Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters











Publication year range
1.
Inflammation ; 45(1): 372-386, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34792688

ABSTRACT

Intestinal inflammation is a common disease which can further lead to inflammatory bowel disease and even intestinal cancer. The increasing focus has come to the role of short-chain fatty acid (SCFA) in various bowel diseases. Hence, this study was designed to explore the specific role of SCFA in intestinal inflammation. In vivo and in vitro models of intestinal inflammation were constructed by lipopolysaccharide (LPS) injection in mice and LPS treatment on intestinal epithelial cells. A possible regulatory mechanism involving SCFA, CCAAT enhancer-binding protein beta (CEBPB), microRNA-145 (miR-145), and dual-specificity phosphatase 6 (DUSP6) in intestinal inflammation was verified by ChIP assay and dual-luciferase reporter gene assay. To evaluate the effects of SCFA on LPS-treated intestinal epithelial cells, the expression of relevant genes and inflammatory factors (IL-6, TNF-α, and IL-1ß) were determined. Last, the role of SCFA in vivo was explored through the scoring of disease activity index (DAI) and observation of colonic histology of LPS-treated mice. SCFA decreased the CEBPB expression in mouse colon tissues and small intestine epithelial cells induced by LPS. Furthermore, CEBPB could bind to the miR-145 promoter to inhibit its expression, thereby promoting the expression of DUSP6. In addition, SCFA improved the DAI, colonic histology, and the expression of serum inflammatory factors in LPS-treated mice and cells, noting that SCFA alleviated intestinal inflammation in vitro and in vivo. To sum up, SCFA inhibited DUSP6 by upregulating miR-145 through CEBPB repression and thus prevented the development of intestinal inflammation.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Colitis/metabolism , Colon/metabolism , Dual Specificity Phosphatase 6/metabolism , Fatty Acids, Volatile/metabolism , Intestinal Mucosa/metabolism , MicroRNAs/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/immunology , Colitis/immunology , Colitis/pathology , Colon/immunology , Colon/pathology , Dual Specificity Phosphatase 6/immunology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fatty Acids, Volatile/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/immunology
2.
J Immunol ; 204(5): 1214-1224, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31980574

ABSTRACT

Leukocytes are rapidly recruited to sites of inflammation via interactions with the vascular endothelium. The steroid hormone dehydroepiandrosterone (DHEA) exerts anti-inflammatory properties; however, the underlying mechanisms are poorly understood. In this study, we show that an anti-inflammatory mechanism of DHEA involves the regulation of developmental endothelial locus 1 (DEL-1) expression. DEL-1 is a secreted homeostatic factor that inhibits ß2-integrin-dependent leukocyte adhesion, and the subsequent leukocyte recruitment and its expression is downregulated upon inflammation. Similarly, DHEA inhibited leukocyte adhesion to the endothelium in venules of the inflamed mouse cremaster muscle. Importantly, in a model of lung inflammation, DHEA limited neutrophil recruitment in a DEL-1-dependent manner. Mechanistically, DHEA counteracted the inhibitory effect of inflammation on DEL-1 expression. Indeed, whereas TNF reduced DEL-1 expression and secretion in endothelial cells by diminishing C/EBPß binding to the DEL-1 gene promoter, DHEA counteracted the inhibitory effect of TNF via activation of tropomyosin receptor kinase A (TRKA) and downstream PI3K/AKT signaling that restored C/EBPß binding to the DEL-1 promoter. In conclusion, DHEA restrains neutrophil recruitment by reversing inflammation-induced downregulation of DEL-1 expression. Therefore, the anti-inflammatory DHEA/DEL-1 axis could be harnessed therapeutically in the context of inflammatory diseases.


Subject(s)
Calcium-Binding Proteins/immunology , Cell Adhesion Molecules/immunology , Dehydroepiandrosterone/pharmacology , Leukocytes/immunology , Signal Transduction/immunology , Animals , CCAAT-Enhancer-Binding Protein-beta/immunology , CD18 Antigens/immunology , Cell Adhesion/immunology , Endothelium, Vascular/immunology , Female , Gene Expression Regulation/immunology , Leukocytes/cytology , Mice , Phosphatidylinositol 3-Kinases/immunology , Promoter Regions, Genetic/immunology , Proto-Oncogene Proteins c-akt/immunology , Receptor, trkA/immunology
3.
BMC Genomics ; 20(1): 878, 2019 Nov 20.
Article in English | MEDLINE | ID: mdl-31747871

ABSTRACT

BACKGROUND: The function of Toll-like receptor 2 (TLR2) in host defense against pathogens, especially Mycobacterium tuberculosis (Mtb) is poorly understood. To investigate the role of TLR2 during mycobacterial infection, we analyzed the response of tlr2 zebrafish mutant larvae to infection with Mycobacterium marinum (Mm), a close relative to Mtb, as a model for tuberculosis. We measured infection phenotypes and transcriptome responses using RNA deep sequencing in mutant and control larvae. RESULTS: tlr2 mutant embryos at 2 dpf do not show differences in numbers of macrophages and neutrophils compared to control embryos. However, we found substantial changes in gene expression in these mutants, particularly in metabolic pathways, when compared with the heterozygote tlr2+/- control. At 4 days after Mm infection, the total bacterial burden and the presence of extracellular bacteria were higher in tlr2-/- larvae than in tlr2+/-, or tlr2+/+ larvae, whereas granuloma numbers were reduced, showing a function of Tlr2 in zebrafish host defense. RNAseq analysis of infected tlr2-/- versus tlr2+/- shows that the number of up-regulated and down-regulated genes in response to infection was greatly diminished in tlr2 mutants by at least 2 fold and 10 fold, respectively. Analysis of the transcriptome data and qPCR validation shows that Mm infection of tlr2 mutants leads to decreased mRNA levels of genes involved in inflammation and immune responses, including il1b, tnfb, cxcl11aa/ac, fosl1a, and cebpb. Furthermore, RNAseq analyses revealed that the expression of genes for Maf family transcription factors, vitamin D receptors, and Dicps proteins is altered in tlr2 mutants with or without infection. In addition, the data indicate a function of Tlr2 in the control of induction of cytokines and chemokines, such as the CXCR3-CXCL11 signaling axis. CONCLUSION: The transcriptome and infection burden analyses show a function of Tlr2 as a protective factor against mycobacteria. Transcriptome analysis revealed tlr2-specific pathways involved in Mm infection, which are related to responses to Mtb infection in human macrophages. Considering its dominant function in control of transcriptional processes that govern defense responses and metabolism, the TLR2 protein can be expected to be also of importance for other infectious diseases and interactions with the microbiome.


Subject(s)
Fish Diseases/genetics , Gene Expression Regulation, Developmental , Mycobacterium Infections, Nontuberculous/genetics , Mycobacterium Infections, Nontuberculous/veterinary , Toll-Like Receptor 2/genetics , Zebrafish/genetics , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/immunology , Chemokine CXCL11/genetics , Chemokine CXCL11/immunology , Disease Resistance/genetics , Embryo, Nonmammalian , Fish Diseases/immunology , Fish Diseases/microbiology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunity, Innate , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Larva/genetics , Larva/growth & development , Larva/immunology , Larva/microbiology , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/immunology , Macrophages/immunology , Macrophages/microbiology , Maf Transcription Factors/genetics , Maf Transcription Factors/immunology , Metabolic Networks and Pathways/genetics , Metabolic Networks and Pathways/immunology , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium marinum/immunology , Mycobacterium marinum/pathogenicity , Neutrophils/immunology , Neutrophils/microbiology , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/immunology , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/immunology , Transcriptome/immunology , Zebrafish/growth & development , Zebrafish/immunology , Zebrafish/microbiology , Zebrafish Proteins/genetics , Zebrafish Proteins/immunology
4.
BMC Res Notes ; 12(1): 717, 2019 Oct 31.
Article in English | MEDLINE | ID: mdl-31672165

ABSTRACT

OBJECTIVE: CCAAT/Enhancer Binding proteins (C/EBPs) are transcription factors involved in the regulation of a variety of cellular processes. We used the Abcam Recombinant Anti-C/EBP beta antibody (E299) to detect C/EBPß expression during myogenesis. Though the antibody is monoclonal, and the immunogen used is highly specific to C/EBPß, we identified an intense band at 23 kDa on western blot that did not correspond to any of the known isoforms of C/EBPß, or family members predicted to cross-react. Absent in myoblast cells overexpressing C/EBPß, the band was present when C/EBPß was knocked down, confirming specificity for a protein other than C/EBPß. The objective of this work was to identify the contaminating reactivity. RESULTS: We performed immunoprecipitation followed by mass spectrometry to identified myosin light chain 4 (MYL4) as the unknown band, suggesting that the Abcam monoclonal antibody directed against C/EBPß is not pure, but contains a contaminating antibody against MYL4. Caution should be used when working in cells lines that express MYL4 to not confound the detection of MYL4 with that of C/EBPß isoforms.


Subject(s)
Antibodies, Monoclonal/immunology , CCAAT-Enhancer-Binding Protein-beta/immunology , Cell Differentiation/immunology , Myoblasts/immunology , Animals , Antibody Specificity/immunology , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Differentiation/genetics , Cell Line , Cross Reactions/immunology , Gene Expression Profiling , HEK293 Cells , Humans , Mice , Muscle Development/genetics , Muscle Development/immunology , Myoblasts/cytology , Myoblasts/metabolism , Myosin Light Chains/genetics , Myosin Light Chains/immunology , Myosin Light Chains/metabolism , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/metabolism , Tandem Mass Spectrometry/methods
5.
Front Immunol ; 9: 2511, 2018.
Article in English | MEDLINE | ID: mdl-30425718

ABSTRACT

Background and Aims: In ulcerative colitis (UC), inflammation begins in the rectum and can extend proximally throughout the entire colon. The extension of inflammation is an important determinant of disease course, and may be limited by the action of regulatory T cells (Tregs). In this cross-sectional study, we evaluated the relationship between UC extension and the proportions of CD3+CD4+Foxp3+ and CD3+CD4+LAP+Foxp3-Tregs in the colonic lamina propria (LP) of 79 UC patients and 29 controls. The role of these cells in UC extension was also investigated in the murine oxazolone-induced colitis model. Methods: Patients: Disease extension was classified according to the Montreal classification. Where possible, endoscopic biopsies of involved and uninvolved tissue were obtained from UC patients. Mouse model: Colitis was induced by intrarectal oxazolone administration. Lamina propria mononuclear cells were isolated from patient biopsies and mouse colon tissue using enzymatic method and the percentage of CD3+CD4+Foxp3+ and CD3+CD4+LAP+Foxp3-cells evaluated by immunofluorescence. Confocal microscopy was applied for the visualization and quantification of CD4+LAP+ cells on tissue histological sections. Results: In UC patients with distal colitis the proportion of LP CD3+CD4+Foxp3+ Tregs was significantly higher in inflamed tissue than uninvolved tissue. As opposite, the proportion of LP CD3+CD4+LAP+ Tregs was significantly higher in uninvolved tissue than involved tissue. Both LP CD3+CD4+Foxp3+ and LP CD3+CD4+LAP+ Tregs proportion in involved tissue was significantly higher than in controls irrespective of the extension of inflammation. In mice with oxazolone-induced distal colitis, treatment with LAP-depleting antibody was associated with the development of extensive colitis. Conclusions: Our findings suggest that CD3+CD4+LAP+Foxp3-Tregs limit the extension of inflammatory lesions in UC patients.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/immunology , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/immunology , Colitis, Ulcerative/immunology , Forkhead Transcription Factors/immunology , Mucous Membrane/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Aged, 80 and over , Animals , Colitis, Ulcerative/chemically induced , Colon/immunology , Cross-Sectional Studies , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Oxazolone/pharmacology
6.
J Cell Biochem ; 119(6): 4751-4759, 2018 06.
Article in English | MEDLINE | ID: mdl-29323736

ABSTRACT

MicroRNAs (miRNAs) mediate post-transcriptional gene suppression and are a critical component of the complex regulatory networks in epithelial immune responses. Transcription of miRNA genes in epithelial cells can be elaborately controlled through Toll-like receptors (TLRs), and associated nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinase (MAPK) pathways, leading to nuclear transcription factor associated-transactivation and transrepression of miRNAs. MiRNA, let-7f is involved in the regulation of innate immune responses post TLR3 stimulation in human endocervical cells (End1/E6E7) and decreased let-7f is associated with poor immune activation. Thus, expression of let-7f is under strict control. However, the mechanism by which let-7f is regulated in these cells is not known. Therefore, in the present study, we have investigated the role of MAPK and NF-κB in the transcription of let-7f. We report that signalling of TLR3, results in activation of multiple signalling pathways including MAPK/ERK, JNK, p38, and NF-κB. Of these MAPK/ p38 and JNK directly influence the expression of let-7f in End1/E6E7 cells. Inhibition of ERK and NF-κB up regulates the expression of let-7f and its transcription factor, C/EBPß. In conclusion, we have identified a system through which TLR3 mediated immune response is regulated by C/EBPß and let-7f through the temporal activation of MAPK and NF-κB in human endocervical cells.


Subject(s)
Epithelial Cells/immunology , Gene Expression Regulation/immunology , MAP Kinase Signaling System/immunology , MicroRNAs/immunology , NF-kappa B/immunology , CCAAT-Enhancer-Binding Protein-beta/immunology , Cell Line, Transformed , Cervix Uteri , Epithelial Cells/cytology , Female , Humans , Toll-Like Receptor 3/immunology
7.
Virus Res ; 244: 90-98, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29141203

ABSTRACT

Human adenovirus (HAdV) type 36 seropositivity has been linked to obesity in humans. That link is supported by a small number of studies using HAdV-36 infection of animals that are not natural hosts for HAdVs. In this study, we infected mice with mouse adenovirus type 1 (MAV-1), a mouse pathogen, to determine whether MAV-1 infected adipose tissue and was associated with adipose tissue inflammation and obesity. We detected MAV-1 in adipose tissue during acute MAV-1 infection, but we did not detect virus-induced increases in adipose tissue cytokine expression or histological evidence of adipose tissue inflammation during acute infection. MAV-1 did not persist in adipose tissue at later times, and we did not detect long-term adipose inflammation, increased adipose tissue mass, or body weight in infected mice. Our data indicate that MAV-1 is not associated with obesity in infected mice.


Subject(s)
Adenoviridae Infections/virology , Adipose Tissue/virology , DNA, Viral/genetics , Gene Expression Regulation/immunology , Host-Pathogen Interactions , Mastadenovirus/genetics , Adenoviridae Infections/genetics , Adenoviridae Infections/immunology , Adipose Tissue/immunology , Animals , Body Weight , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/immunology , Cell Line , Chemokine CCL2/genetics , Chemokine CCL2/immunology , DNA, Viral/immunology , Female , Fibroblasts/virology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Male , Mastadenovirus/growth & development , Mastadenovirus/metabolism , Mice , Mice, Inbred C57BL , PPAR gamma/genetics , PPAR gamma/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Virus Replication
8.
Mol Immunol ; 91: 165-172, 2017 11.
Article in English | MEDLINE | ID: mdl-28934717

ABSTRACT

Sepsis-induced myeloid-derived suppressor cells (MDSCs) contribute to immunosuppression associated with sepsis. We reported that the CCAAT enhancer-binding protein C/EBPß activates microRNA (miR)-21 and miR-181b expressions, which induce transcription factor NFI-A to support the generation and expansion of MDSCs in the bone marrow and spleens of septic mice. Here, using a conditional knockout mouse model lacking C/EBPß in the myeloid lineage, we find that without C/EBPß, myeloid progenitor cells could not express miR-21 or miR-181b, and ectopic expression of C/EBPß in the C/EBPß-deficient myeloid progenitors activated the expression of the two miRNAs. Moreover, C/EBPß-reconstituted myeloid cells expressed IL-10 and reduced T cell proliferation and function, similar to control MDSCs that express C/EBPß. Exogenous expression of miR-21 and miR-181b in the C/EBPß-deficient myeloid progenitors from septic mice produced similar results. Notably, NFI-A-dependent transactivation of NF-kB MDSC generating pathway was reversed in the C/EBPß-deficient myeloid progenitors from septic mice. Together, these results support that decreasing C/EBPß expression prevents MDSC generation and decreases immunosuppression in septic mice, providing a target for sepsis treatment.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/immunology , Gene Expression Regulation/immunology , Immune Tolerance , Interleukin-10/immunology , Myeloid Progenitor Cells/immunology , Sepsis/immunology , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Proliferation/genetics , Gene Expression Regulation/genetics , Interleukin-10/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/immunology , Myeloid Progenitor Cells/pathology , NF-kappa B/genetics , NF-kappa B/immunology , NFI Transcription Factors/genetics , NFI Transcription Factors/immunology , Sepsis/genetics , Sepsis/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transcriptional Activation/immunology
9.
Infect Immun ; 85(11)2017 11.
Article in English | MEDLINE | ID: mdl-28784931

ABSTRACT

Alcohol abuse impairs immune defense. To study the effect of chronic-plus-binge alcohol exposure on the granulopoietic response, acute alcohol intoxication (intraperitoneal injection of 5 g alcohol/kg body weight) was introduced to mice chronically fed on the Lieber-DeCarli low-fat liquid alcohol diet for 5 weeks. Bacteremia was induced by intravenous injection of Escherichia coli Bacteremia caused a remarkable increase in marrow lin- c-kit+ Sca-1+ cells. Activation of cell proliferation supported the increase in marrow lin- c-kit+ Sca-1+ cells. Alcohol administration inhibited this activation of lin- c-kit+ Sca-1+ cells. The bone marrow of pair-fed control mice receiving intraperitoneal saline stored a large number of mature granulocytes expressing a high level of Gr1 (Gr1hi cells). The proportion of Gr1hi cells and the total number of Gr1+ cells were markedly reduced in the bone marrow, along with an increase in the ratio of Gr1+ granulocytes in peripheral white blood cells following bacteremia. E. coli infection stimulated proliferation of granulopoietic precursor cells, resulting in a marked increase in the ratio of immature Gr1lo cells in the bone marrow. Alcohol administration itself triggered marrow release of Gr1+ cells, resulting in reduction of the marrow granulocyte reserve with an elevation of granulocytes in the circulation. Alcohol also impaired activation of granulopoietic precursor proliferation following bacteremia. Alcohol disrupted lipopolysaccharide (LPS)-TLR4-ERK1/2-cyclin D1 signaling and inhibited upregulation of Sca-1 and C/EBPß expression by lineage-negative marrow cells in response to bacteremia. These results indicate that chronic-plus-binge alcohol exposure inhibits the granulopoietic response by disrupting key cell signaling for hematopoietic precursor cell activation and commitment to granulocyte lineage development.


Subject(s)
Bacteremia/immunology , Binge Drinking/immunology , Escherichia coli Infections/immunology , Ethanol/pharmacology , Gene Expression Regulation/drug effects , Hematopoiesis/drug effects , Signal Transduction/drug effects , Animals , Antigens, Ly/genetics , Antigens, Ly/immunology , Bacteremia/genetics , Bacteremia/pathology , Binge Drinking/genetics , Binge Drinking/pathology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/immunology , Cyclin D1/genetics , Cyclin D1/immunology , Disease Models, Animal , Escherichia coli/growth & development , Escherichia coli/immunology , Escherichia coli Infections/genetics , Escherichia coli Infections/pathology , Gene Expression Regulation/immunology , Granulocytes/drug effects , Granulocytes/immunology , Granulocytes/pathology , Hematopoiesis/genetics , Hematopoiesis/immunology , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Nucleotidyltransferases/deficiency , Nucleotidyltransferases/genetics , Nucleotidyltransferases/immunology , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
10.
PLoS Pathog ; 13(5): e1006410, 2017 May.
Article in English | MEDLINE | ID: mdl-28558034

ABSTRACT

For efficient clearance of Mycobacterium tuberculosis (Mtb), macrophages tilt towards M1 polarization leading to the activation of transcription factors associated with the production of antibacterial effector molecules such as nitric oxide (NO) and proinflammatory cytokines such as interleukin 1 ß (IL-1ß) and tumor necrosis factor α (TNF-α). At the same time, resolution of inflammation is associated with M2 polarization with increased production of arginase and cytokines such as IL-10. The transcriptional and post-transcriptional mechanisms that govern the balance between M1 and M2 polarization, and bacteria-containing processes such as autophagy and trafficking of Mtb to lysosomes, are incompletely understood. Here we report for the first time, that the transcription factor KLF4 is targeted by microRNA-26a (miR-26a). During Mtb infection, downregulation of miR-26a (observed both ex vivo and in vivo) facilitates upregulation of KLF4 which in turn favors increased arginase and decreased iNOS activity. We further demonstrate that KLF4 prevents trafficking of Mtb to lysosomes. The CREB-C/EBPß signaling axis also favors M2 polarization. Downregulation of miR-26a and upregulation of C/ebpbeta were observed both in infected macrophages as well as in infected mice. Knockdown of C/ebpbeta repressed the expression of selected M2 markers such as Il10 and Irf4 in infected macrophages. The importance of these pathways is substantiated by observations that expression of miR-26a mimic or knockdown of Klf4 or Creb or C/ebpbeta, attenuated the survival of Mtb in macrophages. Taken together, our results attribute crucial roles for the miR-26a/KLF4 and CREB-C/EBPßsignaling pathways in regulating the survival of Mtb in macrophages. These studies expand our understanding of how Mtb hijacks host signaling pathways to survive in macrophages, and open up new exploratory avenues for host-targeted interventions.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/immunology , CREB-Binding Protein/immunology , Kruppel-Like Transcription Factors/immunology , Lysosomes/microbiology , Macrophages/immunology , MicroRNAs/immunology , Mycobacterium tuberculosis/physiology , Tuberculosis/immunology , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CREB-Binding Protein/genetics , Cell Polarity , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Lysosomes/genetics , Lysosomes/immunology , Macrophages/cytology , Macrophages/microbiology , Mice , MicroRNAs/genetics , Mycobacterium tuberculosis/immunology , RAW 264.7 Cells , Signal Transduction , Tuberculosis/genetics , Tuberculosis/microbiology , Tuberculosis/physiopathology
11.
J Leukoc Biol ; 102(2): 191-200, 2017 08.
Article in English | MEDLINE | ID: mdl-28476751

ABSTRACT

Sepsis inflammation accelerates myeloid cell generation to compensate for rapid mobilization of the myeloid progenitors from bone marrow. This inflammation-driven myelopoiesis, however, generates myeloid progenitors with immunosuppressive functions that are unable to differentiate into mature, innate immune cells. The myeloid-derived suppressor cells (MDSCs) expand markedly in the later phases of sepsis, suppress both innate and adaptive immunity, and thus, elevate mortality. Using a murine model with myeloid-restricted deletion of the C/EBPß transcription factor, we show that sepsis-induced generation of MDSCs depends on C/EBPß. C/EBPß myeloid cell-deficient mice did not generate MDSCs or develop immunosuppression and survived sepsis. However, septic mice still generated Gr1+CD11b+ myeloid progenitors at the steady-state levels similar to the control sham mice, suggesting that C/EBPß is not involved in healthy, steady-state myelopoiesis. C/EBPß-deficient Gr1+CD11b+ cells generated fewer monocyte- and granulocyte-like colonies than control mice did, indicating reduced proliferation potential, but differentiated normally in response to growth factors. Adoptive transfer of C/EBPß-deficient Gr1+CD11b+ cells from late septic mice exacerbated inflammation in control mice undergoing early sepsis, confirming they were not immunosuppressive. These results show that C/EBPß directs a switch from proinflammatory to repressor myeloid cells and identifies a novel treatment target.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/immunology , Immune Tolerance/immunology , Myeloid-Derived Suppressor Cells/immunology , Sepsis/immunology , Animals , Blotting, Western , Cell Differentiation/immunology , Disease Models, Animal , Flow Cytometry , Inflammation/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Real-Time Polymerase Chain Reaction
12.
Pharmacol Res ; 121: 33-41, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28428115

ABSTRACT

Asthma and chronic obstructive pulmonary disease (COPD) are common chronic lung inflammatory diseases. Thrombin and interleukin (IL)-8/C-X-C chemokine ligand 8 (CXCL8) play critical roles in lung inflammation. Our previous study showed that c-Src-dependent IκB kinase (IKK)/IκBα/nuclear factor (NF)-κB and mitogen-activated protein kinase kinase kinase 1 (MEKK1)/extracellular signal-regulated kinase (ERK)/ribosomal S6 protein kinase (RSK)-dependent CAAT/enhancer-binding protein ß (C/EBPß) activation are involved in thrombin-induced IL-8/CXCL8 expression in human lung epithelial cells. In this study, we aimed to investigate the roles of p300 and C/EBPß-reliant IKKß expression in thrombin-induced IL-8/CXCL8 expression. Thrombin-induced increases in IL-8/CXCL8-luciferase activity and IL-8/CXCL8 release were inhibited by p300 small interfering (siRNA). Thrombin-caused histone H3 acetylation was attenuated by p300 siRNA. Stimulation of cells with thrombin for 12h resulted in increases in IKKß expression and phosphorylation in human lung epithelial cells. However, thrombin did not affect p65 expression. Moreover, 12h of thrombin stimulation produced increases in IKKß expression and phosphorylation, and IκBα phosphorylation, which were inhibited by C/EBPß siRNA. Finally, treatment of cells with thrombin caused increases in p300 and C/EBPß complex formation, p65 and C/EBPß complex formation, and recruitment of p300, p65, and C/EBPß to the IL-8/CXCL8 promoter. These results imply that p300-dependent histone H3 acetylation and C/EBPß-regulated IKKß expression contribute to thrombin-induced IL-8/CXCL8 expression in human lung epithelial cells. Results of this study will help clarify C/EBPß signaling pathways involved in thrombin-induced IL-8/CXCL8 expression in human lung epithelial cells.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/immunology , E1A-Associated p300 Protein/immunology , I-kappa B Kinase/genetics , Inflammation/immunology , Interleukin-8/genetics , Respiratory Mucosa/immunology , Thrombin/immunology , Cell Line , Epithelial Cells/cytology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Gene Expression Regulation , Humans , Inflammation/genetics , Lung/cytology , Lung/immunology , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism
13.
Mol Immunol ; 85: 89-99, 2017 05.
Article in English | MEDLINE | ID: mdl-28214650

ABSTRACT

Increasing evidences indicate that 2-Methoxyestradiol (2ME2) plays an essential role in protecting against inflammatory responses. However, its effect on IgG immune complex (IC)-induced acute lung injury (ALI) remains enigmatic. In the study, by using i.p. administration of 2ME2, we evaluated its influence on IgG IC-induced pulmonary injury in mice. We found that during IgG IC-induced ALI, mice treated by 2ME2 displayed a substantial decrease in vascular permeability and neutrophil influx (represented by myeloperoxidase activity) when compared with their counterparts receiving vehicle treatment. Furthermore, 2ME2 treatment significantly decreased pro-inflammatory mediator production and inflammatory cell, especially neutrophil accumulation in bronchoalveolar lavage fluids (BALFs) upon IgG IC stimulation. In vitro, IgG IC-triggered inflammatory mediator production was markedly down-regulated by 2ME2 in macrophages. Moreover, we verified that the activation of the transcription factors, NF-κB and CCAAT/enhancer-binding protein (C/EBP) ß, were inhibited by 2ME2 in IgG IC-challenged macrophages. We demonstrated that alleviation of NF-κB-dependent transcription might be associated with reduced phosphorylation of NF-κB p65, and reduction of C/EBP activation was directly linked to its expression. In addition, we discovered that IgG IC-stimulated phosphorylation of both p38 MAPK and ERK1/2 was alleviated by 2ME2. These data indicated a novel strategy for blockade of IgG IC-induced inflammatory activities.


Subject(s)
Acute Lung Injury/metabolism , CCAAT-Enhancer-Binding Protein-beta/drug effects , Estradiol/analogs & derivatives , Macrophages, Alveolar/drug effects , NF-kappa B/drug effects , 2-Methoxyestradiol , Acute Lung Injury/immunology , Animals , Antigen-Antibody Complex/toxicity , Blotting, Western , CCAAT-Enhancer-Binding Protein-beta/immunology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Disease Models, Animal , Estradiol/pharmacology , Immunoglobulin G/toxicity , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/immunology , NF-kappa B/metabolism , Real-Time Polymerase Chain Reaction
14.
Cytokine ; 92: 24-32, 2017 04.
Article in English | MEDLINE | ID: mdl-28088614

ABSTRACT

The CCAAT/Enhancer Binding Protein ß (C/EBPß) transcription factor is activated by multiple inflammatory stimuli, including IL-17 and LPS, and C/EBPß itself regulates numerous genes involved in inflammation. However, the role of C/EBPß in driving autoimmunity is not well understood. Here, we demonstrate that Cebpb-/- mice are resistant to EAE. Cebpb-/- mice exhibited reduced lymphocyte and APC infiltration into CNS following EAE induction. Furthermore, MOG-induced Th17 cytokine production was impaired in draining LN, indicating defects in Th17 cell priming. In vitro Th17 polarization studies indicated that T cell responses are not inherently defective, instead supporting the known roles for C/EBPß in myeloid lineage cell activation as the likely mechanism for defective Th17 priming in vivo. However, we did uncover an unexpected role for C/EBPß in regulating ll23r expression in APCs. ChIP assays confirmed that C/EBPß binds directly to the Il23r gene promoter in dendritic cells and Th17 cells. These data establish C/EBPß as a key driver of autoimmune inflammation in EAE, and propose a novel role for C/EBPß in regulation of IL-23R expression.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/immunology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Expression Regulation/immunology , Th17 Cells/immunology , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation/genetics , Mice , Mice, Knockout , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , Th17 Cells/pathology
15.
Int J Mol Sci ; 17(4): 535, 2016 Apr 08.
Article in English | MEDLINE | ID: mdl-27070591

ABSTRACT

miR-155 (microRNA-155) is an important non-coding RNA in regulating host crucial biological regulators. However, its regulatory function in mycobacterium infection remains unclear. Our study demonstrates that miR-155 expression is significantly increased in macrophages after Mycobacterium marinum (M.m) infection. Transfection with anti-miR-155 enhances nitric oxide (NO) synthesis and decreases the mycobacterium burden, and vice versa, in interferon γ (IFN-γ) activated macrophages. More importantly, miR-155 can directly bind to the 3'UTR of CCAAT/enhancer binding protein ß (C/EBPß), a positive transcriptional regulator of nitric oxide synthase (NOS2), and regulate C/EBPß expression negatively. Knockdown of C/EBPß inhibit the production of nitric oxide synthase and promoted mycobacterium survival. Collectively, these data suggest that M.m-induced upregulation of miR-155 downregulated the expression of C/EBPß, thus decreasing the production of NO and promoting mycobacterium survival, which may provide an insight into the function of miRNA in subverting the host innate immune response by using mycobacterium for its own profit. Understanding how miRNAs partly regulate microbicidal mechanisms may represent an attractive way to control tuberculosis infectious.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/immunology , Interferon-gamma/immunology , MicroRNAs/immunology , Mycobacterium Infections/immunology , Mycobacterium/immunology , Nitric Oxide/immunology , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , Cells, Cultured , Gene Expression Regulation , HEK293 Cells , Humans , Immunity, Innate , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Mycobacterium Infections/genetics , Mycobacterium Infections/microbiology , RAW 264.7 Cells
16.
Nat Commun ; 6: 8272, 2015 Sep 16.
Article in English | MEDLINE | ID: mdl-26374165

ABSTRACT

Del-1 is an endothelial cell-secreted anti-inflammatory protein. In humans and mice, Del-1 expression is inversely related to that of IL-17, which inhibits Del-1 through hitherto unidentified mechanism(s). Here we show that IL-17 downregulates human endothelial cell expression of Del-1 by targeting a critical transcription factor, C/EBPß. Specifically, IL-17 causes GSK-3ß-dependent phosphorylation of C/EBPß, which is associated with diminished C/EBPß binding to the Del-1 promoter and suppressed Del-1 expression. This inhibitory action of IL-17 can be reversed at the GSK-3ß level by PI3K/Akt signalling induced by D-resolvins. The biological relevance of this regulatory network is confirmed in a mouse model of inflammatory periodontitis. Intriguingly, resolvin-D1 (RvD1) confers protection against IL-17-driven periodontal bone loss in a Del-1-dependent manner, indicating an RvD1-Del-1 axis against IL-17-induced pathological inflammation. The dissection of signalling pathways regulating Del-1 expression provides potential targets to treat inflammatory diseases associated with diminished Del-1 expression, such as periodontitis and multiple sclerosis.


Subject(s)
Alveolar Bone Loss/immunology , CCAAT-Enhancer-Binding Protein-beta/immunology , Carrier Proteins/immunology , Glycogen Synthase Kinase 3/immunology , Interleukin-17/immunology , Periodontitis/immunology , Alveolar Bone Loss/genetics , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Calcium-Binding Proteins , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Adhesion Molecules , Chromatin Immunoprecipitation , Disease Models, Animal , Docosahexaenoic Acids/pharmacology , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Gingiva/metabolism , Glycogen Synthase Kinase 3/drug effects , Glycogen Synthase Kinase 3 beta , Human Umbilical Vein Endothelial Cells , Humans , Immunoblotting , Immunoprecipitation , Intercellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Periodontitis/genetics , Peroxidase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction
17.
Oncotarget ; 6(24): 20026-36, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26343373

ABSTRACT

Regulatory T cells (Tregs) are key players of immune regulation/dysregulation both in physiological and pathophysiological settings. Despite significant advances in understanding Treg function, there is still a pressing need to define reliable and specific markers that can distinguish different Treg subpopulations. Herein we show for the first time that markers of activated Tregs [latency associated peptide (LAP) and glycoprotein A repetitions predominant (GARP, or LRRC32)] are expressed on CD4+FoxP3- T cells expressing Helios (FoxP3-Helios+) in the steady state. Following TCR activation, GARP/LAP are up-regulated on CD4+Helios+ T cells regardless of FoxP3 expression (FoxP3+/-Helios+). We show that CD4+GARP+/-LAP+ Tregs make IL-10 immunosuppressive cytokine but not IFN-γ effector cytokine. Further characterization of FoxP3/Helios subpopulations showed that FoxP3+Helios+ Tregs proliferate in vitro significantly less than FoxP3+Helios- Tregs upon TCR stimulation. Unlike FoxP3+Helios- Tregs, FoxP3+Helios+ Tregs secrete IL-10 but not IFN-γ or IL-2, confirming they are bona fide Tregs with immunosuppressive characteristics. Taken together, Helios, and not FoxP3, is the marker of activated Tregs expressing GARP/LAP, and FoxP3+Helios+ Tregs have more suppressive characteristics, compared with FoxP3+Helios- Tregs. Our work implies that therapeutic modalities for treating autoimmune and inflammatory diseases, allergies and graft rejection should be designed to induce and/or expand FoxP3+Helios+ Tregs, while therapies against cancers or infectious diseases should avoid such expansion/induction.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/immunology , Forkhead Transcription Factors/immunology , Ikaros Transcription Factor/immunology , Membrane Proteins/immunology , T-Lymphocytes, Regulatory/immunology , CCAAT-Enhancer-Binding Protein-beta/biosynthesis , CCAAT-Enhancer-Binding Protein-beta/blood , Cells, Cultured , Forkhead Transcription Factors/blood , Humans , Ikaros Transcription Factor/blood , Interleukin-10/biosynthesis , Interleukin-10/blood , Interleukin-10/immunology , Lymphocyte Activation , Membrane Proteins/biosynthesis , Membrane Proteins/blood
18.
PLoS One ; 10(8): e0136538, 2015.
Article in English | MEDLINE | ID: mdl-26317211

ABSTRACT

Humans or mice subjected to immunosuppression, such as corticosteroids or anti-cytokine biologic therapies, are susceptible to mucosal infections by the commensal fungus Candida albicans. Recently it has become evident that the Th17/IL-17 axis is essential for immunity to candidiasis, but the downstream events that control immunity to this fungus are poorly understood. The CCAAT/Enhancer Binding Protein-ß (C/EBPß) transcription factor is important for signaling by multiple inflammatory stimuli, including IL-17. C/EBPß is regulated in a variety of ways by IL-17, and controls several downstream IL-17 target genes. However, the role of C/EBPß in vivo is poorly understood, in part because C/EBPß-deficient mice are challenging to breed and work with. In this study, we sought to understand the role of C/EBPß in the context of an IL-17-dependent immune response, using C. albicans infection as a model system. Confirming prior findings, we found that C/EBPß is required for immunity to systemic candidiasis. In contrast, C/EBPß(-/-) mice were resistant to oropharyngeal candidiasis (OPC), in a manner indistinguishable from immunocompetent WT mice. However, C/EBPß(-/-) mice experienced more severe OPC than WT mice in the context of cortisone-induced immunosuppression. Expression of the antimicrobial peptide ß-defensin (BD)-3 correlated strongly with susceptibility in C/EBPß(-/-) mice, but no other IL-17-dependent genes were associated with susceptibility. Therefore, C/EBPß contributes to immunity to mucosal candidiasis during cortisone immunosuppression in a manner linked to ß-defensin 3 expression, but is apparently dispensable for the IL-17-dependent response.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/immunology , Candida albicans/immunology , Candidiasis, Oral/immunology , Gene Expression Regulation/immunology , beta-Defensins/immunology , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , Candidiasis, Oral/genetics , Candidiasis, Oral/pathology , Interleukin-17/genetics , Interleukin-17/immunology , Mice , Mice, Knockout , beta-Defensins/genetics
19.
J Reprod Immunol ; 112: 63-7, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26276091

ABSTRACT

The pathophysiology of COX-2 expression in endometriosis is a matter of debate. The aim was to investigate the role of DNA methylation of the NF-IL6 site within the promoter of COX-2 gene in the pathogenesis of endometriosis. The endometrial tissues (ectopic and eutopic) were collected from 60 women with endometriosis and 30 women without endometriosis (control group). The methylation status of COX-2 was examined by methylation-specific PCR. Quantitative real-time PCR (RT-PCR) was performed to measure COX-2 mRNA levels in endometrial tissues. We found significantly higher levels of COX-2 in ectopic endometriotic tissue compared with eutopic tissue. Also, we found that the frequencies of methylation status of the NF-IL6 site within the COX-2 promoter in the eutopic and ectopic endometrial tissues of endometriosis groups were significantly decreased in comparison to controls (P=0.002, P=0.000 respectively). Our study demonstrated that DNA hypomethylation of the NF-IL6 site within the promoter of COX-2 gene could be a key mechanism for its elevated expression in the eutopic and ectopic tissues of endometriosis.


Subject(s)
Cyclooxygenase 2/biosynthesis , DNA Methylation , Endometriosis/enzymology , Endometrium/enzymology , Promoter Regions, Genetic , Adult , CCAAT-Enhancer-Binding Protein-beta/immunology , Cyclooxygenase 2/immunology , Endometriosis/immunology , Endometriosis/pathology , Endometriosis/surgery , Endometrium/immunology , Endometrium/pathology , Endometrium/surgery , Female , Gene Expression Regulation, Enzymologic/immunology , Humans
20.
Eur J Immunol ; 45(9): 2661-71, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26118414

ABSTRACT

Macrophages have important functions in tissue homeostasis, but the exact mechanisms regarding wide spectrum of macrophage phenotype remain unresolved. In this study, we report that mouse bone marrow derived naïve macrophages produce prostaglandin E2 (PGE2 ) endogenously, resulting in anti-inflammatory gene expression upon differentiation induced by macrophage colony stimulating factor (M-CSF). Cyclooxygenase (COX) inhibition by indomethacin reduced endogenous PGE2 production of macrophages and subsequently reduced arg1, IL10 and Mrc1, YmI and FizzI gene expressions. Of note, PGE2 phosphorylates CREB via EP2 and EP4 receptor ligation, thereby transcriptionally increasing C/EBP-ß expression in BALB/c bone marrow derived macrophages. Activated CREB directly binds to the CREB-responsive element of the C/EBP-ß promoter, such that PGE2 ultimately reinforces arg1, IL10 and Mrc1 gene expression. Cyclic AMP activator forskolin also phosphorylated CREB and induced the C/EBP-ß cascade, but this was completely blocked by the PKA inhibitor, H89. Consequently, M-CSF grown macrophages inhibited T-cell proliferation but the inhibition ability was reduced when the COX is inhibited by indomethacin or macrophage C/EBP-ß expression was decreased by siRNA transduction. Our results collectively describe the molecular basis for homeostatic macrophage differentiation by endogenous PGE2 .


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/immunology , Cyclic AMP Response Element-Binding Protein/immunology , Dinoprostone/biosynthesis , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/drug effects , Animals , Arginase/genetics , Arginase/immunology , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Differentiation/drug effects , Cell Line , Colforsin/pharmacology , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/immunology , Female , Gene Expression Regulation , Indomethacin/pharmacology , Interleukin-10/genetics , Interleukin-10/immunology , Isoquinolines/pharmacology , Macrophages/cytology , Macrophages/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Phenotype , Primary Cell Culture , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Immunologic , Signal Transduction , Sulfonamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL