Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Innate Immun ; 27(2): 133-142, 2021 02.
Article in English | MEDLINE | ID: mdl-33241977

ABSTRACT

The capacity for macrophages to polarize into distinct functional activation states (e.g., M1, M2) is critical to tune an inflammatory response to the relevant infection or injury. Alternative or M2 polarization of macrophages is most often achieved in vitro in response to IL-4/IL-13 and results in the transcriptional up-regulation of a constellation of characteristic M2 marker genes. In vivo, additional signals from the inflammatory milieu can further increase or decrease M2 marker expression. Particularly, activation of cAMP-generating G protein-coupled receptors is reported to increase M2 markers, but whether this is strictly dependent upon cAMP production is unclear. We report herein that increased cAMP alone can increase IL-4-dependent M2 marker expression through a PKA/C/EBPß/CREB dependent pathway in murine macrophages.


Subject(s)
Biomarkers/metabolism , Cyclic AMP/metabolism , Macrophages/metabolism , Animals , Cell Differentiation , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation , Interleukin-4/metabolism , Macrophage Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , RAW 264.7 Cells , Signal Transduction , Steroid Isomerases/metabolism , Th2 Cells/immunology
2.
Front Immunol ; 11: 629399, 2020.
Article in English | MEDLINE | ID: mdl-33633747

ABSTRACT

Neoantigen formation due to the interaction of drug molecules with human leukocyte antigen (HLA)-peptide complexes can lead to severe hypersensitivity reactions. Flucloxacillin (FLX), a ß-lactam antibiotic for narrow-spectrum gram-positive bacterial infections, has been associated with severe immune-mediated drug-induced liver injury caused by an influx of T-lymphocytes targeting liver cells potentially recognizing drug-haptenated peptides in the context of HLA-B*57:01. To identify immunopeptidome changes that could lead to drug-driven immunogenicity, we used mass spectrometry to characterize the proteome and immunopeptidome of B-lymphoblastoid cells solely expressing HLA-B*57:01 as MHC-I molecules. Selected drug-conjugated peptides identified in these cells were synthesized and tested for their immunogenicity in HLA-B*57:01-transgenic mice. T cell responses were evaluated in vitro by immune assays. The immunopeptidome of FLX-treated cells was more diverse than that of untreated cells, enriched with peptides containing carboxy-terminal tryptophan and FLX-haptenated lysine residues on peptides. Selected FLX-modified peptides with drug on P4 and P6 induced drug-specific CD8+ T cells in vivo. FLX was also found directly linked to the HLA K146 that could interfere with KIR-3DL or peptide interactions. These studies identify a novel effect of antibiotics to alter anchor residue frequencies in HLA-presented peptides which may impact drug-induced inflammation. Covalent FLX-modified lysines on peptides mapped drug-specific immunogenicity primarily at P4 and P6 suggesting these peptide sites as drivers of off-target adverse reactions mediated by FLX. FLX modifications on HLA-B*57:01-exposed lysines may also impact interactions with KIR or TCR and subsequent NK and T cell function.


Subject(s)
Floxacillin/immunology , HLA-B Antigens/immunology , Haptens/immunology , Peptides/immunology , Animals , Cell Line , HLA-B Antigens/genetics , Humans , Mice , Mice, Transgenic , Peptides/genetics
3.
Sci Rep ; 8(1): 11477, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30065306

ABSTRACT

Therapeutic proteins can induce immune responses that affect their safety and efficacy. Product aggregates and innate immune response modulating impurities (IIRMI) are risk factors of product immunogenicity. In this study, we use Intravenous Immunoglobulin (IVIG), Avastin, and Human Serum Albumin (HSA) to explore whether increased aggregates activate innate immune cells or modify the response to IIRMI. We show that increased aggregates (shaken or stirred) in IVIG and Avastin, but not HSA, induced activation of MAPKs (pp38, pERK and pJNK) and transcription of immune-related genes including IL8, IL6, IL1ß, CSF1, CCL2, CCL7, CCL3, CCL24, CXCL2, IRAK1, EGR2, CEBPß, PPARg and TNFSF15 in human PBMC. The immunomodulatory effect was primarily mediated by FcγR, but not by TLR. Interestingly, increased aggregates in IVIG or Avastin magnified innate immune responses to TLR2/4 agonists, but diminished responses to TLR3/9 agonists. This study shows that IIRMI and aggregates can modify the activity of immune cells potentially modifying the milieu where the products are delivered highlighting the complex interplay of different impurities on product immunogenicity risk. Further, we show that aggregates could modify the sensitivity of PBMC-based assays designed to detect IIRMI. Understanding and managing immunogenicity risk is a critical component of product development and regulation.


Subject(s)
Immunity, Innate/immunology , Immunoglobulins, Intravenous/immunology , Serum Albumin, Human/immunology , Animals , Antibody Formation/immunology , Bevacizumab/immunology , Cells, Cultured , HEK293 Cells , Humans , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/immunology , Transcription, Genetic/immunology
4.
Thromb Res ; 159: 39-47, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28968537

ABSTRACT

INTRODUCTION: Heparin-induced thrombocytopenia (HIT) is an immune-mediated complication of heparin anticoagulation therapy resulting in thrombocytopenia frequently accompanied by thrombosis. Current evidence suggests that HIT is associated with antibodies developed in response to multi-molecular complexes formed by platelet factor 4 (PF4) bound to heparin or cell surface glycosaminoglycans. These antibody complexes activate platelets and monocytes typically through FcγRIIA receptors increasing the production of PF4, inflammatory mediators, tissue factor and thrombin. The influence of underlying events in HIT including complex-induced pro-inflammatory cell activation and structural determinants leading to local inflammatory responses are not fully understood. METHODS: The stoichiometry and complex component requirements were determined by incubating fresh peripheral blood mononuclear cells (PBMC) with different concentrations of unfractionated heparin (H), low molecular weight heparin (LMWH), PF4- and anti-PF4-H complex antibodies (KKO). Cytokine mRNA or protein were measured by qRT-PCR or Meso Scale Discovery technology, respectively. Gene expression profile analysis for 594 genes was performed using Nanostring technology and analyzed using Ingenuity Pathway Analysis software. RESULTS AND CONCLUSIONS: The data show that antibodies magnify immune responses induced in PBMCs by PF4 alone or in complex with heparin or LMWH. We propose that following induction of HIT antibodies by heparin-PF4 complexes, binding of the antibodies to PF4 is sufficient to induce a local pro-inflammatory response which may play a role in the progression of HIT. In vitro assays using PBMCs may be useful in characterizing local inflammatory and innate immune responses induced by HIT antibodies in the presence of PF4 and different sources of heparins. FDA DISCLAIMER: The findings and conclusions in this article are solely the responsibility of the authors and are not being formally disseminated by the Food and Drug Administration. Thus, they should not be construed to represent any Agency determination or policy.


Subject(s)
Antibodies, Monoclonal/immunology , Heparin, Low-Molecular-Weight/adverse effects , Inflammation/immunology , Platelet Factor 4/immunology , Thrombocytopenia/chemically induced , Thrombocytopenia/immunology , Cell Line , Humans , Immunity, Innate , Inflammation/blood , Leukocytes, Mononuclear/immunology , Platelet Factor 4/blood , Thrombocytopenia/blood
5.
Sci Rep ; 7(1): 10490, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28874687

ABSTRACT

Immunogenicity can have devastating consequences on the safety and efficacy of therapeutic proteins. Therefore, evaluating and mitigating the risk of product immunogenicity is critical for the development these products. This study, showed that Betaseron and Extavia, which are reported to be more immunogenic among IFNß products in clinical usage, contain residual innate immune response modulating impurities (IIRMIs) capable of activating NF-κB and induced expression of inflammatory mediators. These IIRMIs were undetectable in Rebif or Avonex. The stimulatory effect was attributed solely to IIRMIs because it was evident in murine cells lacking the interferon receptor (IFNAR). The IIRMIs in Betaseron and Extavia triggered NF-κB activation in HEK-293 cells bearing TLR2 and TLR4 in MyD88 dependent manner. Importantly, the IIRMIs in Betaseron induced up-regulation of IL-6, IL-1ß, and ccl5 in the skin of IFNAR knock out mice following subcutaneous administration. This indicates that trace level IIRMIs in Betaseron could contribute to the higher immunogenicity rates seen in clinics. Together these data suggest that cell based assays can reveal subtle but clinically relevant differences in IIRMIs following manufacturing changes or between products with the same active ingredients but different manufacturing processes. Appreciating these differences may inform immunogenicity risk assessments.


Subject(s)
Drug Contamination , Interferon beta-1b/standards , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cells, Cultured , HEK293 Cells , Humans , Interferon beta-1b/chemistry , Interleukins/metabolism , Mice , Mice, Inbred C57BL
6.
J Pharm Sci ; 106(3): 751-760, 2017 03.
Article in English | MEDLINE | ID: mdl-27923493

ABSTRACT

Unwanted immune responses to therapeutic proteins can severely impact their safety and efficacy. Studies show that the presence of trace amounts of host cells and process-related impurities that stimulate pattern recognition receptors (PRR) can cause local inflammation and enhance product immunogenicity. Here we used purified PRR agonists as model impurities to assess the minimal level of individual innate immune response modulating impurities (IIRMIs) that could activate a local immune response. We show that levels of endotoxin as low as 10 pg (0.01 EU), 1 ng for polyinosinic:polycytidylic acid (PolyI:C), 100 ng for synthetic diacylated liopprotein, thiazoloquinolone compound, or muramyl dipeptide, 1 µg for flagellin or ß-glucan, or 5 µg for CpG-oligodeoxynucleotide increased expression of genes linked to innate immune activation and inflammatory processes in the skin of rhesus macaques. Furthermore, spiking studies using rasburicase as a model therapeutic showed that the levels of PRR agonists that induced detectable gene upregulation in the skin were associated with increased immunogenicity for rasburicase. This study underscores the need for testing multiple IIRMIs in biologics, strengthening the connection between the local mRNA induction in skin, innate immune activation, and antibody development in primates, and provides an indication of the levels of IIRMI in therapeutic products that could impact product immunogenicity.


Subject(s)
Endotoxins/immunology , Immunity, Innate/immunology , Immunogenetic Phenomena/physiology , Models, Animal , Skin/immunology , Animals , Antibodies/immunology , Endotoxins/administration & dosage , Female , HEK293 Cells , Humans , Immunity, Innate/drug effects , Immunogenetic Phenomena/drug effects , Injections, Subcutaneous , Macaca mulatta , Male , Poly C/administration & dosage , Poly C/immunology , Receptors, Pattern Recognition/immunology , Skin/drug effects
7.
Breast Cancer Res Treat ; 145(1): 5-22, 2014 May.
Article in English | MEDLINE | ID: mdl-24668500

ABSTRACT

Nicotine, a main addictive compound in tobacco smoke, has been linked to promotion and progression of lung, head and neck, pancreatic, and breast cancers, but the detailed mechanisms of cancer progression remain elusive. Here, we show that nicotine induces the expression of galectin-3 (an anti-apoptotic ß-galactoside-binding lectin) in breast cancer cell line and in primary tumors from breast cancer patients. Nicotine-induced up regulation of galectin-3 is due to an increased expression of α9 isoform of nicotinic acetylcholine receptor (α9nAChR), which activates transcription factor STAT3 that in turn, physically binds to galectin-3 (LGALS3) promoter and induces transcription of galectin-3. Intracellular galectin-3 increased mitochondrial integrity and suppressed chemotherapeutic-induced apoptosis of breast cancer cell. Moreover, nicotine-induced enrichment of side population cells with cancer stem cell-like properties was modulated by galectin-3 expression and could be significantly reduced by transient knock down of LGALS3 and its upstream signaling molecules STAT3 and α9nAChR. Thus, galectin-3 or its upstream signaling molecule STAT3 or α9nAChR could be a potential target to prevent nicotine-induced chemoresistance in breast cancer.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Signal Transduction/drug effects , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Fragmentation , Galectin 3/biosynthesis , Humans , Immunohistochemistry , Neoplastic Stem Cells/pathology , Real-Time Polymerase Chain Reaction , Receptors, Nicotinic/metabolism , STAT3 Transcription Factor/metabolism , Side-Population Cells , Transfection
8.
PLoS Pathog ; 9(7): e1003479, 2013.
Article in English | MEDLINE | ID: mdl-23853595

ABSTRACT

The cell surface/endosomal Toll-like Receptors (TLRs) are instrumental in initiating immune responses to both bacteria and viruses. With the exception of TLR2, all TLRs and cytosolic RIG-I-like receptors (RLRs) with known virus-derived ligands induce type I interferons (IFNs) in macrophages or dendritic cells. Herein, we report that prior ligation of TLR2, an event previously shown to induce "homo" or "hetero" tolerance, strongly "primes" macrophages for increased Type I IFN production in response to subsequent TLR/RLR signaling. This occurs by increasing activation of the transcription factor, IFN Regulatory Factor-3 (IRF-3) that, in turn, leads to enhanced induction of IFN-ß, while expression of other pro-inflammatory genes are suppressed (tolerized). In vitro or in vivo "priming" of murine macrophages with TLR2 ligands increase virus-mediated IFN induction and resistance to infection. This priming effect of TLR2 is mediated by the selective upregulation of the K63 ubiquitin ligase, TRAF3. Thus, we provide a mechanistic explanation for the observed antiviral actions of MyD88-dependent TLR2 and further define the role of TRAF3 in viral innate immunity.


Subject(s)
Cellular Reprogramming , Immunity, Innate , Interferon Type I/biosynthesis , Macrophages, Peritoneal/immunology , TNF Receptor-Associated Factor 3/metabolism , Toll-Like Receptor 2/metabolism , Up-Regulation , Animals , Cell Line , Cells, Cultured , Female , Humans , Influenza A virus/immunology , Interferon Type I/genetics , Interferon Type I/metabolism , Ligands , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Recombinant Proteins/metabolism , Signal Transduction , TNF Receptor-Associated Factor 3/genetics , Toll-Like Receptor 2/genetics , Vaccinia virus/immunology , Vesicular stomatitis Indiana virus/immunology
9.
J Immunol ; 190(9): 4763-72, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23543762

ABSTRACT

Endotoxin tolerance is a complex phenomenon characterized primarily by decreased production of proinflammatory cytokines, chemokines, and other inflammatory mediators, whereas the expression of other genes are induced or unchanged. Endotoxin tolerance is induced by prior exposure of murine macrophages/human monocytes, experimental animals, or people to TLR ligands. Although recent studies reported a possible relationship between endotoxin tolerance and differentiation of alternatively activated macrophages (AA-MΦs or M2), we show in this study that LPS pretreatment of IL-4Rα(-/-) and STAT6(-/-) macrophages, which fail to develop into AA-MΦs, resulted in tolerance of proinflammatory cytokines, as well as molecules and chemokines previously associated with AA-MΦs (e.g., arginase-1, mannose receptor, CCL2, CCL17, and CCL22). In contrast to LPS, wild-type (WT) MΦs pretreated with IL-4, the prototype inducer of AA-MΦs, did not induce endotoxin tolerance with respect to proinflammatory cytokines, AA-MΦ-associated chemokines, negative regulators, NF-κB binding and subunit composition, and MAPKs; conversely, IL-13(-/-) macrophages were tolerized equivalently to WT MΦs by LPS pretreatment. Further, IL-4Rα deficiency did not affect the reversal of endotoxin tolerance exerted by the histone deacetylase inhibitor trichostatin A. Like WT mice, 100% of LPS-tolerized IL-4Rα-deficient mice survived LPS + d-galactosamine-induced lethal toxicity and exhibited decreased serum levels of proinflammatory cytokines and AA-MΦ-associated chemokines induced by LPS challenge compared with nontolerized mice. These data indicate that the signaling pathways leading to endotoxin tolerance and differentiation of AA-MΦs are dissociable.


Subject(s)
Cell Differentiation/immunology , Endotoxins/immunology , Immune Tolerance/immunology , Macrophages/immunology , Animals , Cell Differentiation/genetics , Cell Line , Endotoxins/metabolism , Immune Tolerance/genetics , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-13/metabolism , Lipopolysaccharides/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/immunology , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/immunology , STAT6 Transcription Factor/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Transcriptome/genetics , Transcriptome/immunology
10.
Proc Natl Acad Sci U S A ; 109(44): 18054-9, 2012 Oct 30.
Article in English | MEDLINE | ID: mdl-23071332

ABSTRACT

Gaucher disease (GD) is an autosomal recessive disorder caused by mutations in the acid ß-glucocerebrosidase gene. To model GD, we generated human induced pluripotent stem cells (hiPSC), by reprogramming skin fibroblasts from patients with type 1 (N370S/N370S), type 2 (L444P/RecNciI), and type 3 (L444P/L444P) GD. Pluripotency was demonstrated by the ability of GD hiPSC to differentiate to all three germ layers and to form teratomas in vivo. GD hiPSC differentiated efficiently to the cell types most affected in GD, i.e., macrophages and neuronal cells. GD hiPSC-macrophages expressed macrophage-specific markers, were phagocytic, and were capable of releasing inflammatory mediators in response to LPS. Moreover, GD hiPSC-macrophages recapitulated the phenotypic hallmarks of the disease. They exhibited low glucocerebrosidase (GC) enzymatic activity and accumulated sphingolipids, and their lysosomal functions were severely compromised. GD hiPSC-macrophages had a defect in their ability to clear phagocytosed RBC, a phenotype of tissue-infiltrating GD macrophages. The kinetics of RBC clearance by types 1, 2, and 3 GD hiPSC-macrophages correlated with the severity of the mutations. Incubation with recombinant GC completely reversed the delay in RBC clearance from all three types of GD hiPSC-macrophages, indicating that their functional defects were indeed caused by GC deficiency. However, treatment of induced macrophages with the chaperone isofagomine restored phagocytosed RBC clearance only partially, regardless of genotype. These findings are consistent with the known clinical efficacies of recombinant GC and isofagomine. We conclude that cell types derived from GD hiPSC can effectively recapitulate pathologic hallmarks of the disease.


Subject(s)
Gaucher Disease/pathology , Pluripotent Stem Cells/cytology , Cell Differentiation , Cell Lineage , Humans , Macrophage Activation , Models, Biological
11.
J Immunol ; 189(1): 50-60, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22634618

ABSTRACT

IL-33, a member of the IL-1 family of cytokines, is produced by many cell types, including macrophages, yet its regulation is largely unknown. Treatment of primary murine macrophages with a panel of TLR (e.g., TLR2, TLR3, TLR4, and TLR9) agonists and non-TLR (e.g., MDA5, RIG-I) agonists revealed a pattern of gene and protein expression consistent with a role for IFN regulatory factor-3 (IRF-3) in the expression of IL-33. Accordingly, induction of IL-33 mRNA was attenuated in IRF-3(-/-) macrophages and TBK-1(-/-) mouse embryonic fibroblasts. Despite the fact that all IL-33 agonists were IRF-3 dependent, LPS-induced IL-33 mRNA was fully inducible in IFN-ß(-/-) macrophages, indicating that IL-33 is not dependent on IFN-ß as an intermediate. Epinephrine and Bordetella pertussis adenylate cyclase toxin (ACT), cAMP-activating agents, activate CREB and greatly synergize with LPS to induce IL-33 mRNA in macrophages. Both LPS-induced and ACT/LPS-enhanced expression of IL-33 mRNA was partially, but significantly, inhibited by the protein kinase A inhibitor H-89 but not by tyrosine kinase or protein kinase C inhibitors. Two IL-33 mRNA species derived from two alternative promoters encode full-length IL-33; however, the shorter "A" species is preferentially induced by all IL-33-inducing agonists except Newcastle disease virus, a RIG-I agonist that induced expression of both "A" and "B" transcripts. Together, these studies greatly extend what is currently known about the regulation of IL-33 induction in macrophages stimulated by bacterial and viral agonists that engage distinct innate immune signaling pathways.


Subject(s)
Interleukins/biosynthesis , Toll-Like Receptors/agonists , Toll-Like Receptors/physiology , Transcriptional Activation/immunology , Animals , Cells, Cultured , Fibroblasts/immunology , Fibroblasts/microbiology , Fibroblasts/virology , Immunity, Innate/genetics , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-3/genetics , Interleukin-33 , Interleukins/genetics , Ligands , Macrophages/immunology , Macrophages/microbiology , Macrophages/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/biosynthesis , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptors/metabolism , Transcriptional Activation/genetics
12.
J Biol Chem ; 286(4): 2817-25, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21123181

ABSTRACT

Inflammation underlying immune pathology and tissue damage involves an intricate interplay between multiple immunological and biochemical mediators. Cytokines represent the key immune mediators that trigger a cascade of reactions that drive processes such as angiogenesis and proteolytic damage to tissues. IL-17 has now been shown to be a pivotal cytokine in many autoimmune diseases, supplanting the traditional Th1-Th2 paradigm. Also, the dual role of proinflammatory IFN-γ has unraveled new complexities in the cytokine biology of such disorders. A major hurdle in fully understanding the effector pathways in these disorders is the lack of information regarding the temporal kinetics of the cytokines during the course of the disease, as well as the interplay among the key cytokines. Using an experimental model of arthritic inflammation, we demonstrate that the temporal expression of cytokines during the incubation phase is a critical determinant of disease susceptibility. The susceptible rats raised a vigorous IL-17 response early, followed by IFN-γ and IL-27 response in that sequence, whereas the resistant rats displayed an early and concurrent response to these three cytokines. Accordingly, treatment with exogenous IFN-γ/IL-27 successfully controlled arthritic inflammation and inhibited the defined mediators of inflammation, angiogenesis, cell survival, apoptosis, and tissue damage. Furthermore, IFN-γ enhanced IL-27 secretion, revealing a cooperative interplay between the two cytokines. Our results offer a novel immunobiochemical perspective on the pathogenesis of autoimmune arthritis and its therapeutic control.


Subject(s)
Arthritis, Experimental/metabolism , Autoimmune Diseases/metabolism , Interferon-gamma/metabolism , Interleukins/metabolism , Angiogenesis Inducing Agents/immunology , Angiogenesis Inducing Agents/metabolism , Animals , Apoptosis/immunology , Arthritis, Experimental/chemically induced , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Autoimmune Diseases/chemically induced , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Cell Survival/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Interferon-gamma/immunology , Interferon-gamma/pharmacology , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukins/immunology , Interleukins/pharmacology , Rats , Rats, Inbred Lew , Rats, Inbred WKY , Species Specificity
13.
Arthritis Rheum ; 60(1): 103-13, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19116924

ABSTRACT

OBJECTIVE: Pretreatment of Lewis rats with soluble mycobacterial Hsp65 affords protection against subsequent adjuvant-induced arthritis (AIA). This study was aimed at unraveling the mechanisms underlying mycobacterial Hsp65-induced protection against arthritis, using contemporary parameters of immunity. METHODS: Lewis rats were given 3 intraperitoneal injections of mycobacterial Hsp65 in solution prior to the initiation of AIA with heat-killed Mycobacterium tuberculosis. Thereafter, mycobacterial Hsp65-specific T cell proliferative, cytokine, and antibody responses were tested in tolerized rats. The roles of anergy and the indoleamine 2,3 dioxygenase (IDO)-tryptophan pathway in tolerance induction were assessed, and the frequency and suppressive function of CD4+FoxP3+ Treg cells were monitored. Also tested was the effect of mycobacterial Hsp65 tolerization on T cell responses to AIA-related mycobacterial Hsp70, mycobacterial Hsp10, and rat Hsp65. RESULTS: The AIA-protective effect of mycobacterial Hsp65-induced tolerance was associated with a significantly reduced T cell proliferative response to mycobacterial Hsp65, which was reversed by interleukin-2 (IL-2), indicating anergy induction. The production of interferon-gamma (but not IL-4/IL-10) was increased, with concurrent down-regulation of IL-17 expression by mycobacterial Hsp65-primed T cells. Neither the frequency nor the suppressive activity of CD4+FoxP3+ T cells changed following tolerization, but the serum level of anti-mycobacterial Hsp65 antibodies was increased. However, no evidence was observed for a role of IDO or cross-tolerance to mycobacterial Hsp70, mycobacterial Hsp10, or rat Hsp65. CONCLUSION: Tolerization with soluble mycobacterial Hsp65 leads to suppression of IL-17, anergy induction, and enhanced production of anti-mycobacterial Hsp65 antibodies, which play a role in protection against AIA. These results are relevant to the development of effective immunotherapeutic approaches for autoimmune arthritis.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Experimental/prevention & control , Bacterial Proteins/pharmacology , Chaperonins/pharmacology , Immune Tolerance/immunology , Interferon-gamma/metabolism , Interleukin-17/metabolism , Animals , Autoantibodies/immunology , Bacterial Proteins/immunology , CD4 Antigens/metabolism , Chaperonin 10/immunology , Chaperonin 10/pharmacology , Chaperonin 60 , Chaperonins/immunology , Cross Reactions/immunology , Down-Regulation/immunology , Forkhead Transcription Factors/metabolism , HSP70 Heat-Shock Proteins/immunology , HSP70 Heat-Shock Proteins/pharmacology , Immune Tolerance/drug effects , Injections, Intraperitoneal , Interleukin-2/metabolism , Male , Rats , Rats, Inbred Lew , Solubility , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Up-Regulation/immunology
14.
J Immunol ; 179(1): 236-46, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17579043

ABSTRACT

Ligation of FcgammaR concurrent with LPS stimulation of murine macrophages results in decreased IL-12 and increased IL-10 production. Because PI3K deficiency has been associated with increased IL-12, we hypothesized that PI3K was central to the anti-inflammatory effect of FcgammaR ligation on TLR-induced IL-12. FcgammaR ligation of macrophages increased pAKT, a correlate of PI3K activity, above levels induced by TLR4 or TLR2 agonists. This increase was blocked by PI3K inhibitors, wortmannin or LY294002, as was the effect of FcgammaR ligation on TLR-induced IL-12 and IL-10. LPS-induced binding of NF-kappaB to the IL-12 p40 promoter NF-kappaB-binding site was not affected by FcgammaR ligation at 1 h; however, by 4 h, NF-kappaB binding was markedly inhibited, confirmed in situ by chromatin immunoprecipitation analysis. This effect was wortmannin sensitive. Although TLR-induced IkappaBalpha degradation was not affected by FcgammaR ligation, IkappaBalpha accumulated in the nuclei of cells treated with LPS and FcgammaR ligation for 4 h, and was blocked by PI3K inhibitors. LPS-induced IFN regulatory factor-8/IFN consensus sequence-binding protein mRNA, and an IFN regulatory factor-8-dependent gene, Nos2, were inhibited by concurrent FcgammaR ligation, and this was also reversed by wortmannin. Thus, FcgammaR ligation modulates LPS-induced IL-12 via multiple PI3K-sensitive pathways that affect production, accumulation, and binding of key DNA-binding proteins required for IL-12 induction.


Subject(s)
Interleukin-10/genetics , Interleukin-12/genetics , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Phosphatidylinositol 3-Kinases/physiology , Receptors, IgG/metabolism , Toll-Like Receptors/physiology , Transcription, Genetic/immunology , Animals , Cells, Cultured , Enzyme Inhibitors/pharmacology , Interleukin-10/biosynthesis , Interleukin-12/biosynthesis , Ligands , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Oncogene Protein v-akt/antagonists & inhibitors , Oncogene Protein v-akt/physiology , Phosphoinositide-3 Kinase Inhibitors , Protein Binding/genetics , Protein Binding/immunology , Receptors, IgG/immunology , Signal Transduction/genetics , Signal Transduction/immunology
15.
J Immunol ; 178(6): 3602-11, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17339457

ABSTRACT

IFN regulatory factor (IRF)-2(-/-) mice are significantly more resistant to LPS challenge than wild-type littermates, and this was correlated with increased numbers of apoptotic Kupffer cells. To assess the generality of this observation, and to understand the role of IRF-2 in apoptosis, responses of peritoneal macrophages from IRF-2(+/+) and IRF-2(-/-) mice to apoptotic stimuli, including the fungal metabolite, gliotoxin, were compared. IRF-2(-/-) macrophages exhibited a consistently higher incidence of apoptosis that failed to correlate with caspase-3/7 activity. Using microarray gene expression profiling of liver RNA samples derived from IRF-2(+/+) and IRF-2(-/-) mice treated with saline or LPS, we identified >40 genes that were significantly down-regulated in IRF-2(-/-) mice, including Stat3, which has been reported to regulate apoptosis. Compared with IRF-2(+/+) macrophages, STAT3alpha mRNA was up-regulated constitutively or after gliotoxin treatment of IRF-2(-/-) macrophages, whereas STAT3beta mRNA was down-regulated. Phospho-Y705-STAT3, phospho-S727-STAT1, and phospho-p38 protein levels were also significantly higher in IRF-2(-/-) than control macrophages. Activation of the STAT signaling pathway has been shown to elicit expression of CASP1 and apoptosis. IRF-2(-/-) macrophages exhibited increased basal and gliotoxin-induced caspase-1 mRNA expression and enhanced caspase-1 activity. Pharmacologic inhibition of STAT3 and caspase-1 abolished gliotoxin-induced apoptosis in IRF-2(-/-) macrophages. A novel IFN-stimulated response element, identified within the murine promoter of Casp1, was determined to be functional by EMSA and supershift analysis. Collectively, these data support the hypothesis that IRF-2 acts as a transcriptional repressor of Casp1, and that the absence of IRF-2 renders macrophages more sensitive to apoptotic stimuli in a caspase-1-dependent process.


Subject(s)
Apoptosis/immunology , Caspase 3/immunology , Caspase 7/immunology , Interferon Regulatory Factor-2/immunology , Kupffer Cells/immunology , Macrophages, Peritoneal/immunology , Repressor Proteins/immunology , STAT1 Transcription Factor/immunology , STAT3 Transcription Factor/immunology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Caspase 3/biosynthesis , Caspase 7/biosynthesis , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/genetics , Down-Regulation/immunology , Gliotoxin/pharmacology , Immunosuppressive Agents/pharmacology , Interferon Regulatory Factor-2/biosynthesis , Interferon Regulatory Factor-2/deficiency , Kupffer Cells/metabolism , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/metabolism , Mice , Mice, Knockout , Promoter Regions, Genetic/immunology , Repressor Proteins/biosynthesis , STAT1 Transcription Factor/biosynthesis , STAT3 Transcription Factor/biosynthesis
16.
J Comp Neurol ; 501(6): 944-58, 2007 Apr 20.
Article in English | MEDLINE | ID: mdl-17311327

ABSTRACT

Sodium/calcium (Na(+)/Ca(2+)) exchangers are membrane transport systems that regulate Ca(2+)-homeostasis in many eukaryotic cells. In olfactory and vomeronasal sensory neurons ligand-induced olfactory signal transduction is associated with influx and elevation of intracellular Ca(2+), [Ca(2+)](i). While much effort has been devoted to the characterization of Ca(2+)-related excitation and adaptation events of olfactory chemosensory neurons (OSNs), much less is known about mechanisms that return [Ca(2+)](i) to the resting state. To identify proteins participating in the poststimulus Ca(2+)-clearance of mouse OSNs, we analyzed the expression of three potassium (K(+))-independent (NCX1, 2, 3) and three K(+)-dependent (NCKX1, 2, 3) Na(+)/Ca(2+) exchangers. In situ hybridization showed that mRNAs of all six Na(+)/Ca(2+) exchangers coexist in neurons of the olfactory and vomeronasal systems, and that some are already detectable in the embryo. Of these, NCX1 and NCKX1 represent the most and least abundant mRNAs, respectively. Moreover, immunohistochemistry revealed that the NCX1, 2, and 3 proteins are expressed in nearly all neurons of the olfactory epithelium, the vomeronasal organ, the septal organ of Masera, and the Grueneberg ganglion. These three exchanger proteins display different expression profiles in dendrites, knobs, and plasma membranes of OSNs and in sustentacular cells. Furthermore, we show that NCX1 mRNA in rat olfactory mucosa is expressed as 8 alternative splice variants. This is the first comprehensive analysis of Na(+)/Ca(2+) exchanger expression in the mammalian olfactory system. Our results suggest that Ca(2+)-extrusion by OSNs utilizes multiple different Na(+)/Ca(2+) exchangers and that different subtypes are targeted to different subcellular compartments.


Subject(s)
Neurons, Afferent/metabolism , Olfactory Pathways/metabolism , Smell/physiology , Sodium-Calcium Exchanger/metabolism , Vomeronasal Organ/metabolism , Age Factors , Animals , Calcium/metabolism , Female , Male , Mice , Neurons, Afferent/cytology , Olfactory Mucosa/cytology , Olfactory Mucosa/metabolism , Olfactory Pathways/cytology , Protein Isoforms/metabolism , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Sodium-Calcium Exchanger/classification , Sodium-Calcium Exchanger/genetics , Vomeronasal Organ/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...