Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 209
Filter
1.
J Virol ; 91(5)2017 03 01.
Article in English | MEDLINE | ID: mdl-28031362

ABSTRACT

Many types of small GTPases are widely expressed in eukaryotes and have different functions. As a crucial member of the Rho GTPase family, Cdc42 serves a number of functions, such as regulating cell growth, migration, and cell movement. Several RNA viruses employ Cdc42-hijacking tactics in their target cell entry processes. However, the function of Cdc42 in shrimp antiviral immunity is not clear. In this study, we identified a Cdc42 protein in the kuruma shrimp (Marsupenaeus japonicus) and named it MjCdc42. MjCdc42 was upregulated in shrimp challenged by white spot syndrome virus (WSSV). The knockdown of MjCdc42 and injection of Cdc42 inhibitors increased the proliferation of WSSV. Further experiments determined that MjCdc42 interacted with an arginine kinase (MjAK). By analyzing the binding activity and enzyme activity of MjAK and its mutant, ΔMjAK, we found that MjAK could enhance the replication of WSSV in shrimp. MjAK interacted with the envelope protein VP26 of WSSV. An inhibitor of AK activity, quercetin, could impair the function of MjAK in WSSV replication. Further study demonstrated that the binding of MjCdc42 and MjAK depends on Cys271 of MjAK and suppresses the WSSV replication-promoting effect of MjAK. By interacting with the active site of MjAK and suppressing its enzyme activity, MjCdc42 inhibits WSSV replication in shrimp. Our results demonstrate a new function of Cdc42 in the cellular defense against viral infection in addition to the regulation of actin and phagocytosis, which has been reported in previous studies. IMPORTANCE The interaction of Cdc42 with arginine kinase plays a crucial role in the host defense against WSSV infection. This study identifies a new mechanism of Cdc42 in innate immunity and enriches the knowledge of the antiviral innate immunity of invertebrates.


Subject(s)
Arginine Kinase/metabolism , Arthropod Proteins/metabolism , Penaeidae/virology , Virus Replication , White spot syndrome virus 1/physiology , cdc42 GTP-Binding Protein/metabolism , Amino Acid Sequence , Animals , Arginine Kinase/chemistry , Arthropod Proteins/chemistry , Conserved Sequence , Enzyme Induction/immunology , Escherichia coli , Host-Pathogen Interactions , Immunity, Innate , Molecular Docking Simulation , Penaeidae/enzymology , Penaeidae/immunology , Protein Binding , Protein Interaction Maps , Up-Regulation , cdc42 GTP-Binding Protein/chemistry
2.
J Biol Chem ; 291(25): 13206-15, 2016 Jun 17.
Article in English | MEDLINE | ID: mdl-27129230

ABSTRACT

Viral infection or lipopolysaccharide (LPS) treatment induces expression of a large array of genes, the products of which play a critical role in host antipathogen immunity and inflammation. We have previously reported that the expression of ubiquitin-specific protease 25 (USP25) is significantly up-regulated after viral infection or LPS treatment, and this is essential for innate immune signaling. However, the mechanism behind this phenomenon is unclear. In this study, we found that viral infection-induced up-regulation of Usp25 is diminished in cells lacking interferon regulatory factor 7 (IRF7) or interferon α receptor 1 (IFNAR1) but not p65. Sendai virus- or type I interferon-induced up-regulation of Usp25 requires de novo protein synthesis of IRF7. Furthermore, IRF7 directly binds to the two conserved IRF binding sites on the USP25 promoter to drive transcription of Usp25, and mutation of these two sites abolished Sendai virus-induced IRF7-mediated activation of the USP25 promoter. Our study has uncovered a previously unknown mechanism by which viral infection or LPS induces up-regulation of USP25.


Subject(s)
Interferon Regulatory Factor-7/physiology , Interferon Type I/physiology , Ubiquitin Thiolesterase/genetics , Animals , Cells, Cultured , Enzyme Induction/immunology , Herpes Simplex/enzymology , Herpesvirus 1, Human/physiology , Lipopolysaccharides/pharmacology , Mice, Knockout , Promoter Regions, Genetic , Protein Binding , Signal Transduction , Transcription, Genetic , Ubiquitin Thiolesterase/metabolism , Up-Regulation/immunology
3.
J Biol Chem ; 290(45): 27158-27167, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26408197

ABSTRACT

Efficient immune responses require regulated antigen presentation to CD4 T cells. IL-10 inhibits the ability of dendritic cells (DCs) and macrophages to stimulate antigen-specific CD4 T cells; however, the mechanisms by which IL-10 suppresses antigen presentation remain poorly understood. We now report that IL-10 stimulates expression of the E3 ubiquitin ligase March-I in activated macrophages, thereby down-regulating MHC-II, CD86, and antigen presentation to CD4 T cells. By contrast, IL-10 does not stimulate March-I expression in DCs, does not suppress MHC-II or CD86 expression on either resting or activated DCs, and does not affect antigen presentation by activated DCs. IL-10 does, however, inhibit the process of DC activation itself, thereby reducing the efficiency of antigen presentation in a March-I-independent manner. Thus, IL-10 suppression of antigen presenting cell function in macrophages is March-I-dependent, whereas in DCs, suppression is March- I-independent.


Subject(s)
Antigen Presentation , Immune Tolerance/physiology , Interleukin-10/immunology , Macrophages/enzymology , Macrophages/immunology , Ubiquitin-Protein Ligases/biosynthesis , Animals , B7-2 Antigen/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/enzymology , Dendritic Cells/immunology , Down-Regulation , Enzyme Induction/immunology , Female , Histocompatibility Antigens Class II/metabolism , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Peptide Fragments/immunology , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
4.
J Leukoc Biol ; 97(2): 379-90, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25420916

ABSTRACT

Borrelia burgdorferi, the bacterial agent of Lyme disease, induces the production of type I IFNs by human DCs through TLR7 and TLR9 signaling. This type I IFN response occurs in a genotype-dependent manner, with significantly higher levels of IFN-α elicited by B. burgdorferi strains that have a greater capacity for causing disseminated infection. A B. burgdorferi strain that was previously shown to induce IFN-α was found to elicit significantly higher levels of IDO1 protein and its downstream metabolite, kynurenine, compared with a B. burgdorferi mutant that lacks a single linear plasmid (lp36); this mutant is unable to induce IFN-α and is severely attenuated for infectivity in mice. Production of IDO by mDC and pDC populations, present within human PBMCs, was concomitant with increased expression of the DC maturation markers, CD83 and CCR7. The defects in IDO production and expression of CD83 and CCR7 could be restored by complementation of the mutant with lp36. Maximal IDO production in response to the wild-type strain was dependent on contributions by both type I IFN and IFN-γ, the type II IFN. Induction of IDO was mediated by the same TLR7-dependent recognition of B. burgdorferi RNA that contributes to the production of type I IFNs by human DCs. The ability of IFN-α-inducing B. burgdorferi strains to stimulate production of IDO and kynurenines may be a mechanism that is used by the pathogen to promote localized immunosuppression and facilitate hematogenous dissemination.


Subject(s)
Borrelia burgdorferi/immunology , Dendritic Cells/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Leukocytes, Mononuclear/immunology , Lyme Disease/immunology , Adult , Aged , Animals , Antigens, CD/immunology , Borrelia burgdorferi/genetics , Dendritic Cells/pathology , Enzyme Induction/immunology , Female , Humans , Immunoglobulins/immunology , Interferon-alpha/immunology , Interferon-gamma/immunology , Leukocytes, Mononuclear/pathology , Lyme Disease/pathology , Male , Membrane Glycoproteins/immunology , Mice , Middle Aged , Mutation , Receptors, CCR7/immunology , Toll-Like Receptor 7/immunology , CD83 Antigen
5.
J Immunol ; 193(1): 335-43, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24879791

ABSTRACT

Response to endotoxins is an important part of the organismal reaction to Gram-negative bacteria and plays a critical role in sepsis and septic shock, as well as other conditions such as metabolic endotoxemia. Humans are generally more sensitive to endotoxins when compared with experimental animals such as mice. Inflammatory caspases mediate endotoxin-induced IL-1ß secretion and lethality in mice, and caspase-4 is an inflammatory caspase that is found in the human, and not mouse, genome. To test whether caspase-4 is involved in endotoxin sensitivity, we developed a transgenic mouse expressing human caspase-4 in its genomic context. Caspase-4 transgenic mice exhibited significantly higher endotoxin sensitivity, as measured by enhanced cytokine secretion and lethality following LPS challenge. Using bone marrow-derived macrophages, we then observed that caspase-4 can support activation of caspase-1 and secretion of IL-1ß and IL-18 in response to priming signals (LPS or Pam3CSK4) alone, without the need for second signals to stimulate the assembly of the inflammasome. These findings indicate that the regulation of caspase-1 activity by human caspase-4 could represent a unique mechanism in humans, as compared with laboratory rodents, and may partially explain the higher sensitivity to endotoxins observed in humans. Regulation of the expression, activation, or activity of caspase-4 therefore represents targets for systemic inflammatory response syndrome, sepsis, septic shock, and related disorders.


Subject(s)
Caspases, Initiator/immunology , Caspases/immunology , Lipopeptides/toxicity , Lipopolysaccharides/toxicity , Macrophages/immunology , Animals , Caspases/genetics , Caspases, Initiator/genetics , Cell Line , Enzyme Induction/drug effects , Enzyme Induction/genetics , Enzyme Induction/immunology , Humans , Inflammasomes/genetics , Inflammasomes/immunology , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Mice , Mice, Knockout
6.
J Immunol ; 192(12): 5481-9, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24808371

ABSTRACT

Eosinophilia and its cellular activation are hallmark features of asthma, as well as other allergic/Th2 disorders, yet there are few, if any, reliable surface markers of eosinophil activation. We have used a FACS-based genome-wide screening system to identify transcriptional alterations in murine lung eosinophils recruited and activated by pulmonary allergen exposure. Using a relatively stringent screen with false-positive correction, we identified 82 candidate genes that could serve as eosinophil activation markers and/or pathogenic effector markers in asthma. Carbonic anhydrase IV (Car4) was a top dysregulated gene with 36-fold induction in allergen-elicited pulmonary eosinophils, which was validated by quantitative PCR, immunohistochemistry, and flow cytometry. Eosinophil CAR4 expression was kinetically regulated by IL-5, but not IL-13. IL-5 was both necessary and sufficient for induction of eosinophil CAR4. Although CAR4-deficient mice did not have a defect in eosinophil recruitment to the lung, nor a change in eosinophil pH-buffering capacity, allergen-challenged chimeric mice that contained Car4(-/-) hematopoietic cells aberrantly expressed a series of genes enriched in biological processes involved in epithelial differentiation, keratinization, and anion exchange. In conclusion, we have determined that eosinophils express CAR4 following IL-5 or allergen exposure, and that CAR4 is involved in regulating the lung transcriptome associated with allergic airway inflammation; therefore, CAR4 has potential value for diagnosing and monitoring eosinophilic responses.


Subject(s)
Asthma/immunology , Carbonic Anhydrase IV/immunology , Eosinophils/immunology , Interleukin-5/immunology , Allergens/genetics , Allergens/immunology , Animals , Asthma/diagnosis , Asthma/genetics , Asthma/metabolism , Asthma/pathology , Carbonic Anhydrase IV/biosynthesis , Carbonic Anhydrase IV/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Enzyme Induction/genetics , Enzyme Induction/immunology , Eosinophils/metabolism , Eosinophils/pathology , Hematopoietic Stem Cells , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-13/metabolism , Interleukin-5/genetics , Interleukin-5/metabolism , Lung/immunology , Lung/metabolism , Lung/pathology , Mice , Mice, Knockout
7.
Dev Comp Immunol ; 46(2): 530-6, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24699445

ABSTRACT

Hydrogen sulfide (H2S) is an important gasotransmitter, which plays indispensable roles in cardiovascular, nervous and immune systems of vertebrates. However, the information about the immunomodulation of H2S in invertebrates is still very limited. In the present study, the temporal expression profile of cystathionine γ lyase in oyster Crassostrea gigas (CgCSE) was investigated after the oysters were stimulated by lipopolysaccharide. The expression levels of CgCSE mRNA transcripts in hemocytes increased significantly at 12h (1.31-fold of the PBS group, P<0.05) after LPS stimulation. The immunomodulation of inducible H2S in oyster was examined by monitoring the alterations of both cellular and humoral immune parameters in response to the stimulations of LPS, LPS+Na2S and LPS+propargylglycine (PAG). The total hemocyte counts (THC) and hemolymph PO activity increased significantly after LPS stimulation, and the increase could be further enhanced by adding PAG, while inhibited by appending Na2S. The phagocytosis activity of hemocytes was also increased firstly after LPS treatment, and the increase was enhanced by adding Na2S but inhibited after appending PAG. The anti-bacterial activity in hemolymph increased at 3h post LPS treatment, and then decreased after adding PAG. The total SOD activity of hemolymph was also elevated at 6h post LPS treatment, and the elevated activity was depressed by adding Na2S. These results collectively indicated that H2S might play crucial roles in the immune response of oyster via modulating the turnover and phagocytosis of hemocytes, and regulating the anti-bacterial activity and proPO activation in the hemolymph.


Subject(s)
Crassostrea/metabolism , Hydrogen Sulfide/metabolism , Animals , Catechol Oxidase/metabolism , Cells, Cultured , Crassostrea/immunology , Cystathionine gamma-Lyase/metabolism , Enzyme Induction/immunology , Enzyme Precursors/metabolism , Escherichia coli/immunology , Hemocytes/enzymology , Hemocytes/immunology , Immunomodulation , Lipopolysaccharides/pharmacology , Phagocytosis , Superoxide Dismutase/metabolism
8.
J Immunol ; 192(11): 5296-304, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24778451

ABSTRACT

High levels of inspired oxygen, hyperoxia, are frequently used in patients with acute respiratory failure. Hyperoxia can exacerbate acute respiratory failure, which has high mortality and no specific therapies. We identified novel roles for PTEN-induced putative kinase 1 (PINK1), a mitochondrial protein, and the cytosolic innate immune protein NLRP3 in the lung and endothelium. We generated double knockouts (PINK1(-/-)/NLRP3(-/-)), as well as cell-targeted PINK1 silencing and lung-targeted overexpression constructs, to specifically show that PINK1 mediates cytoprotection in wild-type and NLRP3(-/-) mice. The ability to resist hyperoxia is proportional to PINK1 expression. PINK1(-/-) mice were the most susceptible; wild-type mice, which induced PINK1 after hyperoxia, had intermediate susceptibility; and NLRP3(-/-) mice, which had high basal and hyperoxia-induced PINK1, were the least susceptible. Genetic deletion of PINK1 or PINK1 silencing in the lung endothelium increased susceptibility to hyperoxia via alterations in autophagy/mitophagy, proteasome activation, apoptosis, and oxidant generation.


Subject(s)
Carrier Proteins/immunology , Endothelium/immunology , Hyperoxia/immunology , Lung/immunology , Oxidants/adverse effects , Protein Kinases/immunology , Animals , Carrier Proteins/genetics , Endothelium/pathology , Enzyme Induction/drug effects , Enzyme Induction/genetics , Enzyme Induction/immunology , Hyperoxia/genetics , Hyperoxia/pathology , Hyperoxia/prevention & control , Lung/pathology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Kinases/genetics
9.
Dev Comp Immunol ; 45(1): 74-86, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24560653

ABSTRACT

Nitric oxide is well known for its roles in immune responses. As such, its synthesizing enzymes have been extensively studied from various species including some teleost fish species. However, the NOS genes have not been characterized in channel catfish (Ictalurus punctatus). In this study, we identified and characterized three NOS genes including one NOS1 and two NOS2 genes in channel catfish. Comparing with the NOS genes from other fish species, the catfish NOS genes are highly conserved in their structural features. Phylogenetic and syntenic analyses allowed determination of NOS1 and NOS2 genes of channel catfish and their orthology relationships. Syntenic analysis, as well as the phylogenetic analysis, indicated that the two NOS2 genes of catfish were lineage-specific duplication. The NOS genes were broadly expressed in most tested tissues, with NOS1 being expressed at the highest levels in the brain, NOS2b1 highly expressed in the skin and gill, and NOS2b2 lowly expressed in most of the tested tissues. The most striking findings of this study was that the expression of the NOS genes are highly regulated after bacterial infection, with time-dependent expression patterns that parallel the migration of macrophages. After Edwardsiella ictaluri challenge, dramatically different responses among the three NOS genes were observed. NOS1 was only significantly in the skin early after infection, while NOS2b1 was rapidly upregulated in gill, but more up-regulated in trunk kidney with the progression of the disease, suggesting such differences in gene expression may be reflective of the migration of macrophages among various tissues of the infected fish. In contrast to NOS1 and NOS2b1, NOS2b2 was normally expressed at very low levels, but it is induced in the brain and liver while significantly down-regulated in most other tissues.


Subject(s)
Enterobacteriaceae Infections/veterinary , Fish Diseases/enzymology , Fish Proteins/genetics , Flavobacteriaceae Infections/veterinary , Ictaluridae/genetics , Nitric Oxide Synthase/genetics , Amino Acid Sequence , Animals , Conserved Sequence , Edwardsiella ictaluri/immunology , Enterobacteriaceae Infections/enzymology , Enterobacteriaceae Infections/immunology , Enzyme Induction/immunology , Fish Diseases/microbiology , Fish Proteins/chemistry , Fish Proteins/metabolism , Flavobacteriaceae Infections/enzymology , Flavobacteriaceae Infections/immunology , Flavobacterium/immunology , Gene Expression/immunology , Ictaluridae/immunology , Ictaluridae/microbiology , Immunity, Innate , Molecular Sequence Data , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase/metabolism , Organ Specificity , Phylogeny , Synteny
10.
Biosci Biotechnol Biochem ; 76(12): 2301-6, 2012.
Article in English | MEDLINE | ID: mdl-23221714

ABSTRACT

Systemic acquired resistance (SAR) is a potent innate immunity system in plants and has been used in rice fields. Development of SAR, involving priming, is achieved by activation of salicylic acid (SA)-mediated pathway. To determine whether heat shock (HS) treatment can induce SAR, we analyzed the effects of HS on Arabidopsis. HS treatment induced disease resistance, expression of SAR marker genes, and SA accumulation in wild-type but not in SA-deficient sid2 and NahG plants, indicating induction of SAR. Time course analysis of the effects of HS indicated that SAR was activated transiently, differently from biological induction, with a peak at 2-3 d after HS, and that it ceased in several days. Production of reactive oxygen species was observed before SA biosynthesis, which might be a trigger for SAR activation. The data presented here suggest that HS can induce SAR, but there exist unknown regulation mechanisms for the maintenance of SAR.


Subject(s)
Arabidopsis/immunology , Arabidopsis/physiology , Heat-Shock Response/immunology , Immunity, Innate , Arabidopsis/enzymology , Arabidopsis/metabolism , Benzoic Acid/metabolism , Disease Resistance/immunology , Enzyme Induction/immunology , Methyltransferases/biosynthesis , Reactive Oxygen Species/metabolism , Salicylic Acid/metabolism
11.
J Immunol ; 189(8): 3848-58, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22962681

ABSTRACT

Hepatic stellate cells (HSCs) are critical for hepatic wound repair and tissue remodeling. They also produce cytokines and chemokines that may contribute to the maintenance of hepatic immune homeostasis and the inherent tolerogenicity of the liver. The functional relationship between HSCs and the professional migratory APCs in the liver, that is, dendritic cells (DCs), has not been evaluated. In this article, we report that murine liver DCs colocalize with HSCs in vivo under normal, steady-state conditions, and cluster with HSCs in vitro. In vitro, HSCs secrete high levels of DC chemoattractants, such as MΙP-1α and MCP-1, as well as cytokines that modulate DC activation, including TNF-α, IL-6, and IL-1ß. Culture of HSCs with conventional liver myeloid (m) DCs resulted in increased IL-6 and IL-10 secretion compared with that of either cell population alone. Coculture also resulted in enhanced expression of costimulatory (CD80, CD86) and coinhibitory (B7-H1) molecules on mDCs. HSC-induced mDC maturation required cell-cell contact and could be blocked, in part, by neutralizing MΙP-1α or MCP-1. HSC-induced mDC maturation was dependent on activation of STAT3 in mDCs and, in part, on HSC-secreted IL-6. Despite upregulation of costimulatory molecules, mDCs conditioned by HSCs demonstrated impaired ability to induce allogeneic T cell proliferation, which was independent of B7-H1, but dependent upon HSC-induced STAT3 activation and subsequent upregulation of IDO. In conclusion, by promoting IDO expression, HSCs may act as potent regulators of liver mDCs and function to maintain hepatic homeostasis and tolerogenicity.


Subject(s)
Dendritic Cells/immunology , Down-Regulation/immunology , Hepatic Stellate Cells/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Liver/immunology , Myeloid Cells/immunology , STAT3 Transcription Factor/physiology , Animals , Cells, Cultured , Coculture Techniques , Enzyme Induction/genetics , Enzyme Induction/immunology , Hepatic Stellate Cells/enzymology , Hepatic Stellate Cells/metabolism , Immunophenotyping , Isoantigens/genetics , Isoantigens/physiology , Liver/cytology , Liver/enzymology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
12.
J Biol Chem ; 287(41): 34801-8, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-22896697

ABSTRACT

Multiple studies have indicated that the TET oxidases and, more controversially, the activation-induced cytidine deaminase/APOBEC deaminases have the capacity to convert genomic DNA 5-methylcytosine (MeC) into altered nucleobases that provoke excision repair and culminate in the replacement of the original MeC with a normal cytosine (C). We show that human APOBEC3A (A3A) efficiently deaminates both MeC to thymine (T) and normal C to uracil (U) in single-stranded DNA substrates. In comparison, the related enzyme APOBEC3G (A3G) has undetectable MeC to T activity and 10-fold less C to U activity. Upon 100-fold induction of endogenous A3A by interferon, the MeC status of bulk chromosomal DNA is unaltered, whereas both MeC and C nucleobases in transfected plasmid DNA substrates are highly susceptible to editing. Knockdown experiments show that endogenous A3A is the source of both of these cellular DNA deaminase activities. This is the first evidence for nonchromosomal DNA MeC to T editing in human cells. These biochemical and cellular data combine to suggest a model in which the expanded substrate versatility of A3A may be an evolutionary adaptation that occurred to fortify its innate immune function in foreign DNA clearance by myeloid lineage cell types.


Subject(s)
5-Methylcytosine/metabolism , Cytidine Deaminase/metabolism , DNA/metabolism , Immunity, Innate , Proteins/metabolism , 5-Methylcytosine/immunology , Cytidine Deaminase/immunology , DNA/immunology , Deamination , Enzyme Induction/drug effects , Enzyme Induction/immunology , HEK293 Cells , Humans , Interferons/immunology , Interferons/pharmacology , Plasmids/immunology , Plasmids/pharmacology , Proteins/immunology , Thymine/immunology , Thymine/metabolism , Uracil/immunology , Uracil/metabolism
13.
Immunol Cell Biol ; 90(8): 831-5, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22430249

ABSTRACT

Interleukin (IL)-27 exerts an anti-inflammatory effect on human and mice CD4(+) T cells by inducing IL-10-producing T regulatory 1 cells through induction of IL-21. However, the role of IL-27 and how it regulates IL-21 from human CD8(+) T cells is unclear. Here, we show that the IL-27 receptor is expressed on human CD8(+) T cells and stimulation of human naïve CD8(+) T cells in the presence of IL-27 leads to an increase in IL-21 and interferon (IFN)-γ production. IL-21 induction in IL-27-stimulated human CD8(+) T cells correlates specifically with expression of the transcription factor T-bet. IL-27 stimulation of naïve CD8(+) T cells induces a double-positive T-bet(+) IL-21(+) expressing CD8(+) T-cell population. Furthermore, IL-27 stimulation of human naïve CD8(+) T cells greatly increases expression of granzyme B. Antibody-mediated neutralization of IL-21 abrogates IL-27-induced granzyme B expression. Moreover, direct addition of IL-21 greatly amplifies granzyme B expression in human naïve CD8(+) T cells. Our findings identify IL-27-induced IL-21 as a key autocrine regulator of granzyme B expression in human CD8(+) T cells.


Subject(s)
Autocrine Communication , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/immunology , Granzymes/biosynthesis , Interleukins/metabolism , Interleukins/pharmacology , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Proliferation , Enzyme Induction/immunology , Humans , Interferon-gamma/metabolism , Lymphocyte Activation/immunology , Mice
14.
J Leukoc Biol ; 91(4): 657-66, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22301793

ABSTRACT

Infection with West Nile virus (WNV) via a mosquito bite results in local viral replication in the skin, followed by viremia. Thus, tissue macrophages are ideally located to prevent the dissemination of WNV throughout the host. The current study shows that WNV infection of human monocyte-derived macrophages (MDM) results in increased WNV mRNA, protein, and infectious virions at 24 h p.i. with a decline in titer after 48 h. Concomitant with viral control was the robust induction of indoleamine 2,3-dioxygenase (IDO) and resultant metabolism of L-tryptophan (L-Trp) to kynurenine. In WNV-exposed cultures, IDO protein was induced primarily in noninfected versus viral-infected MDM. Whereas WNV infection increased the production of IFN-α, IFN-ß, and TNF, only antibody neutralization of TNF attenuated IDO expression and activity. WNV infection also activated NF-κB, and inhibition of this pathway with BMS-345541 abrogated IDO induction. Similar results were also obtained with MDM infected with the related flavivirus, Japanese encephalitis virus. Whereas IDO-mediated L-Trp metabolism can exhibit antiviral properties, inhibition of IDO activity in MDM with L-1-MT or the addition of excess L-Trp did not affect viral control. However, culturing MDM in L-Trp-deficient medium or overexpression of IDO in cells prior to infection significantly attenuated WNV replication, which was reversed by adding excess L-Trp. Together, these data support that although IDO is not required by MDM for the clearance of established viral infection, the spread of flavivirus infection is limited by IDO expressed in uninfected, neighboring cells.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Macrophages/immunology , Monocytes/immunology , NF-kappa B/immunology , West Nile Fever/immunology , West Nile virus/immunology , Animals , Cell Line , Cricetinae , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Enzyme Induction/genetics , Enzyme Induction/immunology , Humans , Imidazoles/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Macrophages/enzymology , Macrophages/virology , Monocytes/metabolism , Monocytes/virology , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , Quinoxalines/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Viral/biosynthesis , RNA, Viral/genetics , RNA, Viral/immunology , Tryptophan/genetics , Tryptophan/immunology , Tryptophan/metabolism , West Nile Fever/genetics , West Nile Fever/metabolism , West Nile virus/metabolism
15.
J Immunol ; 188(1): 417-25, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22131328

ABSTRACT

Eosinophils play important roles in regulation of cellular responses under conditions of homeostasis or infection. Intestinal infection with the parasitic nematode, Trichinella spiralis, induces a pronounced eosinophilia that coincides with establishment of larval stages in skeletal muscle. We have shown previously that in mouse strains in which the eosinophil lineage is ablated, large numbers of T. spiralis larvae are killed by NO, implicating the eosinophil as an immune regulator. In this report, we show that parasite death in eosinophil-ablated mice correlates with reduced recruitment of IL-4(+) T cells and enhanced recruitment of inducible NO synthase (iNOS)-producing neutrophils to infected muscle, as well as increased iNOS in local F4/80(+)CD11b(+)Ly6C(+) macrophages. Actively growing T. spiralis larvae were susceptible to killing by NO in vitro, whereas mature larvae were highly resistant. Growth of larvae was impaired in eosinophil-ablated mice, potentially extending the period of susceptibility to the effects of NO and enhancing parasite clearance. Transfer of eosinophils into eosinophil-ablated ΔdblGATA mice restored larval growth and survival. Regulation of immunity was not dependent upon eosinophil peroxidase or major basic protein 1 and did not correlate with activity of the IDO pathway. Our results suggest that eosinophils support parasite growth and survival by promoting accumulation of Th2 cells and preventing induction of iNOS in macrophages and neutrophils. These findings begin to define the cellular interactions that occur at an extraintestinal site of nematode infection in which the eosinophil functions as a pivotal regulator of immunity.


Subject(s)
Eosinophils/immunology , Macrophages/immunology , Neutrophils/immunology , Nitric Oxide Synthase Type II/immunology , Trichinella spiralis/immunology , Trichinellosis/immunology , Animals , Enzyme Induction/genetics , Enzyme Induction/immunology , Eosinophilia/enzymology , Eosinophilia/immunology , Eosinophilia/parasitology , Eosinophilia/pathology , Eosinophils/enzymology , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-4/metabolism , Larva/growth & development , Larva/immunology , Larva/metabolism , Macrophages/enzymology , Mice , Mice, Knockout , Neutrophils/enzymology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/pathology , Trichinella spiralis/metabolism , Trichinellosis/enzymology , Trichinellosis/genetics , Trichinellosis/pathology
16.
Pharmacol Biochem Behav ; 100(1): 25-32, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21777607

ABSTRACT

Challenging early life events can dramatically affect mental health and wellbeing. Childhood trauma and neglect can increase the risk for developing depressive, anxiety, and substance abuse disorders. Early maternal separation in rodents has been extensively studied and induces long-lasting alterations in affective and stress responses. However, other developmental periods (e.g., the pubertal period) comprise a critical window whereby social and environmental complexity can exert lasting changes on the brain and behavior. In this study, we tested whether early life environmental complexity impacts affective responses, aggressive behaviors, and expression of neuronal nitric oxide synthase (nNOS), the synthetic enzyme for nitric oxide, in adulthood. Mice were weaned into social+nonsocial enrichment, social only enrichment, or standard (isolated) laboratory environments and were tested in open field, elevated plus maze, forced swim, and resident-intruder aggression tests 60 days later. Social+nonsocial enrichment reduced locomotor behavior and anxiety-like responses in the open field and reduced depressive-like responses in the forced swim test. Social housing increased open arm exploration in the elevated plus maze. Both social+nonsocial enrichment and social housing only reduced aggressive behaviors compared with isolation. Social+nonsocial enrichment also increased body mass gain throughout the study. Finally, socially-housed mice had reduced corticosterone concentrations compared with social+nonsocial-enriched mice. Behavioral testing reduced nNOS-positive neurons in the basolateral amygdala and the ventral lateral septum, but not in the social+nonsocial-enriched mice, suggesting that environmental complexity may buffer the brain against some environmental perturbations.


Subject(s)
Motor Activity/immunology , Nitric Oxide Synthase Type I/biosynthesis , Social Behavior , Social Environment , Social Isolation , Weaning , Age Factors , Animals , Enzyme Induction/immunology , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type I/immunology , Nitric Oxide Synthase Type I/metabolism , Social Isolation/psychology
17.
J Immunol ; 186(6): 3632-41, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21296975

ABSTRACT

Helicobacter pylori infection persists for the life of the host due to the failure of the immune response to eradicate the bacterium. Determining how H. pylori escapes the immune response in its gastric niche is clinically important. We have demonstrated in vitro that macrophage NO production can kill H. pylori, but induction of macrophage arginase II (Arg2) inhibits inducible NO synthase (iNOS) translation, causes apoptosis, and restricts bacterial killing. Using a chronic H. pylori infection model, we determined whether Arg2 impairs host defense in vivo. In C57BL/6 mice, expression of Arg2, but not arginase I, was abundant and localized to gastric macrophages. Arg2(-/-) mice had increased histologic gastritis and decreased bacterial colonization compared with wild-type (WT) mice. Increased gastritis scores correlated with decreased colonization in individual Arg2(-/-) mice but not in WT mice. When mice infected with H. pylori were compared, Arg2(-/-) mice had more gastric macrophages, more of these cells were iNOS(+), and these cells expressed higher levels of iNOS protein, as determined by flow cytometry and immunofluorescence microscopy. There was enhanced nitrotyrosine staining in infected Arg2(-/-) versus WT mice, indicating increased NO generation. Infected Arg2(-/-) mice exhibited decreased macrophage apoptosis, as well as enhanced IFN-γ, IL-17a, and IL-12p40 expression, and reduced IL-10 levels consistent with a more vigorous Th1/Th17 response. These studies demonstrate that Arg2 contributes to the immune evasion of H. pylori by limiting macrophage iNOS protein expression and NO production, mediating macrophage apoptosis, and restraining proinflammatory cytokine responses.


Subject(s)
Arginase/biosynthesis , Helicobacter pylori/immunology , Immune Evasion , Macrophages/enzymology , Macrophages/immunology , Animals , Arginase/genetics , Arginase/metabolism , Disease Models, Animal , Enzyme Induction/genetics , Enzyme Induction/immunology , Helicobacter Infections/enzymology , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Intestinal Mucosa/enzymology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/biosynthesis
18.
Mar Biotechnol (NY) ; 13(3): 544-56, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20936320

ABSTRACT

In order to identify genes/proteins involved in copper tolerance, the marine alga Ulva compressa was cultivated with 10 µM copper for 3 days. The activities of antioxidant enzymes ascorbate peroxidase (AP), peroxiredoxin (PRX), thioredoxin (TRX), and glutathione-S-transferase (GST) and the level of lipoperoxides were determined in the alga cultivated with and without copper addition. Antioxidant enzyme activities and lipoperoxides level increased in response to copper excess, indicating that the alga was under oxidative stress. A cDNA library was prepared using U. compressa cultivated with 10 µM copper for 3 days. A total of 3 × 10(4) clones were isolated and 480 clones were sequenced, resulting in 235 non-redundant ESTs, of which 104 encode proteins with known functions. Among them, we identified proteins involved in (1) antioxidant metabolism such as AP, PRX, TRX, GST, and metalothionein (MET), (2) signal transduction, such as calmodulin (CAM), (3) calcium-dependent protein kinase (CDPK) and nucleoside diphosphate kinase (NDK), (4) gene expression, (5) protein synthesis and degradation, and (6) chloroplast and mitochondria electron transport chains. Half of the identified proteins are potentially localized in organelles. The relative level of 18 genes, including those coding for AP, PRX, TRX, GST, MET, CAM, CDPK, and NDK were determined by quantitative RT-PCR in the alga cultivated with 10 µM copper for 0 to 7 days. Transcript levels increased in response to copper stress and most of them reached a maximum at days 3 and 5. Thus, the selected genes are induced by copper stress and they are probably involved in copper acclimation and tolerance.


Subject(s)
Ascorbate Peroxidases/genetics , Copper/toxicity , Glutathione Transferase/genetics , Peroxiredoxins/genetics , Signal Transduction/genetics , Thioredoxins/genetics , Ulva , Analysis of Variance , Ascorbate Peroxidases/metabolism , Base Sequence , Chile , Cloning, Molecular , Electron Transport Chain Complex Proteins/genetics , Enzyme Induction/immunology , Expressed Sequence Tags , Gene Library , Glutathione Transferase/metabolism , Lipid Peroxides/metabolism , Molecular Sequence Data , Oxidative Stress/drug effects , Peroxiredoxins/metabolism , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Thioredoxins/metabolism
19.
J Immunol ; 185(1): 55-63, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20511557

ABSTRACT

Human Vgamma9Vdelta2 T cells, a major innate-like peripheral T cell subset, are thought to play in vivo an important role in innate and adaptive immune responses to infection agents and tumors. However, the mechanisms regulating their broad effector functions, such as cytotoxicity and cytokine responses, remain poorly understood. In this study, we used single-cell calcium video imaging to analyze the early intracellular events associated with TCR-induced Vgamma9Vdelta2 T cell functional responses. When compared with other human T cell subsets, including NKT and Vdelta2(neg) gammadelta T cells, TCR/CD3-activated Vgamma9Vdelta2 T cells displayed an unusually delayed and sustained intracellular calcium mobilization, which was dramatically quickened and shortened on costimulation by NKG2D, a main activating NKR regulating gammadelta T cell tumor cytolysis. Importantly, the protein kinase C transduction pathway was identified as a main regulator of the NKG2D-mediated costimulation of antitumor Vgamma9Vdelta2 cytolytic responses. Therefore, this study identifies a new mechanism regulating Vgamma9Vdelta2 T cell functional plasticity through fine-tuning of early signal transduction events.


Subject(s)
Calcium Signaling/immunology , Cytotoxicity Tests, Immunologic , Isoenzymes/physiology , NK Cell Lectin-Like Receptor Subfamily K/physiology , Natural Killer T-Cells/immunology , Neoplasms, Experimental/immunology , Protein Kinase C/physiology , Receptors, Antigen, T-Cell, gamma-delta/biosynthesis , Receptors, Antigen, T-Cell/physiology , Animals , CD3 Complex/biosynthesis , CD3 Complex/physiology , Cell Communication/immunology , Cell Line, Tumor , Clone Cells , Cytotoxicity Tests, Immunologic/methods , Enzyme Induction/immunology , Humans , Intracellular Fluid/enzymology , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Lymphocyte Activation/immunology , MAP Kinase Signaling System/immunology , Mice , Natural Killer T-Cells/enzymology , Natural Killer T-Cells/metabolism , Neoplasms, Experimental/prevention & control , Protein Kinase C-theta , Receptors, Antigen, T-Cell, gamma-delta/physiology , T-Lymphocyte Subsets/enzymology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
20.
Clin Exp Immunol ; 160(3): 450-6, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20345980

ABSTRACT

Many cellular stresses and inflammatory stimuli can activate p38 mitogen-activated protein kinase (MAPK), a serine/threonine kinase in the MAPK family. The different stimuli act via different receptors or signalling pathways to induce phosphorylation of the cytosolic protein p47(phox), one subunit of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. Formyl-methionyl-leucyl-phenylalanine (fMLP) has been shown to induce the p38 MAPK phosphorylation during the respiratory burst in human neutrophils. Here, we show that treatment with S(+)-ketamine or R(-)-ketamine at different concentrations (50, 100, 200, 400 microM) reduced fMLP-induced superoxide anion generation and p47(phox) phosphorylation in neutrophils in a concentration-dependent manner (y = -0.093x + 93.35 for S(+)-ketamine and y = -0.0982x + 95.603 for R(-)-ketamine, respectively). While treatment with 50 microM ketamine inhibited fMLP-induced superoxide generation by 10%, treatment with 400 microM S(+)-ketamine and R(-)-ketamine reduced fMLP-induced superoxide generation to 60.5 +/- 8.3% and 60.0 +/- 8.5%, respectively, compared with that in neutrophils treated with fMLP alone. Furthermore, treatment with ketamine down-regulated both fMLP-induced p47(phox) and isoproterenol-induced p38 MAPK phosphorylation and superoxide production. Interestingly, treatment with SB203580, the p38 MAPK inhibitor, also mitigated fMLP-induced superoxide anion generation and p38 MAPK and p47(phox) phosphorylation as well as apoptosis in a concentration-dependent fashion in neutrophils. Therefore, ketamine racemes inhibited fMLP-induced superoxide anion generation and p47(phox) phosphorylation by modulating fMLP-mediated p38 MAPK activation in neutrophils.


Subject(s)
Analgesics/pharmacology , Ketamine/pharmacology , MAP Kinase Signaling System/drug effects , NADPH Oxidases/immunology , Superoxides/immunology , p38 Mitogen-Activated Protein Kinases/immunology , Apoptosis/drug effects , Apoptosis/immunology , Cardiotonic Agents/pharmacology , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/immunology , Enzyme Induction/drug effects , Enzyme Induction/immunology , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/pharmacology , Isoproterenol/pharmacology , MAP Kinase Signaling System/immunology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , NADPH Oxidases/biosynthesis , Neutrophils , Phosphorylation/drug effects , Phosphorylation/immunology , Pyridines/pharmacology , Superoxides/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...