Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters










Publication year range
1.
Immune Netw ; 23(5): e37, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37970231

ABSTRACT

Forkhead box P3-positive (Foxp3+)-inducible Tregs (iTregs) are readily generated by TGF-ß1 at low TCR signaling intensity. TGF-ß1-mediated Foxp3 expression is further enhanced by retinoic acid (RA) and lactoferrin (LF). However, the intensity of TCR signaling required for induction of Foxp3 expression by TGF-ß1 in combination with RA and LF is unknown. Here, we found that either RA or LF alone decreased TGF-ß1-mediated Foxp3 expression at low TCR signaling intensity. In contrast, at high TCR signaling intensity, the addition of either RA or LF strongly increased TGF-ß1-mediated Foxp3 expression. Moreover, decreased CD28 stimulation was more favorable for TGF-ß1/LF-mediated Foxp3 expression. Lastly, we found that at high signaling intensities of both TCR and CD28, combined treatment with TGF-ß1, RA, and LF induced robust expression of Foxp3, in parallel with powerful suppressive activity against responder T cell proliferation. Our findings that TGFß/RA/LF strongly generate high affinity Ag-specific iTreg population would be useful for the control of unwanted hypersensitive immune reactions such as various autoimmune diseases.

2.
Immunology ; 168(1): 110-119, 2023 01.
Article in English | MEDLINE | ID: mdl-36054548

ABSTRACT

We recently reported that lactoferrin (LF) induces Foxp3+ Treg differentiation through binding to TGFß receptor III (TßRIII), and this activity was further enhanced by TGFß1. Generally, a low T-cell receptor (TCR) signal strength is favourable for Foxp3+ Treg differentiation. In the present study, we explored the effect of lactoferrin chimera (LFch, containing lactoferricin [aa 17-30] and lactoferrampin [aa 265-284]), along with TGFß1 on Foxp3+ Treg differentiation. LFch alone did not induce Foxp3 expression, yet LFch dramatically enhanced TGFß1-induced Foxp3 expression. LFch had little effect on the phosphorylation of Smad3, a canonical transcriptional factor of TGFß1. Instead, LFch attenuated the phosphorylation of S6 (a target of mTOR), IκB and PI3K. These activities of LFch were completely abrogated by pretreatment of LFch with soluble TGFß1 receptor III (sTßRIII). Consistent with this, the activity of LFch on TGFß1-induced Foxp3 expression was also abrogated by treatment with sTßRIII. Finally, the TGFß1/LFch-induced T cell population substantially suppressed the proliferation of responder CD4+ T cells. These results indicate that LFch robustly enhances TGFß1-induced Foxp3+ Treg differentiation by diminishing TCR/CD28 signal intensity.


Subject(s)
CD28 Antigens , T-Lymphocytes, Regulatory , T-Lymphocytes, Regulatory/metabolism , Lactoferrin/pharmacology , Lactoferrin/metabolism , Receptors, Antigen, T-Cell/metabolism , Cell Differentiation , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism
3.
J Immunol ; 207(10): 2456-2464, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34615735

ABSTRACT

Lactoferrin (LF) is known to possess anti-inflammatory activity, although its mechanisms of action are not well-understood. The present study asked whether LF affects the commitment of inducible regulatory T cells (Tregs). LF substantially promoted Foxp3 expression by mouse activated CD4+T cells, and this activity was further enhanced by TGF-ß1. Interestingly, blocking TGF-ß with anti-TGF-ß Ab completely abolished LF-induced Foxp3 expression. However, no significant amount of soluble TGF-ß was released by LF-stimulated T cells, suggesting that membrane TGF-ß (mTGF-ß) is associated. Subsequently, it was found that LF binds to TGF-ß receptor III, which induces reactive oxygen species production and diminishes the expression of mTGF-ß-bound latency-associated peptide, leading to the activation of mTGF-ß. It was followed by phosphorylation of Smad3 and enhanced Foxp3 expression. These results suggest that LF induces Foxp3+ Tregs through TGF-ß receptor III/reactive oxygen species-mediated mTGF-ß activation, triggering canonical Smad3-dependent signaling. Finally, we found that the suppressive activity of LF-induced Tregs is facilitated mainly by CD39/CD73-induced adenosine generation and that this suppressor activity alleviates inflammatory bowel disease.


Subject(s)
Lactoferrin/metabolism , Receptors, Transforming Growth Factor beta/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Animals , Cell Differentiation/drug effects , Cell Differentiation/immunology , Colitis/immunology , Colitis/metabolism , Lactoferrin/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice, Inbred BALB C , Receptors, Transforming Growth Factor beta/drug effects , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/metabolism
4.
J Immunol ; 206(3): 481-493, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33380497

ABSTRACT

B cells in the germinal center (GC) are programmed to form plasma cells (PCs) or memory B cells according to signals received by receptors that are translated to carry out appropriate activities of transcription factors. However, the precise mechanism underlying this process to complete the GC reaction is unclear. In this study, we show that both genetic ablation and pharmacological inhibition of glycogen synthase kinase 3 (GSK3) in GC B cells of mice facilitate the cell fate decision toward PC formation, accompanied by acquisition of dark zone B cell properties. Mechanistically, under stimulation with CD40L and IL-21, GSK3 inactivation synergistically induced the transcription factors Foxo1 and c-Myc, leading to increased levels of key transcription factors required for PC differentiation, including IRF4. This GSK3-mediated alteration of transcriptional factors in turn facilitated the dark zone transition and consequent PC fate commitment. Our study thus reveals the upstream master regulator responsible for interpreting external cues in GC B cells to form PCs mediated by key transcription factors.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Glycogen Synthase Kinase 3/metabolism , Plasma Cells/immunology , Animals , CD40 Ligand/metabolism , Cell Differentiation , Cells, Cultured , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Gene Expression Regulation , Glycogen Synthase Kinase 3/genetics , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Interleukins/metabolism , Lymphocyte Activation , Mice , Mice, Knockout , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myb/metabolism
5.
Immune Netw ; 20(5): e38, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33163246

ABSTRACT

Dendritic cells (DCs) are professional antigen-presenting cells (APCs) that initiate both T-cell responses and tolerance. Tolerogenic DCs (tDCs) are regulatory DCs that suppress immune responses through the induction of T-cell anergy and Tregs. Because lactoferrin (LF) was demonstrated to induce functional Tregs and has a protective effect against inflammatory bowel disease, we explored the tolerogenic effects of LF on mouse bone marrow-derived DCs (BMDCs). The expression of CD80/86 and MHC class II was diminished in LF-treated BMDCs (LF-BMDCs). LF facilitated BMDCs to suppress proliferation and elevate Foxp3+ induced Treg (iTreg) differentiation in ovalbumin-specific CD4+ T-cell culture. Foxp3 expression was further increased by blockade of the B7 molecule using CTLA4-Ig but was diminished by additional CD28 stimulation using anti-CD28 Ab. On the other hand, the levels of arginase-1 and indoleamine 2,3-dioxygenase-1 (known as key T-cell suppressive molecules) were increased in LF-BMDCs. Consistently, the suppressive activity of LF-BMDCs was partially restored by inhibitors of these molecules. Collectively, these results suggest that LF effectively causes DCs to be tolerogenic by both the suppression of T-cell proliferation and enhancement of iTreg differentiation. This tolerogenic effect of LF is due to the reduction of costimulatory molecules and enhancement of suppressive molecules.

6.
Cells ; 9(6)2020 06 26.
Article in English | MEDLINE | ID: mdl-32604872

ABSTRACT

The effector function of tumor-infiltrated CD4+ T cells is readily suppressed by many types of immune regulators in the tumor microenvironment, which is one of the major mechanisms of immune tolerance against cancer. Cathelicidin-related antimicrobial peptide (CRAMP), the mouse analog of LL-37 peptide in humans, is a cationic antimicrobial peptide belonging to the cathelicidin family; however, its secretion by cancer cells and role in the tumor microenvironment (TME) remain unclear. In this study, we explored the possibility of an interaction between effector CD4+ T cells and CRAMP using in vitro-generated mouse Th17 cells. We found that CRAMP stimulates Th17 cells to express the ectonucleotidase CD73, while simultaneously inducing cell death. This finding suggested that CD73-expressing Th17 cells may function as immune suppressor cells instead of effector cells. In addition, treatment of pharmacological inhibitors of the transforming growth factor-beta (TGF-ß) signaling pathway showed that induction of CD73 expression is mediated by the p38 signaling pathway. Overall, our findings suggest that tumor-derived LL-37 likely functions as an immune suppressor that induces immune tolerance against tumors through shaping effector Th17 cells into suppressor Th17 cells, suggesting a new intervention target to improve cancer immunotherapy.


Subject(s)
Antigens, Neoplasm/metabolism , Antimicrobial Cationic Peptides/metabolism , Tetraspanins/metabolism , Th17 Cells/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Female , Humans , Mice
7.
Sci Rep ; 9(1): 8397, 2019 06 10.
Article in English | MEDLINE | ID: mdl-31182790

ABSTRACT

Inhibitor of kappa B (IκB)-ζ transcription is rapidly induced by stimulation with TLR ligands and IL-1. Despite high IκBζ expression in inflammation sites, the association of IκBζ with host defence via systemic immune responses against bacterial infection remains unclear. Oral immunisation with a recombinant attenuated Salmonella vaccine (RASV) strain did not protect IκBζ-deficient mice against a lethal Salmonella challenge. IκBζ-deficient mice failed to produce Salmonella LPS-specific IgG, especially IgG2a, although inflammatory cytokine production and immune cell infiltration into the liver increased after oral RASV administration. Moreover, IκBζ-deficient mice exhibited enhanced splenic germinal centre reactions followed by increased total IgG production, despite IκBζ-deficient B cells having an intrinsic antibody class switching defect. IκBζ-deficient CD4+ T cells poorly differentiated into Th1 cells. IFN-γ production by CD4+ T cells from IκBζ-deficient mice immunised with RASV significantly decreased after restimulation with heat-killed RASV in vitro, suggesting that IκBζ-deficient mice failed to mount protective immune responses against Salmonella infection because of insufficient Th1 and IgG production. Therefore, IκBζ is crucial in protecting against Salmonella infection by inducing Th1 differentiation followed by IgG production.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation , Immunity , Immunoglobulin G/biosynthesis , Salmonella Infections/immunology , Salmonella Infections/prevention & control , Th1 Cells/immunology , Administration, Oral , Animals , Chronic Disease , Germinal Center/metabolism , Immunization , Inflammation/pathology , Interferon-gamma/metabolism , Lipopolysaccharides , Mice, Inbred C57BL , Salmonella Infections/parasitology , Salmonella Vaccines/immunology , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Vaccines, Attenuated/immunology , Virulence
8.
J Ginseng Res ; 43(2): 291-299, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30976167

ABSTRACT

BACKGROUND: Ginsenosides of Korean Red Ginseng extracts (RGE) and its saponin components suppress secretion of inflammasome-mediating cytokines, whereas the nonsaponin fraction (NS) of RGE oppositely stimulates cytokine secretion. Although direct exposure of NS to macrophages in mice induces cytokine production, oral administration of NS has not been studied in inflammasome-related disease in animal models. METHODS: Mice were fed RGE or NS for 7 days and then developed peritonitis. Peritoneal cytokines were measured, and peritoneal exudate cells (PECs) were collected to assay expression levels of a set of toll-like receptors (TLRs) and cytokines in response to NS ingestion. In addition, the role of intestinal bacteria in NS-fed mice was assessed. The effect of preexposure to NS in bone marrow-derived macrophages (BMDMs) on cytokine production was further confirmed. RESULTS: NS ingestion attenuated secretion of peritoneal cytokines resulting from peritonitis. In addition, the isolated PECs from NS-fed mice presented lower TLR transcription levels than PECs from control diet-fed mice. BMDMs treated with NS showed downregulation of TLR4 mRNA and protein expression, which was mediated by the TLR4-MyD88-NFκB signal pathway. BMDMs pretreated with NS produced less cytokines in response to TLR4 ligands. CONCLUSION: NS administration directly inhibits TLR4 expression in inflammatory cells such as macrophages, thereby reducing secretion of cytokines during peritonitis.

9.
Cell Immunol ; 338: 1-8, 2019 04.
Article in English | MEDLINE | ID: mdl-30850088

ABSTRACT

Imiquimod (IMQ) is a selective toll-like receptor 7 (TLR7) agonist. TLR7 activation leads to the production of IFN-γ and pro-inflammatory cytokines by innate immune cells. However, the role of TLR7 in B cells is not fully understood. In this study, we investigated the direct effect of in vitro stimulation with IMQ on Ab production and isotype switching in B cells. IMQ selectively diminished IL-4-induced IgE and IgG1 production in anti-CD40-activated mouse B cells. IMQ also inhibited germline ε transcripts (GLTε)/GLTγ1 and post-switch ε transcripts (PSTε)/PSTγ1 expression, while enhancing GLTγ2c and PSTγ2c expression in anti-CD40/IL-4-stimulated B cells. Interestingly, IMQ abrogated IL-4-induced circle transcripts ε-γ1 (CTε-γ1) expression, indicative of sequential switching from IgG1 to IgE. Furthermore, IMQ repressed IL-4-induced surface IgE/IgG1 expression while increasing surface IgG2c expression. The selective inhibition of IgE synthesis was not due to IMQ-induced production of IFN-γ or IL-12 in the same culture. IMQ also enhanced BCL6 expression, a transcriptional repressor for the GLTε promoter, in anti-CD40/IL-4-stimulated B cells. In addition, BCL6 siRNA restored IMQ-mediated suppression of GLTε transcription. Therefore, these results indicate that TLR7 engagement by IMQ inhibits IL-4-induced GLTε transcription by enhancing BCL6 expression and inhibits IL-4-induced sequential switching from IgM to IgE via IgG1, thus resulting in the downregulation of IgE production by B cells.


Subject(s)
B-Lymphocytes/physiology , Imiquimod/pharmacology , Immunoglobulin E/metabolism , Proto-Oncogene Proteins c-bcl-6/metabolism , Toll-Like Receptor 7/agonists , Animals , Antibody Formation , CD40 Antigens/metabolism , Cells, Cultured , Gene Expression Regulation , Immunoglobulin Class Switching , Immunoglobulin E/genetics , Immunoglobulin G/genetics , Immunoglobulin epsilon-Chains/genetics , Interleukin-4/metabolism , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-bcl-6/genetics , RNA, Small Interfering/genetics
10.
J Biomed Res ; 32(5): 401-410, 2018 Nov 20.
Article in English | MEDLINE | ID: mdl-30514828

ABSTRACT

Inflammatory responses are essential in eliminating harmful substrates from damaged tissue and inducing recovery. Several cytokines participate in and facilitate this response. Certain cytokines such as interleukin (IL)-1ß and IL-18 are initially produced in precursor form in response to toll-like receptor (TLR) ligands and undergo maturation by inflammasomes, which are cytosolic multi-protein complexes containing nucleotide-binding oligomerization domain (NOD)-containing protein 2-like receptors (NLRs). Immune modulators targeting inflammasomes have been investigated to control inflammatory diseases such as metabolic syndrome. However, most immune modulators possessing anti-inflammasome properties attenuate production of other cytokines, which are essential for host defense. In this review, we analyzed the effect of anti-inflammasome agents on the production of cytokines which are not regulated by inflammasome and involving in initial immune responses. As a result, the inflammasome inhibitors are put into three categories: non-effector, stimulator, or inhibitor of cytokine production. Even the stimulator of cytokine production ameliorated symptoms resulting from inflammasome activation in mouse models. Thus, we suggest ideal immune modulators targeting inflammasomes in order to enhance cytokine production while inhibiting cytokine maturation.

11.
Immune Netw ; 18(4): e25, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30181913

ABSTRACT

γδ T cells are abundant in the gut mucosa and play an important role in adaptive immunity as well as innate immunity. Although γδ T cells are supposed to be associated with the enhancement of Ab production, the status of γδ T cells, particularly in the synthesis of IgA isotype, remains unclear. We compared Ig expression in T cell receptor delta chain deficient (TCRδ-/-) mice with wild-type mice. The amount of IgA in fecal pellets was substantially elevated in TCRδ-/- mice. This was paralleled by an increase in surface IgA expression and total IgA production by Peyer's patches (PPs) and mesenteric lymph node (MLN) cells. Likewise, the TCRδ-/- mice produced much higher levels of serum IgA isotype. Here, surface IgA expression and number of IgA secreting cells were also elevated in the culture of spleen and bone marrow (BM) B cells. Germ-line α transcript, an indicator of IgA class switch recombination, higher in PP and MLN B cells from TCRδ-/- mice, while it was not seen in inactivated B cells. Nevertheless, the frequency of IgA+ B cells was much higher in the spleen from TCRδ-/- mice. These results suggest that γδ T cells control the early phase of B cells, in order to prevent unnecessary IgA isotype switching. Furthermore, this regulatory role of γδ T cells had lasting effects on the long-lived IgA-producing plasma cells in the BM.

12.
Sci Rep ; 8(1): 13659, 2018 09 12.
Article in English | MEDLINE | ID: mdl-30209319

ABSTRACT

Exposure to heavy metals can cause several diseases associated with the immune system. Although the effects of heavy metals on production of inflammatory cytokines have been previously studied, the role of heavy metals in inflammasome activation remains poorly studied. The inflammasome is an intracellular multi-protein complex that detects intracellular danger signals, resulting in inflammatory responses such as cytokine maturation and pyroptosis. In this study, we elucidated the effects of four heavy metals, including cadmium (Cd), mercury (Hg), arsenic (As), and lead (Pb), on the activation of NLRP3, NLRC4, and AIM2 inflammasomes. In our results, mercury and arsenic inhibited interleukin (IL)-1ß and IL-18 secretion resulting from canonical and non-canonical NLRP3 inflammasome activation in macrophages and attenuated elevation of serum IL-1ß in response to LPS treatment in mice. In the mechanical studies, mercury interrupted production of mitochondrial reactive oxygen species, release of mitochondrial DNA, and activity of recombinant caspase-1, whereas arsenic down-regulated expression of promyelocytic leukemia protein. Both mercury and arsenic inhibited Asc pyroptosome formation and gasdermin D cleavage. Thus, we suggest that exposure to mercury and/or arsenic could disrupt inflammasome-mediated inflammatory responses, which might cause unexpected side effects.


Subject(s)
Arsenic/toxicity , Inflammasomes/metabolism , Mercury/toxicity , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins/biosynthesis , Cadmium/toxicity , Calcium-Binding Proteins/metabolism , Caspase 1/metabolism , Cell Line , DNA, Mitochondrial/genetics , DNA-Binding Proteins/metabolism , Female , Interleukin-13/blood , Interleukin-13/metabolism , Interleukin-18/blood , Interleukin-18/metabolism , Intracellular Signaling Peptides and Proteins , Lead/toxicity , Macrophages/immunology , Mice , Mice, Inbred C57BL , Phosphate-Binding Proteins , Promyelocytic Leukemia Protein/biosynthesis , Reactive Oxygen Species/metabolism
13.
Sci Rep ; 8(1): 14307, 2018 09 24.
Article in English | MEDLINE | ID: mdl-30250171

ABSTRACT

Bacteria move toward attractants and away from repellants by rotating their flagellum. The bacterial flagellum assembles through the ordered organization of more than 30 different proteins. Among the diverse flagellar proteins, FlgL forms the junction between the hook and the filament in the flagellum together with FlgK and provides a structural base where flagellin, a filament-forming protein, is inserted for the initiation of filament elongation. However, the functional and structural information available for FlgL is highly limited. To provide structural insights into the cross-linkage between the FlgL junction and the flagellin filament, we determined the crystal structures of FlgL from gram-positive Bacillus cereus (bcFlgL) and gram-negative Xanthomonas campestris (xcFlgL). bcFlgL contains one domain (D1), whereas xcFlgL adopts a two-domain structure that consists of the D1 and D2 domains. The constant D1 domain of FlgL adopts a rod structure that is generated by four longitudinal segments. This four-segment structure is recapitulated in filament and junction proteins but not in hook and rod proteins, allowing us to propose a junction-filament assembly mechanism based on a quasi-homotypic interaction. The D2 domain of xcFlgL resembles that of another junction protein, FlgK, suggesting the structural and functional relatedness of FlgL and FlgK.


Subject(s)
Bacterial Proteins/chemistry , Flagella/metabolism , Flagellin/chemistry , Crystallography, X-Ray , Models, Molecular , Protein Domains , Protein Structure, Secondary
14.
FEBS J ; 285(20): 3786-3800, 2018 10.
Article in English | MEDLINE | ID: mdl-30095229

ABSTRACT

VanR is a negative transcriptional regulator of bacteria that belongs to the PadR family and modulates the expression of vanillate transport and degradation proteins in response to vanillate. Although VanR plays a key role in the utilization of vanillate as a carbon source, it is barely understood how VanR recognizes its effector. Thus, our knowledge concerning the gene regulatory mechanism of VanR is limited. Here, we reveal the vanillate-binding mode of VanR through structural, biophysical, and mutational studies. Similar to other PadR family members, VanR forms a functional dimer, and each VanR subunit consists of an N-terminal DNA-binding domain (NTD) and a C-terminal dimerization domain (CTD). One VanR dimer simultaneously binds two vanillate molecules using two interdomain cavities, as observed in PadR. In contrast to these common features, VanR contains an additional α-helix, αi, that has not been found in other PadR family members. The αi helix functions as an interdomain crosslinker that mediates interactions between the NTD and the CTD. In addition, the VanR-specific αi helix plays a key role in the formation of a unique effector-binding site. As a result, the effector-binding mode of VanR is distinguishable from that of PadR in the location and accessibility of the effector-binding site as well as the orientation of its bound effector. Furthermore, we propose the DNA-binding mode and vanillate-mediated transcriptional regulation mechanism of VanR based on comparative structural and mutational analyses. DATABASES: The atomic coordinates and the structure factors for VanR (PDB ID 5Z7B) have been deposited in the Protein Data Bank, www.pdb.org.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Benzaldehydes/metabolism , Corynebacterium glutamicum/metabolism , DNA, Bacterial/metabolism , Protein Conformation, alpha-Helical , Transcription Factors/chemistry , Transcription Factors/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Crystallography, X-Ray , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Models, Molecular , Mutation , Promoter Regions, Genetic , Protein Binding , Protein Domains , Protein Multimerization , Sequence Homology , Transcription Factors/genetics
15.
Cell Host Microbe ; 24(2): 249-260.e4, 2018 08 08.
Article in English | MEDLINE | ID: mdl-30092201

ABSTRACT

Innate lymphoid cells (ILCs) are important regulators of early infection at mucosal barriers. ILCs are divided into three groups based on expression profiles, and are activated by cytokines and neuropeptides. Yet, it remains unknown if ILCs integrate other signals in providing protection. We show that signaling through herpes virus entry mediator (HVEM), a member of the tumor necrosis factor (TNF) receptor superfamily, in ILC3 is important for host defense against oral infection with the bacterial pathogen Yersinia enterocolitica. HVEM stimulates protective interferon-γ (IFN-γ) secretion from ILCs, and mice with HVEM-deficient ILC3 exhibit reduced IFN-γ production, higher bacterial burdens and increased mortality. In addition, IFN-γ production is critical as adoptive transfer of wild-type but not IFN-γ-deficient ILC3 can restore protection to mice lacking ILCs. We identify the TNF superfamily member, LIGHT, as the ligand inducing HVEM signals in ILCs. Thus HVEM signaling mediated by LIGHT plays a critical role in regulating ILC3-derived IFN-γ production for protection following infection. VIDEO ABSTRACT.


Subject(s)
Enterobacteriaceae Infections/prevention & control , Interferon-gamma/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Receptors, Tumor Necrosis Factor, Member 14/immunology , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Signal Transduction , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Adoptive Transfer , Adult , Animals , Cytokines/metabolism , Disease Models, Animal , Enterobacteriaceae Infections/pathology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/metabolism , Protein Transport , Receptors, CCR6/genetics , Receptors, CCR6/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Spleen/microbiology , Spleen/pathology , Yersinia enterocolitica/pathogenicity
16.
Vet Res Commun ; 42(4): 265-273, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30097755

ABSTRACT

Pigs are an important livestock and serve as a large animal model due to physiological and anatomical similarities with humans. Thus, components of the porcine immune system such as inflammasomes need to be characterized for disease control, vaccination, and translational research purposes. Previously, we and others elucidated porcine nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family Pyrin domain containing 3 (NLRP3) inflammasome activation. However, until now, porcine NLR family caspase recruitment domain (CARD)-containing 4 (NLRC4) and absent in melanoma 2 (AIM2) inflammasomes have been not well studied. In this study, we treated well defined NLRC4 and AIM2 inflammasome triggers to porcine peripheral blood mononuclear cells (PBMCs) and murine bone-marrow derived macrophages (BMDMs) and observed interleukin (IL)-1ß maturation as a readout of inflammasome activation. NLRC4 (flagellin) and AIM2 (dsDNA) triggers led to IL-1ß secretion in both porcine PBMCs and mice macrophages. In addition, porcine and mouse NLRC4 and AIM2 inflammasomes responded differently to NLRP3 inhibitors. Bacterial inflammasome triggers, Salmonella enterica serovar Typhimurium, Listeria monocytogenes, and Escherichia coli, also induced IL-1ß secretion in porcine PBMCs. Taken together, we suggest that known triggers of NLRC4 and AIM2 inflammasomes in mice induce IL-1ß secretion in porcine PBMCs.


Subject(s)
DNA-Binding Proteins/metabolism , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Nod Signaling Adaptor Proteins/metabolism , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , Inflammasomes/drug effects , Leukocytes, Mononuclear/metabolism , Mice , Mice, Inbred C57BL , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/metabolism , Swine , Swine Diseases/immunology , Swine Diseases/metabolism , Swine Diseases/microbiology
17.
Antiviral Res ; 151: 87-96, 2018 03.
Article in English | MEDLINE | ID: mdl-29407486

ABSTRACT

Human rhinovirus (HRV) infection causes more than 80% of all common colds and is associated with severe complications in patients with asthma and chronic obstructive pulmonary disease. To identify antiviral drug against HRV infection, we screened 800 FDA-approved drugs and found budesonide as one of the possible drug candidates. Budesonide is a corticosteroid, which is commonly used to prevent exacerbation of asthma and symptoms of common cold. Budesonide specifically protects host cells from cytotoxicity following HRV infection, which depend on the expression of glucocorticoid receptor. Intranasal administration of budesonide lowered the pulmonary HRV load and the levels of IL-1ß cytokine leading to decreased lung inflammation. Budesonide regulates IL-1ß production following HRV infection independent of inflammasome activation. Instead, budesonide induces mitochondrial reactive oxygen species followed by activation of autophagy. Further, the inhibition of autophagy following chloroquine or bafilomycin A1 treatment reduced the anti-viral effect of budesonide against HRV, suggesting that the antiviral activity of budesonide was mediated via autophagy. The results suggest that budesonide represents a promising antiviral and anti-inflammatory drug candidate for the treatment of human rhinovirus infection.


Subject(s)
Autophagy/drug effects , Budesonide/administration & dosage , Budesonide/pharmacology , Picornaviridae Infections/drug therapy , Rhinovirus/drug effects , Administration, Intranasal , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacology , Cell Line , Disease Models, Animal , Female , Humans , Inflammation/metabolism , Inflammation/virology , Interleukin-1beta/metabolism , Lung/pathology , Lung/virology , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Viral Load/drug effects , Virus Replication/drug effects
18.
Cell Mol Immunol ; 15(2): 111-119, 2018 02.
Article in English | MEDLINE | ID: mdl-27133472

ABSTRACT

Poly-gamma-glutamic acid (γ-PGA) is a natural, edible and non-toxic polymer synthesized by Bacillus subtilis and is suggested as a safe biomaterial for the use in hydrogels and vaccine adjuvants. However, the effect of γ-PGA on inflammasome activation has not yet been studied in macrophages. Inflammasomes, which are intracellular multi-protein complexes, promote acute and chronic inflammation via interleukin-1ß or interleukin-18 maturation, and they are known targets for metabolic syndromes and cancer. In this study, we observed that γ-PGA attenuated NLRP3, NLRC4 and AIM2 inflammasome activation, whereas it upregulated pro-inflammatory cytokine expression in human and murine macrophages. Although γ-PGA had conflicting effects on cytokine production and maturation, it clearly alleviated the severity of lipopolysaccharide-induced endotoxin shock in an animal model. Thus, we suggest γ-PGA as a candidate to control inflammasome-mediated disorders.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Bacillus subtilis/chemistry , Calcium-Binding Proteins/metabolism , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Polyglutamic Acid/analogs & derivatives , Up-Regulation/drug effects , Animals , Caspase 1/metabolism , Glutamic Acid/pharmacology , Inflammation/pathology , Inflammation Mediators/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Polyglutamic Acid/chemistry , Polyglutamic Acid/pharmacology , Signal Transduction/drug effects , Glycine max , Toll-Like Receptor 4/metabolism
19.
Sci Rep ; 7(1): 12409, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28963531

ABSTRACT

Methylene blue (MB), which has antioxidant, anti-inflammatory, neuroprotective, and mitochondria protective effects, has been widely used as a dye and medication. However, the effect of MB on inflammasome activation has not yet been studied. Inflammasomes are multi-protein complexes that induce maturation of interleukins (ILs)-1ß and -18 as well as caspase-1-mediated cell death, known as pyroptosis. Dysregulation of inflammasomes causes several diseases such as type 2 diabetes, Alzheimer's disease, and gout. In this study, we assess the effect of MB on inflammasome activation in macrophages. As the result, MB attenuated activation of canonical inflammasomes such as NLRP3, NLRC4, and AIM2 as well as non-canonical inflammasome activation. In addition, MB inhibited upstream signals such as inflammasome assembly, phagocytosis, and gene expression of inflammasome components via inhibition of NF-κB signaling. Furthermore, MB reduced the activity of caspase-1. The anti-inflammasome properties of MB were further confirmed in mice models. Thus, we suggest that MB is a broad-spectrum anti-inflammasome candidate molecule.


Subject(s)
Inflammasomes/drug effects , Macrophages/drug effects , Methylene Blue/pharmacology , Animals , Apoptosis Regulatory Proteins/antagonists & inhibitors , CARD Signaling Adaptor Proteins/antagonists & inhibitors , Calcium-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/antagonists & inhibitors , Gene Expression/drug effects , Humans , Inflammasomes/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Phagocytosis/drug effects , Pyroptosis/drug effects , RAW 264.7 Cells , THP-1 Cells
20.
Cell Immunol ; 322: 49-55, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29042055

ABSTRACT

The present study extends an earlier report that retinoic acid (RA) down-regulates IgE Ab synthesis in vitro. Here, we show the suppressive activity of RA on IgE production in vivo and its underlying mechanisms. We found that RA down-regulated IgE class switching recombination (CSR) mainly through RA receptor α (RARα). Additionally, RA inhibited histone acetylation of germ-line ε (GL ε) promoter, leading to suppression of IgE CSR. Consistently, serum IgE levels were substantially elevated in vitamin A-deficient (VAD) mice and this was more dramatic in VAD-lecithin:retinol acyltransferase deficient (LRAT-/-) mice. Further, serum mouse mast cell protease-1 (mMCP-1) level was elevated while frequency of intestinal regulatory T cells (Tregs) were diminished in VAD LRAT-/- mice, reflecting that deprivation of RA leads to allergic immune response. Taken together, our results reveal that RA has an IgE-repressive activity in vivo, which may ameliorate IgE-mediated allergic disease.


Subject(s)
Immunoglobulin Class Switching/drug effects , Immunoglobulin E/biosynthesis , Interleukin-4/metabolism , Tretinoin/pharmacology , Vitamin A Deficiency/blood , Acyltransferases/deficiency , Acyltransferases/genetics , Animals , Chymases/metabolism , Food Hypersensitivity/drug therapy , Food Hypersensitivity/immunology , Immunoglobulin Class Switching/immunology , Immunoglobulin E/blood , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Retinoic Acid Receptor alpha/immunology , T-Lymphocytes, Regulatory/immunology , Vitamin A/genetics , Vitamin A Deficiency/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...