Regulation , Cells, Cultured , Databases, Chemical , Dinoprostone/metabolism , Down-Regulation/drug effects , Fibroblasts/cytology , Fibroblasts/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Interleukin-2/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/toxicity , Protein Binding , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Receptor, Metabotropic Glutamate 5/chemistry , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Calcitriol/agonists , Receptors, Calcitriol/metabolism , Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/metabolism , Skin Diseases/metabolism , Skin Diseases/pathology , Small Molecule Libraries/chemistry , Small Molecule Libraries/toxicity , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects
5.
PLoS One ; 11(10): e0163323, 2016.
Article in English | MEDLINE | ID: mdl-27723765

ABSTRACT

In the context of the human airway, interleukin-17A (IL-17A) signaling is associated with severe inflammation, as well as protection against pathogenic infection, particularly at mucosal surfaces such as the airway. The intracellular molecule Act1 has been demonstrated to be an essential mediator of IL-17A signaling. In the cytoplasm, it serves as an adaptor protein, binding to both the intracellular domain of the IL-17 receptor as well as members of the canonical nuclear factor kappa B (NF-κB) pathway. It also has enzymatic activity, and serves as an E3 ubiquitin ligase. In the context of airway epithelial cells, we demonstrate for the first time that Act1 is also present in the nucleus, especially after IL-17A stimulation. Ectopic Act1 expression can also increase the nuclear localization of Act1. Act1 can up-regulate the expression and promoter activity of a subset of IL-17A target genes in the absence of IL-17A signaling in a manner that is dependent on its N- and C-terminal domains, but is NF-κB independent. Finally, we show that nuclear Act1 can bind to both distal and proximal promoter regions of DEFB4, one of the IL-17A responsive genes. This transcriptional regulatory activity represents a novel function for Act1. Taken together, this is the first report to describe a non-adaptor function of Act1 by directly binding to the promoter region of IL-17A responsive genes and directly regulate their transcription.


Subject(s)
Response Elements/physiology , Signal Transduction/physiology , Transcription, Genetic/physiology , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Up-Regulation/physiology , A549 Cells , Adaptor Proteins, Signal Transducing , Humans , Interleukin-17/biosynthesis , Interleukin-17/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Domains , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , beta-Defensins/biosynthesis , beta-Defensins/genetics
6.
Cancer Res ; 75(23): 5106-5119, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26552700

ABSTRACT

The histone acetyltransferases CBP/p300 are involved in recurrent leukemia-associated chromosomal translocations and are key regulators of cell growth. Therefore, efforts to generate inhibitors of CBP/p300 are of clinical value. We developed a specific and potent acetyl-lysine competitive protein-protein interaction inhibitor, I-CBP112, that targets the CBP/p300 bromodomains. Exposure of human and mouse leukemic cell lines to I-CBP112 resulted in substantially impaired colony formation and induced cellular differentiation without significant cytotoxicity. I-CBP112 significantly reduced the leukemia-initiating potential of MLL-AF9(+) acute myeloid leukemia cells in a dose-dependent manner in vitro and in vivo. Interestingly, I-CBP112 increased the cytotoxic activity of BET bromodomain inhibitor JQ1 as well as doxorubicin. Collectively, we report the development and preclinical evaluation of a novel, potent inhibitor targeting CBP/p300 bromodomains that impairs aberrant self-renewal of leukemic cells. The synergistic effects of I-CBP112 and current standard therapy (doxorubicin) as well as emerging treatment strategies (BET inhibition) provide new opportunities for combinatorial treatment of leukemia and potentially other cancers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Enzyme Inhibitors/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Oxazepines/pharmacology , p300-CBP Transcription Factors/antagonists & inhibitors , Amino Acid Sequence , Animals , Cell Line, Tumor , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Synergism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Humans , Leukemia, Myeloid, Acute/enzymology , Mice , Models, Molecular , Molecular Sequence Data , Oxazepines/administration & dosage , Oxazepines/chemistry , Protein Structure, Tertiary , Xenograft Model Antitumor Assays , p300-CBP Transcription Factors/chemistry
7.
Clin Dev Immunol ; 2013: 267971, 2013.
Article in English | MEDLINE | ID: mdl-23956759

ABSTRACT

The significance of Th17 cells and interleukin- (IL-)17A signaling in host defense and disease development has been demonstrated in various infection and autoimmune models. Numerous studies have indicated that Th17 cells and its signature cytokine IL-17A are critical to the airway's immune response against various bacteria and fungal infection. Cytokines such as IL-23, which are involved in Th17 differentiation, play a critical role in controlling Klebsiella pneumonia (K. pneumonia) infection. IL-17A acts on nonimmune cells in infected tissues to strengthen innate immunity by inducing the expression of antimicrobial proteins, cytokines, and chemokines. Mice deficient in IL-17 receptor (IL-17R) expression are susceptible to infection by various pathogens. In this review, we summarize the recent advances in unraveling the mechanism behind Th17 cell differentiation, IL-17A/IL-17R signaling, and also the importance of IL-17A in pulmonary infection.


Subject(s)
Interleukin-17/metabolism , Pneumonia/immunology , Pneumonia/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , Cell Differentiation , Gene Expression Regulation , Humans , Immunity, Innate , Pneumonia/genetics , Pneumonia/microbiology , Receptors, Interleukin-17/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Signal Transduction , Th17 Cells/cytology
8.
Am J Respir Cell Mol Biol ; 45(2): 270-6, 2011 Aug.
Article in English | MEDLINE | ID: mdl-20971882

ABSTRACT

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a potent environmental toxicant. Epidemiological studies have associated TCDD exposure with the development of chronic obstructive pulmonary disease, which is manifested by mucous/goblet cell hyperplasia. The purpose of this research was to elucidate the pathway/mechanisms that lead to TCDD-induced gene expression in both primary normal human bronchial epithelial cells and an immortalized cell line, HBE1, under air-liquid interface conditions. TCDD exposure induced a time-dependent elevation of MUC5AC mRNA and protein synthesis, and cytochrome p450 1A1 (CYP1A1) expression in these cells. Treatment with an aryl hydrocarbon receptor antagonist had no effect on TCDD-induced MUC5AC expression, but significantly suppressed CYP1A1 induction. However, treatments with inhibitors of signaling pathways and the expression of dominant negative mutants of epidermal growth factor receptor (EGFR), extracellular signal-regulated kinase (ERK) and p38, but not the inhibition of c-Jun N-terminal kinase pathway, abrogated MUC5AC induction, but not that of CYP1A1. These effects also occurred at the MUC5AC promoter-reporter level using the chimeric construct for a transient transfection study. Western blot analysis confirmed the phosphorylation of activated EGFR, ERK, and p38 signaling molecules, but not the c-Jun N-terminal kinase, in cells after TCDD exposure. Specificity protein 1 (Sp1) phosphorylation also occurred in cells after TCDD exposure. Both MUC5AC expression and the promoter activity were inhibited by mithramycin A, an inhibitor specific to Sp1-based transcription. These results lead to the conclusion that TCDD induced MUC5AC expression through a noncanonical aryl hydrocarbon receptor-independent, EGFR/ERK/p38-mediated signaling pathway-mediated/Sp1-based transcriptional mechanism.


Subject(s)
Biomarkers, Tumor/metabolism , Cytochrome P-450 CYP1A1/genetics , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Mucin 5AC/genetics , Polychlorinated Dibenzodioxins/pharmacology , Receptors, Immunologic/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Biomarkers, Tumor/genetics , Blotting, Western , Bronchi/cytology , Bronchi/drug effects , Bronchi/metabolism , Cells, Cultured , Cytochrome P-450 CYP1A1/metabolism , Environmental Pollutants/pharmacology , Enzyme Inhibitors/pharmacology , ErbB Receptors/genetics , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Humans , Mucin 5AC/metabolism , RNA, Messenger/genetics , Receptors, Immunologic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Transcription, Genetic/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
9.
Am J Respir Cell Mol Biol ; 45(2): 246-52, 2011 Aug.
Article in English | MEDLINE | ID: mdl-20935193

ABSTRACT

A major pathological feature of chronic airway diseases is the elevated expression of gel-forming mucins. NF-κB activation in airway epithelial cells has been shown to play a proinflammatory role in chronic airway diseases; however, the specific role of NF-κB in mucin gene expression has not been characterized. In this study, we show that the proinflammatory cytokines, IL-1ß and IL-17A, both of which use the NF-κB pathway, are potent inducers of MUC5B mRNA expression in both well differentiated primary normal human bronchial epithelial cells and the human bronchial epithelial cell line, HBE1. MUC5B induction by these cytokines was both time- and dose-dependent, and was attenuated by the small molecule inhibitor, NF-κB inhibitor III, as well as p65 small interfering RNA, suggesting that the regulation of MUC5B expression by these cytokines is via an NF-κB-based transcriptional mechanism. Deletion analysis of the MUC5B promoter demonstrated that IL-1ß- and IL-17A-induced promoter activity resides within the -4.17-kb to -2.56-kb region relative to the transcriptional start site. This region contains three putative κB-binding sites (NF-κB-1, -3,786/-3,774; NF-κB-2, -3,173/-3,161; and NF-κB-3, -2,921/-2,909). Chromatin immunoprecipitation analysis confirmed enhanced binding of the p50 NF-κB subunit to the NF-κB-3 site after cytokine stimulation. We conclude that an NF-κB-based transcriptional mechanism is involved in MUC5B regulation by IL-1ß and IL-17A in airway epithelium. This is the first demonstration of the participation of NF-κB and its specific binding site in cytokine-mediated airway MUC5B expression.


Subject(s)
Bronchi/metabolism , Gene Expression Regulation , Interleukin-17/metabolism , Interleukin-1beta/metabolism , Mucin-5B/genetics , Mucin-5B/metabolism , NF-kappa B/metabolism , Blotting, Western , Bronchi/cytology , Cells, Cultured , Chromatin Immunoprecipitation , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/metabolism , Humans , Interleukin-17/genetics , Interleukin-1beta/genetics , Mucin-5B/antagonists & inhibitors , NF-kappa B/genetics , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Respiratory System/cytology , Respiratory System/metabolism , Reverse Transcriptase Polymerase Chain Reaction
10.
Int J Cell Biol ; 2010: 529376, 2010.
Article in English | MEDLINE | ID: mdl-21274427

ABSTRACT

We present evidence of a link between interferonß-1b (IFN-ß) and G-protein signaling by demonstrating that IFN-ß can induce the expression of the negative regulator of G-protein signaling 1 (RGS1). RGS1 reduces G-protein activation and immune cell migration by interacting with heterotrimeric G-proteins and enhancing their intrinsic GTPase activity. In this study, IFN-ß treatment resulted in the induction of RGS1 in peripheral blood mononuclear cells (PBMCs), monocytes, T cells, and B cells. Induction of RGS1 by IFN-ß was concentration dependent and observed at both the RNA and protein level. Other members of the RGS family were not induced by IFN-ß, and induction of RGS1 required the activation of the IFN receptor. In addition, RGS1 induction was observed in PBMCs obtained from IFN-ß-treated multiple sclerosis patients suggesting a possible, as yet unexplored, involvement of G-protein regulation in disease treatment. The upregulation of RGS1 by IFN-ß has not been previously reported.

11.
Inflamm Bowel Dis ; 16(3): 428-41, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19639560

ABSTRACT

BACKGROUND: Granulocyte-macrophage colony-stimulating factor (GM-CSF) therapy is effective in treating some Crohn's disease (CD) patients and protects mice from colitis induced by dextran sulfate sodium (DSS) administration. However, its mechanisms of action remain elusive. We hypothesized that GM-CSF affects intestinal mucosal repair. METHODS: DSS colitic mice were treated with daily pegylated GM-CSF or saline and clinical, histological, and inflammatory parameters were kinetically evaluated. Further, the role of bone marrow-derived cells in the impact of GM-CSF therapy on DSS colitis was addressed using cell transfers. RESULTS: GM-CSF therapy reduced clinical signs of colitis and the release of inflammatory mediators. GM-CSF therapy improved mucosal repair, with faster ulcer reepithelialization, accelerated hyperproliferative response of epithelial cells in ulcer-adjacent crypts, and lower colonoscopic ulceration scores in GM-CSF-administered mice relative to untreated mice. We observed that GM-CSF-induced promotion of mucosal repair is timely associated with a reduction in neutrophil numbers and increased accumulation of CD11b(+) monocytic cells in colon tissues. Importantly, transfer of splenic GM-CSF-induced CD11b(+) myeloid cells into DSS-exposed mice improved colitis, and lethally irradiated GM-CSF receptor-deficient mice reconstituted with wildtype bone marrow cells were protected from DSS-induced colitis upon GM-CSF therapy. Lastly, GM-CSF-induced CD11b(+) myeloid cells were shown to promote in vitro wound repair. CONCLUSIONS: Our study shows that GM-CSF-dependent stimulation of bone marrow-derived cells during DSS-induced colitis accelerates colonic tissue repair. These data provide a putative mechanism for the observed beneficial effects of GM-CSF therapy in Crohn's disease.


Subject(s)
Bone Marrow Cells/drug effects , Colitis/drug therapy , Colitis/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Wound Healing/drug effects , Acute Disease , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Colitis/chemically induced , Colon/drug effects , Colon/pathology , Colon/physiology , Crohn Disease/drug therapy , Crohn Disease/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Female , Mice , Mice, Inbred BALB C , Mice, Knockout , Mucous Membrane/drug effects , Mucous Membrane/pathology , Regeneration/drug effects , Signal Transduction/drug effects , Specific Pathogen-Free Organisms , Wound Healing/physiology
12.
J Immunol ; 183(10): 6236-43, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19841186

ABSTRACT

Mucin over-production is one of the hallmarks of chronic airway diseases such as chronic obstructive pulmonary disease, asthma, and cystic fibrosis. NF-kappaB activation in airway epithelial cells has been shown to play a positive inflammatory role in chronic airway diseases; however, the role of NF-kappaB in mucin gene expression is unresolved. In this study, we have shown that the proinflammatory cytokines, IL-1beta and IL-17A, both of which utilize the NF-kappaB pathway, are potent inducers of mucin (MUC)5AC mRNA and protein synthesis by both well-differentiated primary normal human bronchial epithelial cells and the human bronchial epithelial cell line, HBE1. MUC5AC induction by these cytokines was both time- and dose-dependent and occurred at the level of promoter activation, as measured by a reporter gene assay. These effects were attenuated by the small molecule inhibitor NF-kappaB inhibitor III, as well as p65 small-interfering RNA, suggesting that the regulation of MUC5AC expression by these cytokines is via an NF-kappaB-based transcriptional mechanism. Further investigation of the promoter region identified a putative NF-kappaB binding site at position-3594/-3582 in the promoter of MUC5AC as critical for the regulation of MUC5AC expression by both IL-1beta and IL-17A. Chromatin immunoprecipitation analysis confirmed enhanced binding of the NF-kappaB subunit p50 to this region following cytokine stimulation. We conclude that an NF-kappaB-based transcriptional mechanism is involved in MUC5AC regulation by IL-1beta and IL-17A in the airway epithelium. This is the first demonstration of the participation of NF-kappaB and its specific binding site in cytokine-mediated airway MUC5AC expression.


Subject(s)
Bronchi/immunology , Mucin 5AC/metabolism , NF-kappa B/metabolism , Respiratory Mucosa/immunology , Transcription Factor RelA/metabolism , Bronchi/drug effects , Cell Line , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Humans , Interleukin-17/pharmacology , Interleukin-1beta/pharmacology , Mucin 5AC/agonists , Mucin 5AC/genetics , Mucin 5AC/immunology , NF-kappa B/antagonists & inhibitors , NF-kappa B/immunology , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , RNA, Messenger/immunology , RNA, Messenger/metabolism , RNA, Small Interfering/immunology , RNA, Small Interfering/metabolism , Recombinant Proteins/pharmacology , Respiratory Mucosa/drug effects , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , Transfection
13.
J Interferon Cytokine Res ; 28(5): 317-31, 2008 May.
Article in English | MEDLINE | ID: mdl-18547162

ABSTRACT

Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS). Interferon-beta (IFN-beta) therapy for MS is hypothesized to cause short-term and long-term changes in gene expression that shift the inflammation from Th1 to Th2. In vivo gene induction to define kinetics of response to IFN-beta therapy in a large cohort of MS patients is described. Differential gene expression in peripheral blood mononuclear cells (PBMCs) obtained from relapsing-remitting MS patients (RRMS) was assessed using high content microarrays. Rapid onset of gene expression appeared within 4 h of subcutaneous IFN-beta administration, returning to baseline levels at 42 h in clinically stable RRMS. IFN-beta therapy in vivo rapidly but transiently induced strong upregulation of genes mediating immune modulation, IFN signaling, and antiviral responses. RT-PCR showed significant patient-to-patient variation in the magnitude of expression of multiple genes, especially for IFN-beta-inducible genes, such as MxA, IRF7, and CCL8, a Th1 product. Variation among patients in IFN-beta-induced RNA transcription was not explained by neutralizing antibodies or IFN receptor expression. Surprisingly, genes regulated in vivo by IFN-beta therapy do not support a simple Th1 to Th2 shift. A complex interplay between both proinflammatory and anti-inflammatory immune regulatory genes is likely to act in concert in the treatment of RRMS.


Subject(s)
Antibodies/immunology , Gene Expression Regulation/drug effects , Interferon-beta/pharmacology , Multiple Sclerosis, Relapsing-Remitting/genetics , Multiple Sclerosis, Relapsing-Remitting/immunology , Receptors, Interferon/genetics , Adult , Female , Flow Cytometry , Genes, Reporter , Humans , Inflammation/genetics , Interferon-beta/administration & dosage , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Male , Neutralization Tests , Oligonucleotide Array Sequence Analysis , Principal Component Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interferon/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics
14.
J Neuroimmunol ; 195(1-2): 116-20, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18279974

ABSTRACT

The molecular mechanism by which interferon beta (IFN-beta) is effective in treating multiple sclerosis is not well understood. Mononuclear cells from therapy-naïve MS patients, IFN-beta-1b-treated MS patients, and healthy controls were analyzed to examine mRNA changes that characterize both the disease and its treatment. The scientific literature was comprehensively searched for all protein-protein interactions. In MS patients who had never been treated with IFN-beta, statistical analysis revealed coordinate changes in mRNA expression for proteins reported in the literature as "regulated by IFN-beta." As a positive control for this approach, samples from a separate MS patient cohort showed significant change of these same genes during in vivo treatment with IFN-beta-1b.The strength of effect observed for regulation by IFN-beta was greater than for IFN-alpha, IFN-gamma (Th1), or IL-4 (Th2). Of the sets we investigated, the most strongly affected by disease was the subset defined by regulation by both IFN-beta and IFN-alpha. Changes in cells from therapy-naïve MS patients thus anticipated the importance of IFN-beta in therapy. These findings are a significant step towards marrying MS disease etiology and IFN-beta mechanism of action at a molecular level.


Subject(s)
Gene Expression Regulation/drug effects , Interferon-beta/pharmacology , Leukocytes, Mononuclear/metabolism , Multiple Sclerosis, Relapsing-Remitting/pathology , Cohort Studies , Female , Humans , Interferon-beta/therapeutic use , Leukocytes, Mononuclear/drug effects , Male , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Myasthenia Gravis , Oligonucleotide Array Sequence Analysis/methods , Time Factors
15.
Atherosclerosis ; 197(1): 204-11, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17466308

ABSTRACT

Atherosclerotic vascular disease is an inflammatory disease. Interferon-beta (IFN-beta) is an important immune modulator. However, the role of IFN-beta in atherosclerotic vascular disease is still not clear. The present study is designed to determine the effects of IFN-beta on atherosclerosis, abdominal aortic aneurysm (AAA) formation and proliferative vascular remodeling in apolipoprotein E (apoE) deficient mice. Six-month-old male apoE deficient mice fed a normal chow underwent ligation of the common left carotid artery, and were randomly assigned to receive either vehicle or angiotensin II (Ang II, 1.4 mg/kg daily) via a subcutaneously implanted osmotic infusion pump. The animals were further assigned to groups that were subjected to subcutaneous injection of vehicle or murine IFN-beta (10 MIU/kg, daily). Ang II increased atherosclerotic area in the non-ligated carotid artery and aortic arch, induced AAA, and exacerbated ligation-induced adventitial proliferation and neointimal hyperplasia characterized by smooth muscle cell (SMC) proliferation and macrophage infiltration in the ligated carotid artery. Co-treatment with IFN-beta, had no effects by itself, significantly attenuated Ang II-accelerated increase in the areas of neointima, adventitia, SMC and macrophage in the ligated carotid artery and suppressed Ang II-exacerbated atherosclerosis, but did not affect Ang II-induced AAA formation. These data indicate that IFN-beta can play a prominent anti-atherosclerosis, anti-inflammation, and anti-proliferation role of vasculoprotection.


Subject(s)
Angiotensin II/pharmacology , Apolipoproteins E/genetics , Atherosclerosis/drug therapy , Immunologic Factors/pharmacology , Interferon-beta/pharmacology , Vasoconstrictor Agents/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/immunology , Aorta, Thoracic/pathology , Aortic Aneurysm, Abdominal/drug therapy , Aortic Aneurysm, Abdominal/immunology , Aortic Aneurysm, Abdominal/pathology , Atherosclerosis/immunology , Atherosclerosis/pathology , Carotid Artery, Common/drug effects , Carotid Artery, Common/immunology , Carotid Artery, Common/pathology , Cell Division/drug effects , Drug Interactions , Foam Cells/pathology , Ligation , Male , Mice , Mice, Mutant Strains , Tunica Intima/drug effects , Tunica Intima/immunology , Tunica Intima/pathology
16.
Crit Rev Immunol ; 28(4): 269-79, 2008.
Article in English | MEDLINE | ID: mdl-19166380

ABSTRACT

IL-17 is a proinflammatory cytokine produced by immune cells. Its significance in host defense and disease development has been demonstrated in various infection and autoimmune models. Recently, additional studies have shown that IL-17 is also important in modulating airway immune response in several aspects. Along with the well-established Th1/Th2 model, new discoveries regarding the Th17 lineage and IL-17 functions have added an extra twist to the already complicated cytokine network that regulates airway immunity. The IL-17 receptor is expressed on blood cells, as well as on structural cells such as the epithelial cells in the airway. Therefore, the effect of IL-17 on airway immunity is very broad, covering both the innate and the adaptive aspects. In this review, we summarize the findings of recent studies on IL-17 signaling and function in pulmonary immune response, and the implications of IL-17 in disease pathogenesis.


Subject(s)
Immunity, Innate , Interleukin-17/physiology , Lung/immunology , Animals , Asthma/immunology , Bacterial Infections/immunology , Humans , Receptors, Interleukin-17/analysis , Receptors, Interleukin-17/physiology , Signal Transduction
17.
Am J Physiol Heart Circ Physiol ; 293(1): H69-76, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17434974

ABSTRACT

The present study tested the hypothesis that murine (m)IFN-beta or mIFN-alpha(2) can eliminate cardiac viral load and protect cardiomyocytes from injury in animals infected with coxsackievirus B3 (CVB3). CVB3-inoculated male Balb/c mice exhibited signs of illness, including lethargy, progressive weight loss, and death (10% on day 3 and 100% on day 8). Cardiac viral load was high [4,277 +/- 1,009 plaque-forming units and 25 +/- 5 copies CVB3/hypoxanthine guanine phosphoribosyl transferase 1 mRNA] on day 4. The cardiac tissue exhibited severe inflammatory infiltration and myocyte damage with an average myocarditis integrated pathology score of 2.1 +/- 0.2 on day 7. Most of the mice infected with CVB3 also developed epicarditis, and 55% had intraventricular thrombi present. Treatment with mIFN-beta [2.5 to 10 million international units (MIU)/kg] dose-dependently improved the general health status in CVB3-inoculated mice, as evidenced by reduction in weight loss, prevention of death, elimination of cardiac viral load, protection of myocytes from injury, decrease in inflammatory cell infiltration, and attenuation of intraventricular thrombus formation. Treatment with 10 MIU/kg mIFN-alpha(2) resulted in a similar level of efficacy as that induced by 5 MIU/kg mIFN-beta, with the exception that mIFN-alpha(2) did not reduce cardiac CVB3 mRNA. However, mIFN-alpha(2) , but not any dose group of mIFN-beta, significantly attenuated CVB3-induced epicarditis. These data demonstrate antiviral effects for both mIFN-beta and mIFN-alpha(2), which lead to protection of the mice from CVB3-induced myocarditis. However, the potential mechanisms leading to a differential host response for the two isoforms of mIFN remain to be elucidated.


Subject(s)
Enterovirus/drug effects , Interferon-alpha/administration & dosage , Interferon-beta/administration & dosage , Myocarditis/drug therapy , Myocarditis/virology , Pericardium/drug effects , Pericardium/virology , Animals , Antiviral Agents/administration & dosage , Cardiotonic Agents/administration & dosage , Cells, Cultured , Male , Mice , Mice, Inbred BALB C , Muscle Cells/drug effects , Muscle Cells/virology , Treatment Outcome
18.
Int J Cancer ; 111(1): 32-42, 2004 Aug 10.
Article in English | MEDLINE | ID: mdl-15185340

ABSTRACT

In addition to antiviral effects, Type I interferons (IFN) have potent antiproliferative and immunomodulatory activities. Because of these properties IFNs have been evaluated as therapeutics for the treatment of a number of human diseases, including cancer. Currently, IFNs have been shown to be efficacious for the treatment of only a select number of cancers. The reason for this is unclear. Recent evidence has demonstrated that some cancer cell types seem to be defective in their ability to respond to IFN. It has been suggested that defects in IFN signaling is one mechanism by which cancer cells escape responsiveness to Type I IFNs and growth control in general. We report that transfection and enhanced expression of the Type I IFN receptor chain (IFNAR2c) in 3 different human cancer cell lines markedly increases the sensitivity of these cells to the antiproliferative effects of IFNs. In cancer cells transfected with IFNAR2c, dose response curves demonstrate a significant decrease in the concentrations of IFN required to achieve maximum cell death. Furthermore, in these transfected cells, we observe a significant increase in the number of cells undergoing apoptosis, as measured by DNA fragmentation and Caspase 3 activation. In addition, using an in vivo xenograft tumor model we show an increase in the effectiveness of systemically delivered Betaseron in decreasing tumor burden in animals in which solid tumors were generated from IFNAR2c transfected cells. These data show that specific regulation of IFN receptor expression can play a major role in determining the clinical outcome of IFN-based cancer therapeutics by regulating the relative sensitivity of cancer cells to IFN-dependent growth control.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic , Interferons/pharmacology , Receptors, Interferon/biosynthesis , Animals , Apoptosis , Caspase 3 , Caspases/pharmacology , Cell Line, Tumor , DNA Damage , Female , Genetic Therapy/methods , Humans , Mice , Mice, Nude , Receptor, Interferon alpha-beta , Receptors, Interferon/physiology , Signal Transduction , Transfection , Transplantation, Heterologous
19.
J Biol Chem ; 277(38): 35635-41, 2002 Sep 20.
Article in English | MEDLINE | ID: mdl-12105218

ABSTRACT

Human type I interferons (IFNs) play an important role in the regulation of antiviral defense mechanisms, immunomodulatory activities, and growth control. Recent efforts have demonstrated the importance of IFNs in the activation of signal transducers and activators of transcription (STATs). The role of STAT1 and STAT2 in IFN-dependent JAK-STAT signaling is well established; however, the role of STAT3 and its activation by IFNs remains unclear. Understanding the IFN-dependent regulation of STAT3 is of increasing interest because recent studies have demonstrated that STAT3 may play a role in cancer. Studies have revealed that STAT3 is constitutively active in a number of cancer cell lines and that overexpression of an active form of STAT3 transforms normal fibroblasts. Therefore, STAT3 exhibits properties indicative of known oncogenes. In this report, we define the role of the type I IFN receptor in STAT3 activation and identify for the first time tyrosine residues present in the cytoplasmic domain of IFNAR2c that are critical for STAT3 activation. The regulation of STAT3 activation by IFNs was measured in a human lung fibrosarcoma cell line lacking IFNAR2c but stably expressing various IFNAR2c tyrosine mutants. We show here that in addition to IFN-dependent tyrosine phosphorylation of STAT3, activation using a STAT3-dependent electrophoretic mobility shift assay and a STAT3-specific reporter can also be demonstrated. Furthermore, we demonstrate that type I IFN-dependent activation of STAT3 proceeds through a novel mechanism that is dependent on two tyrosines, Tyr(337) and Tyr(512), present in IFNAR2c and contained within a conserved six-amino acid residue motif, GxGYxM. Surprisingly, both tyrosines were previously shown to be required for type I IFN-dependent STAT1 and STAT2 activation. Our results reveal that type I IFNs activate multiple STATs via the overlapping usage of two tyrosine residues located in the cytoplasmic domain of IFNAR2c.


Subject(s)
DNA-Binding Proteins/metabolism , Interferon Type I/metabolism , Receptors, Interferon/metabolism , Trans-Activators/metabolism , Tyrosine/metabolism , Base Sequence , DNA Primers , Electrophoretic Mobility Shift Assay , Humans , Membrane Proteins , Receptor, Interferon alpha-beta , Receptors, Interferon/chemistry , STAT3 Transcription Factor , Tumor Cells, Cultured
20.
J Biol Chem ; 277(2): 1493-9, 2002 Jan 11.
Article in English | MEDLINE | ID: mdl-11682488

ABSTRACT

Type I interferons (IFNs) are cytokines that play a central role in mediating antiviral, antiproliferative, and immunomodulatory activities in virtually all cells. These activities are entirely dependent on the interaction of IFNs with their particular cell surface receptor. In this report, we identify two specific tyrosine residues located within the cytoplasmic domain of IFNAR2c that are obligatory for IFN-dependent signaling. Various IFNAR2c tyrosine mutants were expressed in a human lung fibroscarcoma cell line lacking IFNAR2c (U5A). Stable clones expressing these mutants were analyzed for their ability to induce STAT1 and STAT2 activation, ISGF3 transcriptional complex formation, gene expression, and cell growth regulation in response to stimulation with type I IFNs. The replacement of all seven cytoplasmic tyrosine residues of IFNAR2c with phenylalanine resulted in a receptor unable to respond to IFN stimulation. Substitution of single tyrosines at amino acid residue 269, 316, 318, 337, or 512 with phenylalanine had no effect on IFN-dependent signaling, suggesting that no single tyrosine is essential for IFN receptor-mediated signaling. In addition, IFNAR2c retaining five proximal tyrosines residues (269, 306, 316, 318, and 337) or either two distal tyrosine residues (411 or 512) continued to be responsive to IFN stimulation. Surprisingly, the presence of only a single tyrosine at either position 337 or 512 was sufficient to restore a complete IFN response. These results indicate that IFN-dependent signaling proceeds through the redundant usage of two tyrosine residues in the cytoplasmic domain of IFNAR2c.


Subject(s)
Interferon Type I/metabolism , Receptors, Interferon/metabolism , Signal Transduction/physiology , Tyrosine/metabolism , Animals , Cell Division/drug effects , Cell Division/physiology , Cell Line , DNA-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Interferon Type I/pharmacology , Interferon-Stimulated Gene Factor 3 , Interferon-Stimulated Gene Factor 3, gamma Subunit , Membrane Proteins , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Receptor, Interferon alpha-beta , Receptors, Interferon/chemistry , STAT1 Transcription Factor , Trans-Activators/metabolism , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL