Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 327
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 15(1): 36-44, 2014 01.
Article in English | MEDLINE | ID: mdl-24212998

ABSTRACT

Eosinophilia is a hallmark characteristic of T helper type 2 (TH2) cell-associated diseases and is critically regulated by the central eosinophil growth factor interleukin 5 (IL-5). Here we demonstrate that IL-5 activity in eosinophils was regulated by paired immunoglobulin-like receptors PIR-A and PIR-B. Upon self-recognition of ß2-microglobulin (ß2M) molecules, PIR-B served as a permissive checkpoint for IL-5-induced development of eosinophils by suppressing the proapoptotic activities of PIR-A, which were mediated by the Grb2-Erk-Bim pathway. PIR-B-deficient bone marrow eosinophils underwent compartmentalized apoptosis, resulting in decreased blood eosinophilia in naive mice and in mice challenged with IL-5. Subsequently, Pirb(-/-) mice displayed impaired aeroallergen-induced lung eosinophilia and induction of lung TH2 cell responses. Collectively, these data uncover an intrinsic, self-limiting pathway regulating IL-5-induced expansion of eosinophils, which has broad implications for eosinophil-associated diseases.


Subject(s)
Cell Differentiation/immunology , Eosinophils/immunology , Interleukin-5/immunology , Receptors, Immunologic/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , Apoptosis Regulatory Proteins/metabolism , Asthma/genetics , Asthma/immunology , Asthma/metabolism , Bcl-2-Like Protein 11 , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Colony-Forming Units Assay/methods , Eosinophils/cytology , Eosinophils/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Flow Cytometry , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/immunology , GRB2 Adaptor Protein/metabolism , Gene Expression/immunology , Interleukin-5/pharmacology , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/immunology , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/immunology
2.
J Virol ; 98(2): e0203523, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38299844

ABSTRACT

Bovine viral diarrhea virus (BVDV) is prevalent worldwide and causes significant economic losses. Gut microbiota is a large microbial community and has a variety of biological functions. However, whether there is a correlation between gut microbiota and BVDV infection and what kind of relation between them have not been reported. Here, we found that gut microbiota composition changed in normal mice after infecting with BVDV, but mainly the low abundance microbe was affected. Interestingly, BVDV infection significantly reduced the diversity of gut microbiota and changed its composition in gut microbiota-dysbiosis mice. Furthermore, compared with normal mice of BVDV infection, there were more viral loads in the duodenum, jejunum, spleen, and liver of the gut microbiota-dysbiosis mice. However, feces microbiota transplantation (FMT) reversed these effects. The data above indicated that the dysbiosis of gut microbiota was a key factor in the high infection rate of BVDV. It is found that the IFN-I signal was involved by investigating the underlying mechanisms. The inhibition of the proliferation and increase in the apoptosis of peripheral blood lymphocytes (PBL) were also observed. However, FMT treatment reversed these changes by regulating PI3K/Akt, ERK, and Caspase-9/Caspase-3 pathways. Furthermore, the involvement of butyrate in the pathogenesis of BVDV was also further confirmed. Our results showed for the first time that gut microbiota acts as a key endogenous defense mechanism against BVDV infection; moreover, targeting regulation of gut microbiota structure and abundance may serve as a new strategy to prevent and control the disease.IMPORTANCEWhether the high infection rate of BVDV is related to gut microbiota has not been reported. In addition, most studies on BVDV focus on in vitro experiments, which limits the study of its prevention and control strategy and its pathogenic mechanism. In this study, we successfully confirmed the causal relationship between gut microbiota and BVDV infection as well as the potential molecular mechanism based on a mouse model of BVDV infection and a mouse model of gut microbiota dysbiosis. Meanwhile, a mouse model which is more susceptible to BVDV provided in this study lays an important foundation for further research on prevention and control strategy of BVDV and its pathogenesis. In addition, the antiviral effect of butyrate, the metabolites of butyrate-producing bacteria, has been further revealed. Overall, our findings provide a promising prevention and control strategy to treat this infectious disease which is distributed worldwide.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease , Diarrhea Viruses, Bovine Viral , Gastrointestinal Microbiome , Animals , Cattle , Mice , Bovine Virus Diarrhea-Mucosal Disease/complications , Bovine Virus Diarrhea-Mucosal Disease/microbiology , Bovine Virus Diarrhea-Mucosal Disease/therapy , Bovine Virus Diarrhea-Mucosal Disease/virology , Butyrates/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Diarrhea , Diarrhea Viruses, Bovine Viral/pathogenicity , Diarrhea Viruses, Bovine Viral/physiology , Dysbiosis/complications , Dysbiosis/microbiology , Dysbiosis/virology , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fecal Microbiota Transplantation , Interferon Type I/immunology , Interferon Type I/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Disease Models, Animal
3.
Immunity ; 43(1): 65-79, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26141582

ABSTRACT

Colonization with a mixture of Clostridium species has been shown to induce accumulation of induced regulatory T (iTreg) cells in the colon. Transforming growth factor-ß (TGF-ß) is an essential factor for iTreg cell induction; however, the relationship between Clostridium species and TGF-ß remains to be clarified. Here we demonstrated that a gram-positive probiotic bacterial strain, Clostridium butyricum (C. butyricum), promoted iTreg cell generation in the intestine through induction of TGF-ß1 from lamina propria dendritic cells (LPDCs). C. butyricum-mediated TGF-ß1 induction was mainly Toll-like receptor 2 (TLR2) dependent, and the ERK-AP-1 kinase pathway played an important role. In addition, the autocrine TGF-ß-Smad3 transcription factor signal was necessary for robust TGF-ß expression in DCs, whereas Smad2 negatively regulated TGF-ß expression. Smad2-deficient DCs expressed higher concentrations of TGF-ß and were tolerogenic for colitis models. This study reveals a novel mechanism of TGF-ß induction by Clostridia through a cooperation between TLR2-AP-1 and TGF-ß-Smad signaling pathways.


Subject(s)
Clostridium butyricum/immunology , Dendritic Cells/immunology , Smad2 Protein/genetics , Smad3 Protein/genetics , Transforming Growth Factor beta1/biosynthesis , Animals , Clostridium Infections/immunology , Clostridium Infections/microbiology , Colitis/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Intestines/immunology , Intestines/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucous Membrane/cytology , Mucous Membrane/immunology , Promoter Regions, Genetic/genetics , T-Lymphocytes, Regulatory/immunology , Toll-Like Receptor 2/immunology , Transcription Factor AP-1/immunology , Transforming Growth Factor beta1/genetics
4.
Nat Immunol ; 12(5): 425-33, 2011 May.
Article in English | MEDLINE | ID: mdl-21441934

ABSTRACT

Clonal deletion of autoreactive B cells is crucial for the prevention of autoimmunity, but the signaling mechanisms that regulate this checkpoint remain undefined. Here we characterize a previously unrecognized Ca(2+)-driven pathway for activation of the kinase Erk, which was proapoptotic and biochemically distinct from Erk activation induced by diacylglycerol (DAG). This pathway required protein kinase C-δ (PKC-δ) and the guanine nucleotide-exchange factor RasGRP and depended on the concentration of the Ca(2+) sensor STIM1, which controls the magnitude of Ca(2+) entry. Developmental regulation of these proteins was associated with selective activation of the pathway in B cells prone to negative selection. This checkpoint was impaired in PKC-δ-deficient mice, which developed B cell autoimmunity. Conversely, overexpression of STIM1 conferred a competitive disadvantage to developing B cells. Our findings establish Ca(2+)-dependent Erk signaling as a critical proapoptotic pathway that mediates the negative selection of B cells.


Subject(s)
B-Lymphocytes/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Guanine Nucleotide Exchange Factors/immunology , Membrane Glycoproteins/immunology , Protein Kinase C-delta/immunology , Animals , Apoptosis/immunology , B-Lymphocytes/enzymology , Calcium Channels , Cell Line , Enzyme Activation/immunology , Flow Cytometry , Gene Expression Regulation, Developmental , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/immunology , Signal Transduction , Stromal Interaction Molecule 1
5.
Nat Immunol ; 12(11): 1119-26, 2011 Oct 02.
Article in English | MEDLINE | ID: mdl-21964606

ABSTRACT

Binding of antigen to the B cell antigen receptor (BCR) triggers both BCR signaling and endocytosis. How endocytosis regulates BCR signaling remains unknown. Here we report that BCR signaling was not extinguished by endocytosis of BCRs; instead, BCR signaling initiated at the plasma membrane continued as the BCR trafficked intracellularly with the sequential phosphorylation of kinases. Blocking the endocytosis of BCRs resulted in the recruitment of both proximal and downstream kinases to the plasma membrane, where mitogen-activated protein kinases (MAPKs) were hyperphosphorylated and the kinase Akt and its downstream target Foxo were hypophosphorylated, which led to the dysregulation of gene transcription controlled by these pathways. Thus, the cellular location of the BCR serves to compartmentalize kinase activation to regulate the outcome of signaling.


Subject(s)
B-Lymphocytes/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Compartmentation/immunology , Cells, Cultured , Endocytosis/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Phosphorylation/immunology , Protein Transport/immunology , Proto-Oncogene Proteins c-akt/immunology , Receptors, Antigen, B-Cell/immunology , Signal Transduction/immunology , Transcriptional Activation/immunology
6.
Nat Immunol ; 12(6): 544-50, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21532575

ABSTRACT

The molecular mechanisms that underlie T cell quiescence are poorly understood. Here we report that mature naive CD8(+) T cells lacking the transcription factor Foxp1 gained effector phenotype and function and proliferated directly in response to interleukin 7 (IL-7) in vitro. Foxp1 repressed expression of the IL-7 receptor α-chain (IL-7Rα) by antagonizing Foxo1 and negatively regulated signaling by the kinases MEK and Erk. Acute deletion of Foxp1 induced naive T cells to gain an effector phenotype and proliferate in lympho-replete mice. Foxp1-deficient naive CD8(+) T cells proliferated even in lymphopenic mice deficient in major histocompatibility complex class I. Our results demonstrate that Foxp1 exerts essential cell-intrinsic regulation of naive T cell quiescence, providing direct evidence that lymphocyte quiescence is achieved through actively maintained mechanisms that include transcriptional regulation.


Subject(s)
Cell Proliferation , Forkhead Transcription Factors/immunology , Repressor Proteins/immunology , T-Lymphocytes/immunology , Animals , Butadienes/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Flow Cytometry , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/drug effects , Imidazoles/pharmacology , Immunoblotting , Interleukin-7/pharmacology , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/immunology , MAP Kinase Kinase Kinases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nitriles/pharmacology , Protein Binding , Pyridines/pharmacology , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , Receptors, Interleukin-7/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
8.
J Immunol ; 207(11): 2688-2698, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34697226

ABSTRACT

Regulation of BCR signaling has important consequences for generating effective Ab responses to pathogens and preventing production of autoreactive B cells during development. Currently defined functions of Fc receptor-like (FCRL) 1 include positive regulation of BCR-induced calcium flux, proliferation, and Ab production; however, the mechanistic basis of FCRL1 signaling and its contributions to B cell development remain undefined. Molecular characterization of FCRL1 signaling shows phosphotyrosine-dependent associations with GRB2, GRAP, SHIP-1, and SOS1, all of which can profoundly influence MAPK signaling. In contrast with previous characterizations of FCRL1 as a strictly activating receptor, we discover a role for FCRL1 in suppressing ERK activation under homeostatic and BCR-stimulated conditions in a GRB2-dependent manner. Our analysis of B cells in Fcrl1 -/- mice shows that ERK suppression by FCRL1 is associated with a restriction in the number of cells surviving splenic maturation in vivo. The capacity of FCRL1 to modulate ERK activation presents a potential for FCRL1 to be a regulator of peripheral B cell tolerance, homeostasis, and activation.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/immunology , GRB2 Adaptor Protein/immunology , Membrane Proteins/immunology , Receptors, Antigen, B-Cell/immunology , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , HEK293 Cells , Humans , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout
9.
EMBO J ; 37(16)2018 08 15.
Article in English | MEDLINE | ID: mdl-30049713

ABSTRACT

T helper (Th)17 cells represent a unique subset of CD4+ T cells and are vital for clearance of extracellular pathogens including bacteria and fungi. However, Th17 cells are also involved in orchestrating autoimmunity. By employing quantitative surface proteomics, we found that the evolutionarily conserved prohibitins (PHB1/2) are highly expressed on the surface of both murine and human Th17 cells. Increased expression of PHBs at the cell surface contributed to enhanced CRAF/MAPK activation in Th17 cells. Targeting surface-expressed PHBs on Th17 cells with ligands such as Vi polysaccharide (Typhim vaccine) inhibited CRAF-MAPK pathway, reduced interleukin (IL)-17 expression and ameliorated disease pathology with an increase in FOXP3+-expressing Tregs in an animal model for multiple sclerosis (MS). Interestingly, we detected a CD4+ T cell population with high PHB1 surface expression in blood samples from MS patients in comparison with age- and sex-matched healthy subjects. Our observations suggest a pivotal role for the PHB-CRAF-MAPK signalling axis in regulating the polarization and pathogenicity of Th17 cells and unveil druggable targets in autoimmune disorders such as MS.


Subject(s)
Autoimmunity , Multiple Sclerosis/immunology , Repressor Proteins/immunology , Signal Transduction/immunology , Th17 Cells/immunology , Animals , Extracellular Signal-Regulated MAP Kinases/immunology , Forkhead Transcription Factors/immunology , HeLa Cells , Humans , Mice , Multiple Sclerosis/pathology , Prohibitins , Rickettsial Vaccines/pharmacology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th17 Cells/pathology
10.
Eur J Immunol ; 51(4): 864-878, 2021 04.
Article in English | MEDLINE | ID: mdl-33616974

ABSTRACT

Intestinal epithelial cells (IECs) are the first to encounter luminal antigens and play an active role in intestinal immune responses. We previously reported that ß-glucans, major fungal cell-wall glycans, induced chemokine secretion by IEC lines in a Dectin-1- and Syk-dependent manner. Here, we show that in contrast to ß-glucans, stimulation of IEC lines with Candida albicans and Saccharomyces cerevisiae did not induce secretion of any of the proinflammatory cytokines IL-8, CCL2, CXCL1, and GM-CSF. Commensal fungi and ß-glucans activated Syk and ERK in IEC lines. However, only ß-glucans activated p38, JNK, and the transcription factors NF-κB p65 and c-JUN, which were necessary for cytokine secretion. Furthermore, costimulation of IEC lines with ß-glucans and C. albicans yielded decreased cytokine secretion compared to stimulation with ß-glucans alone. Finally, ex vivo stimulation of human colonic mucosal explants with zymosan and C. albicans, leads to epithelial Syk and ERK phosphorylation, implying recognition of fungi and similar initial signaling pathways as in IEC lines. Lack of cytokine secretion in response to commensal fungi may reflect IECs' response to fungal glycans, other than ß-glucans, that contribute to mucosal tolerance. Skewed epithelial response to commensal fungi may impair homeostasis and contribute to intestinal inflammation.


Subject(s)
Candida albicans/immunology , Cell Wall/immunology , Epithelial Cells/immunology , Intestinal Mucosa/immunology , beta-Glucans/immunology , Caco-2 Cells , Candida albicans/metabolism , Candida albicans/physiology , Cell Line, Tumor , Cell Wall/metabolism , Cytokines/immunology , Cytokines/metabolism , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , HT29 Cells , Host-Pathogen Interactions/immunology , Humans , Intestinal Mucosa/microbiology , Phosphorylation/immunology , Syk Kinase/immunology , Syk Kinase/metabolism , Zymosan/immunology , Zymosan/metabolism , beta-Glucans/metabolism
11.
Immunity ; 38(3): 475-88, 2013 Mar 21.
Article in English | MEDLINE | ID: mdl-23453634

ABSTRACT

Follicular B cell survival requires signaling from BAFFR, a receptor for BAFF and the B cell antigen receptor (BCR). This "tonic" BCR survival signal is distinct from that induced by antigen binding and may be ligand-independent. We show that inducible inactivation of the Syk tyrosine kinase, a key signal transducer from the BCR following antigen binding, resulted in the death of most follicular B cells because Syk-deficient cells were unable to survive in response to BAFF. Genetic rescue studies demonstrated that Syk transduces BAFFR survival signals via ERK and PI3 kinase. Surprisingly, BAFFR signaling directly induced phosphorylation of both Syk and the BCR-associated Igα signaling subunit, and this Syk phosphorylation required the BCR. We conclude that the BCR and Igα may be required for B cell survival because they function as adaptor proteins in a BAFFR signaling pathway leading to activation of Syk, demonstrating previously unrecognized crosstalk between the two receptors.


Subject(s)
B-Cell Activation Factor Receptor/immunology , B-Lymphocytes/immunology , Intracellular Signaling Peptides and Proteins/immunology , Protein-Tyrosine Kinases/immunology , Receptors, Antigen, B-Cell/immunology , Signal Transduction/immunology , 3-Phosphoinositide-Dependent Protein Kinases , Animals , B-Cell Activating Factor/immunology , B-Cell Activating Factor/pharmacology , B-Cell Activation Factor Receptor/genetics , B-Cell Activation Factor Receptor/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , CD79 Antigens/immunology , CD79 Antigens/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/immunology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Flow Cytometry , Gene Expression Profiling , Immunoblotting , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Models, Immunological , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proteins/genetics , Proteins/immunology , Proteins/metabolism , RNA, Untranslated , Receptor Cross-Talk/immunology , Receptors, Antigen, B-Cell/metabolism , Signal Transduction/drug effects , Syk Kinase , Tamoxifen/pharmacology
12.
Clin Exp Immunol ; 203(1): 125-136, 2021 01.
Article in English | MEDLINE | ID: mdl-33006758

ABSTRACT

Pattern recognition receptors, such as Toll-like receptors (TLRs), play an important role in the host defense against invading microbial pathogens. Their activation must be precisely regulated, as inappropriate activation or overactivation of TLR signaling pathways may result in inflammatory disorders, such as septic shock or autoimmune diseases. TMEM106A is a type II transmembrane protein constitutively expressed in macrophages. Our current study demonstrated that TMEM106A levels were increased in macrophages upon lipopolysaccharide (LPS) stimulation, as well as in the peripheral monocytes of patients with sepsis. Tmem106a knockout mice were more sensitive to lipopolysaccharide (LPS)-induced septic shock than wild-type mice. Further experiments indicated that Tmem106a ablation enhanced the expression of CD80, CD86 and major histocompatibility complex (MHC)-II in mouse macrophages upon LPS stimulation, accompanied with up-regulation of tumor necrosis factor (TNF)-α, interleukin (IL)-6, interferon (IFN)-ß and inducible nitric oxide synthase (iNOS), indicating the activation of macrophages and polarization towards the M1 inflammatory phenotype. Moreover, elevated mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB) signaling were found to be involved in the LPS-induced inflammatory response in Tmem106a-/- macrophages. However, this effect was largely abrogated by macrophage deletion in Tmem106a-/- mice. Therefore, deficiency of Tmem106a in macrophages may enhance the M1 polarization in mice, resulting in inflammation. This suggests that TMEM106A plays an important regulatory role in maintaining macrophage homeostasis.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/immunology , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/immunology , Macrophages, Peritoneal/immunology , NF-kappa B/immunology , Tumor Suppressor Proteins/immunology , Animals , Extracellular Signal-Regulated MAP Kinases/genetics , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , MAP Kinase Signaling System/genetics , Macrophages, Peritoneal/pathology , Mice , Mice, Knockout , NF-kappa B/genetics , Tumor Suppressor Proteins/genetics
13.
Nat Immunol ; 10(3): 257-65, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19182807

ABSTRACT

The mechanisms by which cytokine signals prevent the activation and mitochondrial targeting of the proapoptotic protein Bax are unclear. Here we show, using primary human eosinophils, that in the absence of the prosurvival cytokines granulocyte-macrophage colony-stimulating factor and interleukin 5, Bax spontaneously underwent activation and initiated mitochondrial disruption. Inhibition of Bax resulted in less eosinophil apoptosis, even in the absence of cytokines. Granulocyte-macrophage colony-stimulating factor induced activation of the kinase Erk1/2, which phosphorylated Thr167 of Bax; this facilitated new interaction of Bax with the prolyl isomerase Pin1. Blockade of Pin1 led to cleavage and mitochondrial translocation of Bax and caspase activation, regardless of the presence of cytokines. Our findings indicate that Pin1 is a key mediator of prosurvival signaling and is a regulator of Bax function.


Subject(s)
Eosinophils/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Interleukin-5/immunology , Peptidylprolyl Isomerase/immunology , bcl-2-Associated X Protein/immunology , Cell Death , Cell Survival , Cells, Cultured , Eosinophils/metabolism , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-5/metabolism , Mitochondria/immunology , Mitochondria/metabolism , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/metabolism , Phosphorylation , Protein Transport , bcl-2-Associated X Protein/metabolism
14.
Nat Immunol ; 10(10): 1110-7, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19734904

ABSTRACT

Signals through the pre-B cell antigen receptor (pre-BCR) and interleukin 7 receptor (IL-7R) coordinate pre-B cell population expansion with subsequent recombination of the locus encoding immunoglobulin kappa-chain (Igk). Although many 'downstream' effectors of each receptor are known, how they integrate to mediate development has remained unclear. Here we report that pre-BCR-mediated activation of the Ras-MEK-Erk signaling pathway silenced transcription of Ccnd3 (encoding cyclin D3) and coordinated exit from the cell cycle with induction of the transcription factor E2A and the initiation of Igk recombination. IL-7R-mediated activation of the transcription factor STAT5 opposed this pathway by promoting Ccnd3 expression and concomitantly inhibiting Igk transcription by binding to the Igk intronic enhancer and preventing E2A recruitment. Our data show how pre-BCR signaling poises pre-B cells to undergo differentiation after escape from IL-7R signaling.


Subject(s)
B-Lymphocytes/cytology , Cell Cycle/immunology , Cell Differentiation/immunology , Immunoglobulin Light Chains/genetics , Signal Transduction/immunology , ras Proteins/immunology , Animals , B-Lymphocytes/immunology , Basic Helix-Loop-Helix Transcription Factors/immunology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle/genetics , Cyclin D3 , Cyclins/immunology , Cyclins/metabolism , Electrophoretic Mobility Shift Assay , Extracellular Signal-Regulated MAP Kinases/immunology , Flow Cytometry , Gene Expression Regulation/immunology , Gene Silencing/immunology , Immunoblotting , MAP Kinase Kinase Kinases/immunology , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Receptors, Interleukin-7/immunology , Receptors, Interleukin-7/metabolism , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/immunology , STAT5 Transcription Factor/metabolism , ras Proteins/genetics , ras Proteins/metabolism
15.
J Allergy Clin Immunol ; 146(6): 1387-1396.e13, 2020 12.
Article in English | MEDLINE | ID: mdl-32407837

ABSTRACT

BACKGROUND: Reduced levels of prostaglandin E2 (PGE2) contribute to aspirin-induced hypersensitivity. COX inhibitors are also frequent cofactors in anaphylaxis. Whether alterations in the PGE2 system contribute to anaphylaxis independently of COX inhibitor intake is unclear. OBJECTIVE: Our aim was to test the hypothesis that relative PGE2 deficiency predisposes to anaphylaxis. METHODS: Sera from 48 patients with anaphylaxis and 27 healthy subjects were analyzed for PGE2 levels and correlated against severity; 9α,11ß-PGF2 and PGI2 metabolites were measured for control purposes. PGE2 stabilization by 15-hydroxyprostaglandin dehydrogenase inhibitor or EP2 or EP4 receptor agonists were used in a murine model of passive systemic anaphylaxis. FcεRI-triggered mediator release was determined in bone marrow-derived cultured mast cells (MCs) and human skin-derived MCs. Signaling was studied by Western blot analysis. RESULTS: Patients with anaphylaxis were characterized by markedly reduced PGE2 levels vis-à-vis healthy subjects, whereas prostacyclin metabolite levels were diminished only weakly, and 9α,11ß-PGF2 levels conversely increased. PGE2 was negatively correlated with severity. Lower PGE2 levels and higher susceptibility to anaphylaxis were also found in C57BL/6 mice vis-à-vis in Balb/c mice. Stabilization of PGE2 level by 15-hydroxyprostaglandin dehydrogenase inhibitor protected mice against anaphylaxis. Exogenous PGE2 attenuated bone marrow-derived cultured MC activation through EP2 and EP4 receptors. EP2 and EP4 agonism also curbed FcεRI-mediated degranulation of human MCs. Mechanistically, PGE2 interfered with the phosphorylation of phospholipase C gamma-1 and extracellular signal-regulated kinase. CONCLUSIONS: Homeostatic levels of PGE2 attenuate MC activation via EP2/EP4 and protect against anaphylaxis. Relative deficiency of PGE2 predisposes to anaphylaxis in humans and mice, whereas PGE2 stabilization protects against anaphylactic reactions.


Subject(s)
Anaphylaxis/immunology , Dinoprostone/deficiency , Mast Cells/immunology , Anaphylaxis/pathology , Animals , Dinoprostone/immunology , Disease Susceptibility/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Humans , Mast Cells/pathology , Mice , Mice, Inbred BALB C , Phospholipase C gamma/immunology , Receptors, Prostaglandin E, EP2 Subtype/immunology , Receptors, Prostaglandin E, EP4 Subtype/immunology , Severity of Illness Index
16.
Int J Med Microbiol ; 310(5): 151432, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32654774

ABSTRACT

The extracellular signal-regulated kinases (ERKs) serve as important determinants of cellular signal transduction pathways, and hence may play important roles during infections. Previous work suggested that putative ERK7 of Toxoplasma gondii is required for efficient intracellular replication of the parasite. However, the antigenic and immunostimulatory properties of TgERK7 protein remain unknown. The objective of this study was to produce a recombinant TgERK7 protein in vitro and to evaluate its effect on the induction of humoral and T cell-mediated immune responses against T. gondii infection in BALB/c mice. Immunization using TgERK7 mixed with Freund's adjuvants significantly increased the ratio of CD3e+CD4+ T/CD3e+CD8a+ T lymphocytes in spleen and elevated serum cytokines (IFN-γ, IL-2, IL-4, IL-10, IL-12p70, IL-23, MCP-1, and TNF-α) in immunized mice compared to control mice. On the contrary, immunization did not induce high levels of serum IgG antibodies. Five predicted peptides of TgERK7 were synthesized and conjugated with KLH and used to analyze the antibody specificity in the sera of immunized mice. We detected a progressive increase in the antibody level only against TgERK7 peptide A (DEVDKHVLRKYD). Antibody raised against this peptide significantly decreased intracellular proliferation of T. gondii in vitro, suggesting that peptide A can potentially induce a protective antibody response. We also showed that immunization improved the survival rate of mice challenged with a virulent strain and significantly reduced the parasite cyst burden within the brains of chronically infected mice. Our data show that TgERK7-based immunization induced TgERK7 peptide A-specific immune responses that can impart protective immunity against T. gondii infection. The therapeutic potential of targeting ERK7 signaling pathway for future toxoplasmosis treatment is warranted.


Subject(s)
Antigens, Protozoan/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Toxoplasma/immunology , Toxoplasmosis, Animal/immunology , Animals , Antibodies, Protozoan/blood , Antigens, Protozoan/genetics , Cytokines/blood , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Immunity, Cellular , Immunity, Humoral , Immunization , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Peptides/chemistry , Peptides/genetics , Protein Conformation , Protozoan Vaccines/immunology , Rabbits , Recombinant Proteins/immunology , Toxoplasma/genetics
17.
Cytokine ; 136: 155268, 2020 12.
Article in English | MEDLINE | ID: mdl-32889153

ABSTRACT

Human basophils regulate allergic reactions by secreting histamine, interleukin 4 (IL-4) and IL-13 through key surface receptors FcεRI as well as IL-3R, which are constitutively expressed on basophils. IL-3/IL-3R signaling axis plays key roles in regulating the development and activation of basophils. We and others have shown that IL-3-induced surface receptors e.g. ST2, IL-17RB and IL-2 receptors regulate the biology of basophils. However, the expression and function of IL-3-induced surface proteins on human basophils remain to be elucidated. We in this study aimed to identify new basophil activation regulators by transcriptomic analysis of IL-3-stimulated basophils. Gene expression microarray analysis of IL-3-treated basophils revealed 2050 differentially expressed genes, of which 323 genes encoded surface proteins including GITR. We identified that GITR was preferentially induced by IL-3 rather than anti-IgE, IL-33, fMLP and C5a. IL-3-induced GITR was suppressed by inhibitors targeting JAK2, PI3K and MEK1/2. Stimulation of IL-3-treated basophils by GITR enhanced the expression of IL-4 and IL-13. Moreover, IgE-mediated degranulation was enhanced by GITRL in the presence of IL-3. This transcriptomic analysis of IL-3-activated basophils helps to identify novel activation regulator. IL-3-induced GITR promoted the activation of basophils, adding new evidence supporting GITR as an important player in Th2-associated immune responses.


Subject(s)
Basophils/immunology , Gene Expression Regulation/immunology , Glucocorticoid-Induced TNFR-Related Protein/immunology , Interleukin-3/immunology , MAP Kinase Signaling System/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Female , Humans , Male
18.
J Immunol ; 201(9): 2832-2841, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30275047

ABSTRACT

In addition to essential roles in protein synthesis, lysyl-tRNA synthetase (KRS) is secreted to trigger a proinflammatory function that induces macrophage activation and TNF-α secretion. KRS has been associated with autoimmune diseases such as polymyositis and dermatomyositis. In this study, we investigated the immunomodulatory effects of KRS on bone marrow-derived dendritic cells (DCs) of C57BL/6 mice and subsequent polarization of Th cells and analyzed the underlying mechanisms. KRS-treated DCs increased the expression of cell surface molecules and proinflammatory cytokines associated with DC maturation and activation. Especially, KRS treatment significantly increased production of IL-12, a Th1-polarizing cytokine, in DCs. KRS triggered the nuclear translocation of the NF-κB p65 subunit along with the degradation of IκB proteins and the phosphorylation of MAPKs in DCs. Additionally, JNK, p38, and ERK inhibitors markedly recovered the degradation of IκB proteins, suggesting the involvement of MAPKs as the upstream regulators of NF-κB in the KRS-induced DC maturation and activation. Importantly, KRS-treated DCs strongly increased the differentiation of Th1 cells when cocultured with CD4+ T cells. The addition of anti-IL-12-neutralizing Ab abolished the secretion of IFN-γ in the coculture, indicating that KRS induces Th1 cell response via DC-derived IL-12. Moreover, KRS enhanced the OVA-specific Th1 cell polarization in vivo following the adoptive transfer of OVA-pulsed DCs. Taken together, these results indicated that KRS effectively induced the maturation and activation of DCs through MAPKs/NF-κB-signaling pathways and favored DC-mediated Th1 cell response.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , Lymphocyte Activation/immunology , Lysine-tRNA Ligase/immunology , Th1 Cells/immunology , Animals , Dendritic Cells/cytology , Dendritic Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Lysine-tRNA Ligase/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/immunology , NF-kappa B/metabolism , Signal Transduction/immunology
19.
J Allergy Clin Immunol ; 143(3): 1047-1057.e8, 2019 03.
Article in English | MEDLINE | ID: mdl-30017554

ABSTRACT

BACKGROUND: Classical FcεRI-induced mast cell (MC) activation causes synthesis of arachidonic acid (AA)-derived eicosanoids (leukotriene [LT] C4, prostaglandin [PG] D2, and thromboxane A2), which mediate vascular leak, bronchoconstriction, and effector cell chemotaxis. Little is known about the significance and regulation of eicosanoid generation in response to nonclassical MC activation mechanisms. OBJECTIVES: We sought to determine the regulation and significance of MC-derived eicosanoids synthesized in response to IL-33, a cytokine critical to innate type 2 immunity. METHODS: We used an ex vivo model of mouse bone marrow-derived mast cells and an IL-33-dependent in vivo model of aspirin-exacerbated respiratory disease (AERD). RESULTS: IL-33 potently liberates AA and elicits LTC4, PGD2, and thromboxane A2 production by bone marrow-derived mast cells. Unexpectedly, the constitutive function of COX-1 is required for IL-33 to activate group IVa cytosolic phospholipase A2 with consequent AA release for synthesis of all eicosanoids, including CysLTs. In contrast, COX-1 was dispensable for FcεRI-driven CysLT production. Inhibition of COX-1 prevented IL-33-induced phosphorylation of extracellular signal-related kinase, an upstream effector of cytosolic phospholipase A2, which was restored by exogenous PGH2, implying that the effects of COX-1 required its catalytic function. Administration of a COX-1-selective antagonist to mice completely prevented the generation of both PGD2 and LTC4 in a model of AERD in which MC activation is IL-33 driven. CONCLUSIONS: MC-intrinsic COX-1 amplifies IL-33-induced activation in the setting of innate type 2 immunity and might help explain the phenomenon of therapeutic desensitization to aspirin by nonselective COX inhibitors in patients with AERD.


Subject(s)
Asthma, Aspirin-Induced/immunology , Cyclooxygenase 1/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Interleukin-33/immunology , Mast Cells/immunology , Membrane Proteins/immunology , Animals , Cells, Cultured , Cyclooxygenase 2/immunology , Cyclooxygenase Inhibitors/pharmacology , Eicosanoids/immunology , Mice, Inbred C57BL , Mice, Knockout , Phospholipases A2, Cytosolic/immunology
20.
J Immunol ; 199(9): 3063-3073, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28972092

ABSTRACT

Unlike conventional B cells, regulatory B cells exhibit immunosuppressive functions to downregulate inflammation via IL-10 production. However, the molecular mechanism regulating the production of IL-10 is not fully understood. In this study, we report the finding that activation-induced cytidine deaminase (AID) is highly upregulated in the IL-10-competent B cell (B10) cell from Innp5dfl/flAicdaCre/+ mice, whereas the 5' inositol phosphatase SHIP-1 is downregulated. Notably, SHIP-1 deficiency in AID+ B cells leads to a reduction in cell count and impaired IL-10 production by B10 cells. Furthermore, the Innp5dfl/flAicdaCre/+ mouse model shows B cell-dependent autoimmune lupus-like phenotypes, such as elevated IgG serum Abs, formation of spontaneous germinal centers, production of anti-dsDNA and anti-nuclear Abs, and the obvious deposition of IgG immune complexes in the kidney with age. We observe that these lupus-like phenotypes can be reversed by the adoptive transfer of B10 cells from control Innp5dfl/fl mice, but not from the Innp5dfl/flAicdaCre/+ mice. This finding highlights the importance of defective B10 cells in Innp5dfl/flAicdaCre/+ mice. Whereas p-Akt is significantly upregulated, MAPK and AP-1 activation is impaired in B10 cells from Innp5dfl/flAicdaCre/+ mice, resulting in the reduced production of IL-10. These results show that SHIP-1 is required for the maintenance of B10 cells and production of IL-10, and collectively suggests that SHIP-1 could be a new potential therapeutic target for the treatment of autoimmune diseases.


Subject(s)
B-Lymphocyte Subsets/immunology , Cytidine Deaminase/immunology , Interleukin-10/immunology , Lupus Erythematosus, Systemic/immunology , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/deficiency , Adoptive Transfer , Animals , B-Lymphocyte Subsets/pathology , Cytidine Deaminase/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/immunology , Germinal Center/immunology , Germinal Center/pathology , Interleukin-10/genetics , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Lupus Erythematosus, Systemic/therapy , Mice , Mice, Transgenic , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology
SELECTION OF CITATIONS
SEARCH DETAIL