Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Cell ; 163(2): 367-80, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26411289

ABSTRACT

Intestinal Th17 cells are induced and accumulate in response to colonization with a subgroup of intestinal microbes such as segmented filamentous bacteria (SFB) and certain extracellular pathogens. Here, we show that adhesion of microbes to intestinal epithelial cells (ECs) is a critical cue for Th17 induction. Upon monocolonization of germ-free mice or rats with SFB indigenous to mice (M-SFB) or rats (R-SFB), M-SFB and R-SFB showed host-specific adhesion to small intestinal ECs, accompanied by host-specific induction of Th17 cells. Citrobacter rodentium and Escherichia coli O157 triggered similar Th17 responses, whereas adhesion-defective mutants of these microbes failed to do so. Moreover, a mixture of 20 bacterial strains, which were selected and isolated from fecal samples of a patient with ulcerative colitis on the basis of their ability to cause a robust induction of Th17 cells in the mouse colon, also exhibited EC-adhesive characteristics.


Subject(s)
Bacterial Adhesion , Citrobacter rodentium/physiology , Enterobacteriaceae Infections/immunology , Escherichia coli Infections/immunology , Escherichia coli O157/physiology , Intestinal Mucosa/immunology , Th17 Cells/immunology , Animals , Bacterial Infections/immunology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelial Cells/ultrastructure , Feces/microbiology , Humans , Immunoglobulin A/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Microscopy, Electron, Scanning , Rats , Rats, Inbred F344 , Species Specificity
2.
J Biol Chem ; 298(2): 101579, 2022 02.
Article in English | MEDLINE | ID: mdl-35031323

ABSTRACT

Rho family small GTPases (Rho) regulate various cell motility processes by spatiotemporally controlling the actin cytoskeleton. Some Rho-specific guanine nucleotide exchange factors (RhoGEFs) are regulated via tyrosine phosphorylation by Src family tyrosine kinase (SFK). We also previously reported that PLEKHG2, a RhoGEF for the GTPases Rac1 and Cdc42, is tyrosine-phosphorylated by SRC. However, the details of the mechanisms by which SFK regulates RhoGEFs are not well understood. In this study, we found for the first time that PLEKHG1, which has very high homology to the Dbl and pleckstrin homology domains of PLEKHG2, activates Cdc42 following activation by FYN, a member of the SFK family. We also show that this activation of PLEKHG1 by FYN requires interaction between these two proteins and FYN-induced tyrosine phosphorylation of PLEKHG1. We also found that the region containing the Src homology 3 and Src homology 2 domains of FYN is required for this interaction. Finally, we demonstrated that tyrosine phosphorylation of Tyr-720 and Tyr-801 in PLEKHG1 is important for the activation of PLEKHG1. These results suggest that FYN is a regulator of PLEKHG1 and may regulate cell morphology through Rho signaling via the interaction with and tyrosine phosphorylation of PLEKHG1.


Subject(s)
Rho Guanine Nucleotide Exchange Factors , rho GTP-Binding Proteins , src-Family Kinases , Phosphorylation , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Tyrosine/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , src-Family Kinases/genetics , src-Family Kinases/metabolism
3.
Biol Pharm Bull ; 43(12): 1911-1916, 2020.
Article in English | MEDLINE | ID: mdl-33268709

ABSTRACT

Royal jelly (RJ) is known as an important functional foodstuff that promotes several health benefits and contains various bioactive substances, including major royal jelly proteins (MRJPs). Among the MRJPs, MRJP3 possesses both cell proliferation and wound healing effects. As the carboxyl domain of MRJP3 contains tandem penta-peptide repeat (TPR) sequences unique to MRJP3 among the MRJPs, we purified the TPRs as glutathione-S-transferase (GST)-fusion proteins and demonstrated their dose-dependent effects on THP-1 and Vero cell proliferation. The GST-TPR protein with 19 repeats (GST-TPR19) showed cell proliferative activity equivalent to MRJP3 and higher than GST-TPR6. GST-TPR19 also exhibited wound healing activity at a level similar to MRJP3. Digestion of GST-TPR19 with trypsin had no effect on its cell proliferative activity, suggesting that the main digested products; i.e., penta-peptides (Q-N-x-N-[K/R]), maintain the cell proliferative ability of MRJP3. In conclusion, the TPRs of MRJP3 are critical to the beneficial effect(s) of RJ.


Subject(s)
Cell Proliferation/drug effects , Fatty Acids/chemistry , Fatty Acids/genetics , Oligopeptides/chemistry , Oligopeptides/genetics , Sequence Analysis, Protein/methods , Animals , Cell Proliferation/physiology , Chlorocebus aethiops , Fatty Acids/pharmacology , Humans , Oligopeptides/pharmacology , THP-1 Cells , Vero Cells
4.
Biol Pharm Bull ; 41(2): 190-197, 2018.
Article in English | MEDLINE | ID: mdl-29386479

ABSTRACT

Application of food-grade Lactococcus lactis (L. lactis) as a safe delivery tool for DNA vaccines and therapeutic proteins has been well investigated. Although some studies showed that eukaryotic expression plasmids were transferred from L. lactis to enterocytes, the precise mechanism of the DNA transfer remains unknown. In this study, we generated an invasive L. lactis strain that expresses "murinized" Internalin A, an invasin of intracellular bacteria Listeria monocytogenes with two amino acid alterations for invasion into murine cells, and confirmed that this L. lactis strain delivered DNA in an invasin-dependent manner into a monolayer of epithelial cells polarized to mimic the gastrointestinal tract environment. Although invasive L. lactis inoculated orally can deliver DNA into enterocytes in the gastrointestinal tract of mice, the efficiency of DNA transfer was similar to that of non-invasive L. lactis strain, suggesting that the in vivo DNA transfer from L. lactis occurs invasin-independently. A ligated-intestinal loop assay, a method for a short-term culturing of the whole intestine filled with materials to evaluate the interaction of the materials with intestinal cells, demonstrated that both non-invasive and invasive L. lactis strains were present in the Peyer's patches of the small intestine. On the other hand, few L. lactis was detected in the non-Peyer's patch epithelial region. Thus, our observations lead us to speculate that DNA transfer from L. lactis occurs predominantly in the Peyer's patches in an invasin-independent manner.


Subject(s)
Bacterial Proteins/metabolism , DNA, Recombinant/metabolism , Drug Delivery Systems , Lactococcus lactis/physiology , Microorganisms, Genetically-Modified/physiology , Peyer's Patches/metabolism , Vaccines, DNA/metabolism , Administration, Oral , Animals , Bacterial Proteins/administration & dosage , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Translocation , Biological Transport , Caco-2 Cells , Cell Line , Cell Polarity , DNA, Recombinant/administration & dosage , Female , Food Microbiology , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestine, Small/cytology , Intestine, Small/metabolism , Intestine, Small/microbiology , Lactococcus lactis/cytology , Lactococcus lactis/genetics , Listeria monocytogenes/cytology , Listeria monocytogenes/genetics , Listeria monocytogenes/physiology , Mice , Mice, Inbred C57BL , Microorganisms, Genetically-Modified/cytology , Microorganisms, Genetically-Modified/genetics , Peyer's Patches/cytology , Peyer's Patches/microbiology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/metabolism , Vaccines, DNA/administration & dosage
5.
J Biol Chem ; 289(14): 10045-56, 2014 Apr 04.
Article in English | MEDLINE | ID: mdl-24554703

ABSTRACT

FLJ00018/PLEKHG2 is a guanine nucleotide exchange factor for the small GTPases Rac and Cdc42 and has been shown to mediate the signaling pathways leading to actin cytoskeleton reorganization. The function of FLJ00018 is regulated by the interaction of heterotrimeric GTP-binding protein Gßγ subunits or cytosolic actin. However, the details underlying the molecular mechanisms of FLJ00018 activation have yet to be elucidated. In the present study we show that FLJ00018 is phosphorylated and activated by ß1-adrenergic receptor stimulation-induced EGF receptor (EGFR) transactivation in addition to Gßγ signaling. FLJ00018 is also phosphorylated and activated by direct EGFR stimulation. The phosphorylation of FLJ00018 by EGFR stimulation is mediated by the Ras/mitogen-activated protein kinase (MAPK) pathway. Through deletion and site-directed mutagenesis studies, we have identified Thr-680 as the major site of phosphorylation by EGFR stimulation. FLJ00018 T680A, in which the phosphorylation site is replaced by alanine, showed a limited response of the Neuro-2a cell morphology to EGF stimulation. Our results provide evidence that stimulation of the Ras/MAPK pathway by EGFR results in FLJ00018 phosphorylation at Thr-680, which in turn controls changes in cell shape.


Subject(s)
ErbB Receptors/metabolism , Guanine Nucleotide Exchange Factors/metabolism , MAP Kinase Signaling System/physiology , Transcriptional Activation/physiology , Amino Acid Substitution , Animals , Cell Shape , ErbB Receptors/genetics , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , Mice , Mutagenesis, Site-Directed , Mutation, Missense , NIH 3T3 Cells , Phosphorylation
6.
Immunopharmacol Immunotoxicol ; 35(1): 1-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22856509

ABSTRACT

The extracts prepared from green algae are reported to possess a variety of biological activities including antioxidant, antitumor and antiviral activities. The acidic polysaccharide fraction from a green alga Coccomyxa gloeobotrydiformi (CmAPS) was isolated and the antiviral action on an in vitro infection of influenza A virus was examined. CmAPS inhibited the growth and yield of all influenza A virus strains tested, such as A/H1N1, A/H2N2, A/H3N2 and A/H1N1 pandemic strains. The 50% inhibitory concentration of CmAPS on the infection of human influenza A virus strains ranged from 26 to 70 µg/mL and the antiviral activity of CmAPS against influenza A/USSR90/77 (H1N1) was the strongest. The antiviral activity of CmAPS was not due to the cytotoxicity against host cells. The antiviral activity of CmAPS required its presence in the inoculation of virus onto MDCK cells. Pretreatment and post-treatment with CmAPS was ineffective for the antiviral activity. CmAPS inhibited influenza A virus-induced erythrocyte hemagglutination and hemolysis. Taken together, CmAPS was suggested to exhibit the anti-influenza virus activity through preventing the interaction of virus and host cells. The detailed antiviral activity of CmAPS is discussed.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Chlorophyta/chemistry , Influenza A virus/drug effects , Influenza, Human/virology , Plant Extracts/pharmacology , Polysaccharides/chemistry , Polysaccharides/pharmacology , Animals , Cells, Cultured , Chickens , Dogs , Erythrocytes/drug effects , Hemagglutination/drug effects , Hemolysis/drug effects , Host-Pathogen Interactions/drug effects , Humans , Influenza, Human/drug therapy , Madin Darby Canine Kidney Cells , Plant Extracts/chemistry , Virus Replication/drug effects
7.
Biochem Biophys Res Commun ; 417(4): 1127-32, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22206674

ABSTRACT

The glial cells missing a (GCMa) transcription factor plays a pivotal role in the placental development by regulating the expression of several genes in the placenta that are responsible for the proper formation of the syncytiotrophoblast. It is well known that the function of GCMa is regulated at both transcriptional and post-translational levels by the cyclic AMP (cAMP)/protein kinase A (PKA)-dependent pathway, the activation of which increases the GCMa protein level and leads to trophoblast differentiation into the syncytiotrophoblast. However, little is known about the regulatory control of GCMa by PKC-dependent signaling mechanism(s). To investigate whether GCMa is regulated by PKC-dependent pathway, we treated the human choriocarcinoma JEG-3 cells with phorbol 12-myristate 13-acetate (PMA) and studied its effect on the GCMa protein using a monoclonal anti-GCMa antibody we prepared. PMA caused a transient decrease in the endogenous GCMa protein level in JEG-3 cells that was accompanied by an increase in GCMa phosphorylation. The phosphorylation and degradation of GCMa by PMA treatment was effectively reduced by pretreatment with protein kinase C (PKC) inhibitors and a mitogen-activated protein kinase (MAPK) kinase (MEK) inhibitor, indicating a PKC- and MEK-dependent mechanism. Furthermore, we identified the serine residues 328, 378 and 383 to be the phosphorylation sites on GCMa that are involved in the PMA-induced degradation of GCMa. Our data demonstrate for the first time that GCMa is phosphorylated by the PKC- and MEK/extracellular signal-regulated kinase (ERK)-dependent mechanism, and that this phosphorylation is involved in its degradation process.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Nuclear Proteins/metabolism , Protein Kinase C/metabolism , Tetradecanoylphorbol Acetate/metabolism , Transcription Factors/metabolism , Animals , Antibodies, Monoclonal , DNA-Binding Proteins , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , HEK293 Cells , Humans , Hybridomas , MAP Kinase Signaling System , Mice , Neuropeptides/genetics , Neuropeptides/immunology , Neuropeptides/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Stability , Serine/genetics , Serine/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factors/genetics , Transcription Factors/immunology
8.
J Virol ; 85(9): 4606-11, 2011 May.
Article in English | MEDLINE | ID: mdl-21345944

ABSTRACT

Plasmacytoid dendritic cells (pDCs) do not produce alpha interferon (IFN-α) unless viruses cause a systemic infection or overcome the first-line defense provided by conventional DCs and macrophages. We show here that even paramyxoviruses, whose infections are restricted to the respiratory tract, have a V protein able to prevent Toll-like receptor 7 (TLR7)- and TLR9-dependent IFN-α induction specific to pDCs. Mutational analysis of human parainfluenza virus type 2 demonstrates that the second Trp residue of the Trp-rich motif (Trp-X(3)-Trp-X(9)-Trp) in the C-terminal domain unique to V, a determinant for IRF7 binding, is critical for the blockade of TLR7/9-dependent signaling.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/virology , Parainfluenza Virus 2, Human/pathogenicity , Signal Transduction , Toll-Like Receptor 7/antagonists & inhibitors , Toll-Like Receptor 9/antagonists & inhibitors , Viral Proteins/metabolism , Humans
9.
Cell Immunol ; 273(1): 73-8, 2012.
Article in English | MEDLINE | ID: mdl-22177846

ABSTRACT

10-Hydroxy-trans-2-decenoic acid (10H2DA) is a major lipid component of royal jelly, a honey bee secretion used to nourish the queen bee and young larvae. In this study, we examined the effect of 10H2DA on interferon (IFN)-γ-induced nitric oxide (NO) production. IFN-γ-induced NO production and activation of the inducible NO synthase promoter were significantly inhibited by 10H2DA. IFN-γ-induced phosphorylation of signal transducer and activator of transcription-1 was not affected by 10H2DA. In contrast, IFN-γ-induced tumor necrosis factor (TNF)-α production and nuclear factor (NF)-κB activation were inhibited by 10H2DA. IFN-γ-mediated induction of interferon regulatory factor (IRF)-8, but not IRF-1, was also inhibited by 10H2DA. IFN-γ-induced TNF-α production followed by activation of NF-κB is known to be essential for NO production. Together, 10H2DA inhibited IFN-γ-induced NO production by inhibiting IRF-8 induction and TNF-α production. 10H2DA might modulate IFN-γ-mediated cellular responses by inhibiting the induction of IRF-8 and IRF-8-dependent genes.


Subject(s)
Fatty Acids, Monounsaturated/pharmacology , Interferon Regulatory Factors/antagonists & inhibitors , Interferon-gamma/antagonists & inhibitors , Macrophages/drug effects , Nitric Oxide/biosynthesis , Animals , Cell Line , Enzyme Activation/immunology , Fatty Acids/pharmacology , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Interferon-gamma/immunology , Macrophages/enzymology , Macrophages/immunology , Mice , NF-kappa B/immunology , Nitric Oxide/antagonists & inhibitors , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/immunology , Transcription, Genetic/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
10.
Biol Pharm Bull ; 35(6): 917-23, 2012.
Article in English | MEDLINE | ID: mdl-22687484

ABSTRACT

Previously, we have shown that chickens immunized with Shiga toxin (Stx) produce Stx-neutralizing egg yolk immunoglobulin Y (IgY) antibody. The anti-Stx-1 IgY and anti-Stx-2 IgY exert their neutralizing activity through their antibody activity against the B subunit of the toxin but not the A subunit. In the present study, chickens were immunized with recombinant Stx-1 B subunit (rStx-1B) and recombinant Stx-2 B subunit (rStx-2B). Induced anti-rStx-1B and anti-rStx-2B IgY neutralized the toxicity of Stx-1 and Stx-2 against HeLa 229 cells. The neutralizing activity of anti-rStx-1B IgY on Stx-1 was almost 10 times stronger than that of anti-Stx-1 IgY, and that of anti-rStx-2B IgY was 2.6 times stronger than that of anti-Stx-2 IgY. Anti-rStx-1B and anti-rStx-2B IgY reacted with multimeric and monomeric forms of the B subunits in contrast to anti-Stx-1 and anti-Stx-2 IgY that reacted with only the multimeric form. These results indicated that recombinant B subunits were promising antigens for induction of neutralizing antibodies in chickens.


Subject(s)
Antibodies, Neutralizing/immunology , Egg Yolk/immunology , Immunoglobulins/immunology , Shiga Toxin/immunology , Animals , Chickens , Enzyme-Linked Immunosorbent Assay , Immunization , Protein Subunits/immunology , Recombinant Proteins/immunology
11.
Immunohorizons ; 5(10): 870-883, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34702763

ABSTRACT

Citrobacter rodentium is a murine pathogenic bacterium that adheres to intestinal epithelial cells, resulting in loss of microvilli and pedestal formation, and alters multiple cellular processes, including actin dynamics. Translocated intimin receptor (Tir), one of its virulence factors, functions as receptor for intimin, a bacterial adhesin, thereby mediating bacterial adhesion to epithelial cells. Although robust immune responses are induced to eliminate pathogenic bacteria in the host, they are suppressed against harmless commensal bacteria. The mechanism(s) underlying such a differentiation remains unclear. This study sought to determine the roles of intimate adhesion in the induction of specific immune responses upon C. rodentium infection. To this end, microbiota-depleted mice were infected with the Tir-F strain expressing full-length Tir or mutant strains expressing the C-terminal truncated Tir that is defective in intimin binding and host cell actin polymerization. There were no differences in the colonization kinetics and Abs responses against C. rodentium LPS among the strains, whereas Abs against the virulence factors were only produced on Tir-F infection. Although there were no differences in the virulence factors mRNA expression levels, colonic hyperplasia, and bacterial translocation to the systemic organs irrespective of the strain, adhesion to colonic epithelial cells was reduced in the mutant strain-infected mice. Furthermore, transcriptomic analysis indicated that robust inflammatory and immune responses were only induced in the Tir-F-infected group and were suppressed in the mutant-infected groups. Taken together, these findings suggest that Tir-mediated intimate adhesion induces inflammatory and immune responses, resulting in the induction of virulence factor-specific Abs.


Subject(s)
Bacterial Adhesion/immunology , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , Intestinal Mucosa/pathology , Virulence Factors/metabolism , Adhesins, Bacterial/metabolism , Animals , Bacterial Adhesion/genetics , Cell Line, Tumor , Citrobacter rodentium/genetics , Citrobacter rodentium/pathogenicity , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/pathology , Female , Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Mice , Mutation , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Specific Pathogen-Free Organisms
12.
Cell Signal ; 61: 93-107, 2019 09.
Article in English | MEDLINE | ID: mdl-31100317

ABSTRACT

The Rho family small GTPases mediate cell responses through actin cytoskeletal rearrangement. We previously reported that PLEKHG2, a Rho-specific guanine nucleotide exchange factor, is regulated via interaction with several proteins. We found that PLEKHG2 interacted with non-receptor tyrosine kinase ABL1, but the cellular function remains unclear. Here, we show that the interaction between PLEKHG2 and ABL1 attenuated the PLEKHG2-induced serum response element-dependent gene transcription in a tyrosine phosphorylation-independent manner. PLEKHG2 and ABL1 were co-localized and accumulated within cells co-expressing PLEKHG2 and ABL1. The cellular fractionation analysis suggested that the accumulation involved actin cytoskeletal reorganization. We also revealed that the co-expression of PLEKHG2 with ABL1, but not BCR-ABL, suppressed cell growth and synergistically enhanced NF-κB-dependent gene transcription. The cell growth suppression was canceled by co-expression with IκBα, a member of the NF-κB inhibitor protein family. This study suggests that the interaction between PLEKHG2 and ABL1 suppresses cell growth through intracellular protein accumulation via the NF-κB signaling pathway.


Subject(s)
Cell Proliferation/genetics , Guanine Nucleotide Exchange Factors/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Signal Transduction/genetics , Actin Cytoskeleton/metabolism , Actins/metabolism , Fusion Proteins, bcr-abl/metabolism , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , NF-KappaB Inhibitor alpha/metabolism , Phosphorylation/genetics , Protein Aggregates/genetics , Protein Binding/genetics , Proto-Oncogene Proteins c-abl/genetics , Serum Response Element/genetics , Transcription, Genetic/genetics , Transfection
13.
J Microbiol Methods ; 159: 62-68, 2019 04.
Article in English | MEDLINE | ID: mdl-30817947

ABSTRACT

Evaluation of the efficacy of vaccine candidates that prevent enteropathogenic and enterohemorrhagic Escherichia coli (EPEC/EHEC) infection in mouse models is difficult due to their limited pathogenicity in mice. Citrobacter rodentium, a murine pathogenic bacterium that shares its infection strategy and virulence genes with EPEC/EHEC, has been used as a model pathogen to develop novel vaccine strategies or platforms for these bacteria. However, there are few reports on the comparative effectiveness of novel vaccine platforms as no C. rodentium vaccines have yet been prepared by standard methods such as bacteria attenuation or inactivation. In this study, we investigated the protective effect of the oral administration of formalin-inactivated C. rodentium (Fo-CR) on C. rodentium infection in two mouse strains, C57BL/6 and C3H/HeN, as these strains have different degrees of susceptibility to infection. In C57BL/6 mice, administration of Fo-CR induced significant C. rodentium-specific mucosal and systemic antibody responses, promoted bacterial clearance from the gut and inhibited colonic hyperplasia. Furthermore, in C3H/HeN mice, the administration followed by lethal C. rodentium infection induced significantly high avidity serum IgG specific to C. rodentium and inhibited death, body weight loss, and bacterial invasion to visceral organs. In conclusion, the oral administration of Fo-CR resulted in the protection of mice from C. rodentium infection, indicating that it serves as a reference method for evaluating the efficacy of novel oral vaccine candidates or platforms.


Subject(s)
Bacterial Vaccines/administration & dosage , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/prevention & control , Administration, Oral , Animals , Antibodies, Bacterial/immunology , Bacterial Vaccines/immunology , Citrobacter rodentium/genetics , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/microbiology , Female , Humans , Immunization , Immunoglobulin G/immunology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
14.
FEMS Immunol Med Microbiol ; 53(3): 306-13, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18625009

ABSTRACT

Tumor necrosis factor-alpha (TNF-alpha) is important for the induction of systemic inflammatory responses that lead to lethal shock. Quercetin and luteolin, which differ by one hydroxyl group, are known to suppress the lipopolysaccharide-induced production of TNF-alpha in vitro. We show differing inhibitory effects of quercetin and luteolin on the induction of lethal shock in Salmonella typhimurium aroA-infected mice. In a time- and dose-dependent manner, quercetin reduced the plasma levels of TNF-alpha, lowered bacterial titers in livers, prevented liver damage and prolonged survival, while luteolin had little or no effect. Compared with luteolin, quercetin increased the infiltration of Gr-1(+)CD69(+) neutrophils into the peritoneal cavity and lowered heat shock protein 70 expression. Obviously, the additional hydroxyl group in quercetin is important for suppressing infection-induced lethal shock in mice.


Subject(s)
Immunologic Factors/pharmacology , Luteolin/pharmacology , Quercetin/pharmacology , Salmonella Infections, Animal/complications , Salmonella typhimurium/isolation & purification , Shock/prevention & control , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Colony Count, Microbial , Dose-Response Relationship, Drug , Female , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/biosynthesis , Liver/microbiology , Liver/pathology , Mice , Molecular Structure , Neutrophils/immunology , Peritoneal Cavity/cytology , Salmonella Infections, Animal/microbiology , Survival Analysis , Tumor Necrosis Factor-alpha/blood
15.
Immunopharmacol Immunotoxicol ; 30(4): 867-82, 2008.
Article in English | MEDLINE | ID: mdl-18720166

ABSTRACT

Quercetin (QUER) and luteolin (LUTE) are dietary flavonoids capable of regulating the production of cytokines, such as tumor necrosis factor-alpha (TNF-alpha), and interleukin-6 (IL-6). However, their mechanisms of action are not fully understood. In lipopolysaccharide-triggered (LPS)-triggered signaling via Toll-like receptor 4 (TLR4), QUER and LUTE suppresses not only the degradation of the inhibitor of kappaB (IkappaB), with resultant activation of nuclear factor-kappaB (NF-kappaB), but also the phosphorylation of p38 and Akt in bone marrow-derived macrophages that have been stimulated with LPS. We report here that, in TNF-alpha-induced signaling, QUER and LUTE significantly suppressed the production of IL-6 and activation of NF-kappaB. Accumulation of lipid rafts, the initial step in the signaling pathway, was significantly inhibited when macrophages were treated with QUER or with LUTE prior to exposure to LPS. Similarly, the accumulation of lipid rafts was inhibited by the flavonoids when B cells were activated via the membrane IgM and when T cells were activated via CD3. In contrast, QUER and LUTE did not inhibit the activation of phorbol myristate acetate-induced NF-kappaB in macrophages. Our observations suggest that QUER and LUTE interact with receptors on the cell surface and suppress the accumulation of lipid rafts that occurs downstream of the activation of the receptors.


Subject(s)
Immunosuppressive Agents/pharmacology , Luteolin/pharmacology , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Quercetin/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 4/physiology , Animals , Cell Line , Dose-Response Relationship, Immunologic , Flavonoids/pharmacology , Humans , Membrane Microdomains/immunology , Mice , Mice, Inbred C57BL , Signal Transduction/immunology
16.
J Endotoxin Res ; 12(6): 346-51, 2006.
Article in English | MEDLINE | ID: mdl-17254388

ABSTRACT

Previously, we found that mouse TH2.52 cells possess the characteristic of CD5(+) B1 cells and proliferate in response to lipopolysaccharide (LPS). The effect of LPS on cytokine production by TH2.52 B1 cells was studied. TH2.52 cells constitutively produced a small amount of tumor necrosis factor (TNF)-alpha and interleukin (IL)-6, and TNF-alpha and IL-6 production was markedly enhanced by LPS stimulation. Although interferon (IFN)-gamma caused the production of various cytokines, such as IL-2, IL-4, IL-6 and TNF-alpha in TH2.52 cells, LPS did not cause the production of such cytokines. LPS did not induce IFN-beta production in TH2.52 cells and TH2.52 cells lacked the expression of several molecules participating in the MyD88-independent pathway in LPS signaling. Defective responsiveness of TH2.52 B1 cells to LPS in cytokine production might be responsible for the failure of IFN-beta production due to the lack of molecules participating in the MyD88-independent pathway.


Subject(s)
Antigens, CD/immunology , B-Lymphocytes/immunology , CD5 Antigens/immunology , Lipopolysaccharides/toxicity , Animals , Cell Division/drug effects , Cell Line , Cytokines/biosynthesis , Mice , Spleen/immunology
17.
FEMS Immunol Med Microbiol ; 43(2): 277-86, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15681159

ABSTRACT

The effect of inhibition of mitogen and stress-activated protein kinases 1/2 (MSK1/2) on lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophage cells was investigated. Pretreatment with Ro 31-8220, an inhibitor of MSK1/2, induced cell death in LPS-stimulated RAW 264.7 cells. In contrast, calphostin C, another inhibitor of protein kinase C, did not cause cell death. Cell death was not mediated by the release of pro-inflammatory mediators from LPS-stimulated RAW 264.7 cells. Cell death was accompanied by DNA fragmentation and annexin V binding, suggesting apoptotic cell death. Further, several caspase inhibitors did not prevent LPS-induced cell death of Ro 31-8220-pretreated RAW 264.7 cells. Nuclear translocation of apoptosis-inducing factor (AIF) was detected in Ro 31-8220-pretreated cells after LPS stimulation. Cell death was due to mitochondrial damage. Ro 31-8220 exclusively inhibited the phosphorylation of cAMP-responsive element binding protein (CREB), a substrate of MSK1/2. RAW 264.7 cells transfected with the dominant-negative MSK1 clones underwent cell death in response to LPS. Hence, it was suggested that MSK1/2 might play a critical role in the survival of LPS-stimulated RAW 264.7 cells.


Subject(s)
Apoptosis , Lipopolysaccharides/immunology , Macrophages/cytology , Macrophages/immunology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Animals , Annexin A5/metabolism , Apoptosis Inducing Factor , Caspase Inhibitors , Cell Line , Cell Nucleus/chemistry , Cell Survival , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , DNA Fragmentation , Enzyme Inhibitors/pharmacology , Flavoproteins/metabolism , Indoles/pharmacology , Macrophages/metabolism , Membrane Proteins/metabolism , Mice , Mitochondria/metabolism , Mitochondria/ultrastructure , Naphthalenes/pharmacology
18.
FEMS Immunol Med Microbiol ; 45(2): 213-9, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16051071

ABSTRACT

The present study was conducted to determine effects of U0126, a specific inhibitor of mitogen-activated kinase kinase 1/2, on production of nitric oxide (NO) in RAW264.7 macrophage cells. U0126 significantly enhanced NO production in lipopolysaccharide (LPS) but not CpG DNA or interferon-gamma-stimulated RAW264.7 cells. In contrast, U0124, a negative control for U0126, did not affect LPS-induced NO production. Further, a series of inhibitors of p38, phosphatidyl-inositol 3-kinase and Janus tyrosine kinase rather caused suppression in LPS-stimulated RAW264.7 cells. U0126 was found to definitely inhibit phosphorylation of extracellular signal-regulated kinase (Erk) 1/2 and augment the levels of inducible type of NO synthase. Antisense oligonucleotides of Erk1/2 also augmented LPS-induced NO production. Inactivation of Erk1/2 by U0126 furthermore inhibited LPS-induced activating protein-1 activation, but not nuclear factor-kappaB activation. The results suggest that Erk1/2 might negatively regulate NO production in LPS-stimulated RAW264.7 cells.


Subject(s)
MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Macrophages/metabolism , Nitric Oxide/biosynthesis , Animals , Base Sequence , Butadienes/pharmacology , Cell Line , Cytokines/biosynthesis , DNA/genetics , Enzyme Inhibitors/pharmacology , Interferon-beta/genetics , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Mice , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Nitriles/pharmacology , Oligodeoxyribonucleotides/pharmacology , Oligonucleotides, Antisense/pharmacology , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins , Tumor Necrosis Factor-alpha/genetics
19.
Clin Rheumatol ; 24(1): 11-3, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15674653

ABSTRACT

The polymorphism of the interleukin-10 (IL-10) promoter and tumor necrosis factor receptor II (TNFRII) in Vietnamese patients with systemic lupus erythematosus (SLE) was examined by using the polymerase chain reaction (PCR) method with genomic DNA and allele-specific primers. In the frequency of IL-10 promoter 1082 genotypes consisting of AA, A/G and GG, the allele frequency of G in the SLE patients was significantly higher than that in the healthy controls. On the other hand, there was no statistical difference in the frequency of TNF receptor (TNFR) II 196 genotypes between the SLE patients and healthy controls. It was therefore suggested that the polymorphism of the IL-10 promoter, but not TNFRII, might participate in the pathogenesis of SLE in Vietnamese.


Subject(s)
Interleukin-10/genetics , Lupus Erythematosus, Systemic/genetics , Polymorphism, Genetic , Promoter Regions, Genetic/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics , Adolescent , Adult , Child , Cross-Sectional Studies , DNA/analysis , Electrophoresis, Agar Gel , Female , Gene Frequency/genetics , Genetic Markers , Genotype , Humans , Interleukin-10/blood , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/epidemiology , Male , Middle Aged , Polymerase Chain Reaction , Receptors, Tumor Necrosis Factor, Type II/blood , Vietnam/epidemiology
20.
J Endotoxin Res ; 10(1): 25-31, 2004.
Article in English | MEDLINE | ID: mdl-15025821

ABSTRACT

The biological actions of lipopolysaccharides (LPSs) from Sinorhizobium meliloti, Mesorhizobium loti and Escherichia coli were compared. In biological activities including lethality, production of tumor necrosis factor (TNF)-alpha and nitric oxide (NO), adjuvant action and Limulus activity, LPS from S. meliloti exhibited stronger actions than LPS from M. loti, but had a weaker action than LPS from E. coli. On the other hand, M. loti LPS showed a higher activity to activate human complement than S. meliloti LPS. Further, there was a significant difference in polymyxin B binding between S. meliloti LPS and M. loti LPS, suggesting a difference in the lipid A structure. LPSs from S. meliloti and M. loti seem to exhibit characteristic biological actions that may be dependent on the difference in the lipid A structure.


Subject(s)
Lipopolysaccharides/isolation & purification , Lipopolysaccharides/pharmacology , Proteobacteria/pathogenicity , Sinorhizobium/pathogenicity , Animals , Biological Assay , Escherichia coli/pathogenicity , Horseshoe Crabs , Nitric Oxide/analysis , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL