Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
J Leukoc Biol ; 100(1): 155-61, 2016 07.
Article in English | MEDLINE | ID: mdl-26819317

ABSTRACT

Differences in human and mouse immune responses may partly reflect species-specific adaptations and can provide important insights into human immunity. In this study, we show that RNF144B, which encodes an E3 ubiquitin ligase, was lipopolysaccharide-inducible in primary human macrophages and in human macrophage-like THP-1 cells. In contrast, Rnf144b was not lipopolysaccharide-inducible in several mouse cell populations, including primary macrophages from C57BL/6 and BALB/c mice and RAW264.7 macrophages. Similarly, Rnf144b was not up-regulated by infection of C57BL/6 mice with Escherichia coli Although the human and mouse RNF144B genes have conserved transcription start sites, cap analysis of gene expression data confirmed that the RNF144B promoter directs transcription in human but not mouse macrophages. The human and mouse RNF144B genes are controlled by highly conserved TATA-containing promoters, but subtle differences in transcription factor binding sites may account for differential regulation. Using gene silencing, we showed that RNF144B is necessary for priming of inflammasome responses in primary human macrophages. Specifically, RNF144B promotes lipopolysaccharide-inducible IL-1b mRNA expression but does not regulate expression of several other lipopolysaccharide-inducible cytokines (e.g., interleukin-10, interferon-γ) or affect expression of inflammasome components or substrates (e.g., procaspase-1, pro-interleukin-18). Our findings thus revealed a species-specific regulatory mechanism for selective inflammasome priming in human macrophages.


Subject(s)
Gene Expression Regulation, Enzymologic/drug effects , Inflammasomes/metabolism , Interleukin-1beta/genetics , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Membrane Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Humans , Interleukin-1beta/metabolism , Macrophages/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Promoter Regions, Genetic , Transcriptional Activation
2.
Sci Rep ; 5: 16149, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26548325

ABSTRACT

Escherichia coli sequence type 131 (ST131) is a globally dominant multidrug resistant clone associated with urinary tract and bloodstream infections. Most ST131 strains exhibit resistance to multiple antibiotics and cause infections associated with limited treatment options. The largest sub-clonal ST131 lineage is resistant to fluoroquinolones, contains the type 1 fimbriae fimH30 allele and expresses an H4 flagella antigen. Flagella are motility organelles that contribute to UPEC colonisation of the upper urinary tract. In this study, we examined the specific role of H4 flagella in ST131 motility and interaction with host epithelial and immune cells. We show that the majority of H4-positive ST131 strains are motile and are enriched for flagella expression during static pellicle growth. We also tested the role of H4 flagella in ST131 through the construction of specific mutants, over-expression strains and isogenic mutants that expressed alternative H1 and H7 flagellar subtypes. Overall, our results revealed that H4, H1 and H7 flagella possess conserved phenotypes with regards to motility, epithelial cell adhesion, invasion and uptake by macrophages. In contrast, H4 flagella trigger enhanced induction of the anti-inflammatory cytokine IL-10 compared to H1 and H7 flagella, a property that may contribute to ST131 fitness in the urinary tract.


Subject(s)
Cell Movement/genetics , Escherichia coli/genetics , Flagella/genetics , Urinary Tract Infections/genetics , Adhesins, Escherichia coli/genetics , Bacterial Adhesion/genetics , Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli/pathogenicity , Fimbriae Proteins/genetics , Fluoroquinolones/administration & dosage , Humans , Interleukin-10/biosynthesis , Interleukin-10/genetics , Microbial Sensitivity Tests , Urinary Tract Infections/microbiology , beta-Lactamases/genetics
3.
Cell Microbiol ; 17(5): 730-46, 2015 May.
Article in English | MEDLINE | ID: mdl-25410299

ABSTRACT

Urinary tract infections (UTI) are among the most common infections in humans. Uropathogenic Escherichia coli (UPEC) can invade and replicate within bladder epithelial cells, and some UPEC strains can also survive within macrophages. To understand the UPEC transcriptional programme associated with intramacrophage survival, we performed host-pathogen co-transcriptome analyses using RNA sequencing. Mouse bone marrow-derived macrophages (BMMs) were challenged over a 24 h time course with two UPEC reference strains that possess contrasting intramacrophage phenotypes: UTI89, which survives in BMMs, and 83972, which is killed by BMMs. Neither of these strains caused significant BMM cell death at the low multiplicity of infection that was used in this study. We developed an effective computational framework that simultaneously separated, annotated and quantified the mammalian and bacterial transcriptomes. Bone marrow-derived macrophages responded to the two UPEC strains with a broadly similar gene expression programme. In contrast, the transcriptional responses of the UPEC strains diverged markedly from each other. We identified UTI89 genes up-regulated at 24 h post-infection, and hypothesized that some may contribute to intramacrophage survival. Indeed, we showed that deletion of one such gene (pspA) significantly reduced UTI89 survival within BMMs. Our study provides a technological framework for simultaneously capturing global changes at the transcriptional level in co-cultures, and has generated new insights into the mechanisms that UPEC use to persist within the intramacrophage environment.


Subject(s)
Escherichia coli/immunology , Escherichia coli/physiology , Gene Expression Profiling , Host-Pathogen Interactions , Macrophages/immunology , Macrophages/microbiology , Animals , Cells, Cultured , Mice , Sequence Analysis, RNA
4.
J Immunol ; 191(10): 5182-95, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24123681

ABSTRACT

The Wnt signaling network, an ancient signaling system governing ontogeny and homeostatic processes, has recently been identified to exert immunoregulatory functions in a variety of inflammatory and infectious disease settings including tuberculosis. In this study, we show that Wnt6 is expressed in granulomatous lesions in the lung of Mycobacterium tuberculosis-infected mice. We identified foamy macrophage-like cells as the primary source of Wnt6 in the infected lung and uncovered a TLR-MyD88-NF-κB-dependent mode of induction in bone marrow-derived macrophages. Analysis of Wnt6-induced signal transduction revealed a pertussis toxin-sensitive, ERK-mediated, but ß-catenin-independent induction of c-Myc, a master regulator of cell proliferation. Increased Ki-67 mRNA expression levels and enhanced thymidine incorporation in Wnt6-treated macrophage cultures demonstrate a proliferation-promoting effect on murine macrophages. Further functional studies in M. tuberculosis-infected macrophages using Wnt6 conditioned medium and Wnt6-deficient macrophages uncovered a Wnt6-dependent induction of macrophage Arginase-1 and downregulation of TNF-α. This identifies Wnt6 as a novel factor driving macrophage polarization toward an M2-like phenotype. Taken together, these findings point to an unexpected role for Wnt6 in macrophage differentiation in the M. tuberculosis-infected lung.


Subject(s)
Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins/metabolism , Toll-Like Receptor 2/metabolism , Tuberculosis, Pulmonary/metabolism , Wnt Proteins/metabolism , Animals , Arginase/biosynthesis , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Differentiation/immunology , Cell Proliferation , Culture Media, Conditioned/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Granuloma/metabolism , Ki-67 Antigen/biosynthesis , Ki-67 Antigen/genetics , Lung/metabolism , Lung/microbiology , Lung/pathology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis/immunology , Myeloid Differentiation Factor 88/genetics , Pertussis Toxin/metabolism , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/biosynthesis , Toll-Like Receptor 2/genetics , Tuberculosis, Pulmonary/immunology , Tumor Necrosis Factor-alpha/genetics , Wnt Proteins/biosynthesis , Wnt Proteins/genetics , Wnt Signaling Pathway/immunology , beta Catenin/metabolism
5.
Immunobiology ; 218(11): 1345-53, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23948647

ABSTRACT

G protein-coupled receptors (GPCRs) are among the most important targets in drug discovery. In this study, we used TaqMan Low Density Arrays to profile the full GPCR repertoire of primary human macrophages differentiated from monocytes using either colony stimulating factor-1 (CSF-1/M-CSF) (CSF-1 Mϕ) or granulocyte macrophage colony stimulating factor (GM-CSF) (GM-CSF Mϕ). The overall trend was a downregulation of GPCRs during monocyte to macrophage differentiation, but a core set of 10 genes (e.g. LGR4, MRGPRF and GPR143) encoding seven transmembrane proteins were upregulated, irrespective of the differentiating agent used. Several of these upregulated GPCRs have not previously been studied in the context of macrophage biology and/or inflammation. As expected, CSF-1 Mϕ and GM-CSF Mϕ exhibited differential inflammatory cytokine profiles in response to the Toll-like Receptor (TLR)4 agonist lipopolysaccharide (LPS). Moreover, 15 GPCRs were differentially expressed between these cell populations in the basal state. For example, EDG1 was expressed at elevated levels in CSF-1 Mϕ versus GM-CSF Mϕ, whereas the reverse was true for EDG6. 101 GPCRs showed differential regulation over an LPS time course, with 65 of these profiles being impacted by the basal differentiation state (e.g. GPRC5A, GPRC5B). Only 14 LPS-regulated GPCRs showed asynchronous behavior (divergent LPS regulation) with respect to differentiation status. Thus, the differentiation state primarily affects the magnitude of LPS-regulated expression, rather than causing major reprogramming of GPCR gene expression profiles. Several GPCRs showing differential profiles between CSF-1 Mϕ and GM-CSF Mϕ (e.g. P2RY8, GPR92, EMR3) have not been widely investigated in macrophage biology and inflammation. Strikingly, several closely related GPCRs displayed completely opposing patterns of regulation during differentiation and/or activation (e.g. EDG1 versus EDG6, LGR4 versus LGR7, GPRC5A versus GPRC5B). We propose that selective regulation of GPCR5A and GPCR5B in CSF-1 Mϕ contributes to skewing toward the M2 macrophage phenotype. Our analysis of the GPCR repertoire expressed during primary human monocyte to macrophage differentiation and TLR4-mediated activation provides a valuable new platform for conducting future functional analyses of individual GPCRs in human macrophage inflammatory pathways.


Subject(s)
Inflammation/immunology , Macrophages/metabolism , RNA, Messenger/genetics , Receptors, G-Protein-Coupled/metabolism , Cell Differentiation/immunology , Down-Regulation , Gene Expression Profiling , Gene Expression Regulation/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Lipopolysaccharides , Macrophage Activation/immunology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/immunology , Monocytes/metabolism , Receptors, G-Protein-Coupled/biosynthesis , Receptors, Lysosphingolipid/biosynthesis , Sphingosine-1-Phosphate Receptors , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/metabolism , Up-Regulation
6.
Curr Opin Microbiol ; 16(1): 100-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23403118

ABSTRACT

Urinary tract infections (UTI) are among the most common infectious diseases of humans and are the most common nosocomial infections in the developed world. It is estimated that 40-50% of women and 5% of men will develop a UTI in their lifetime, and UTI accounts for more than 1 million hospitalizations and $1.6 billion in medical expenses each year in the USA. Uropathogenic Escherichia coli (UPEC) is the primary cause of UTI. This review presents an overview of recent discoveries related to the primary virulence factors of UPEC and major innate immune responses to infection of the lower urinary tract. New and emerging themes in UPEC research are discussed in the context of the interface between host and pathogen.


Subject(s)
Escherichia coli Infections/immunology , Escherichia coli Infections/pathology , Immunity, Innate , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/immunology , Uropathogenic Escherichia coli/pathogenicity , Escherichia coli Proteins/metabolism , Humans , Urinary Tract Infections/immunology , Urinary Tract Infections/pathology , Virulence Factors/metabolism
7.
Eur J Cell Biol ; 90(6-7): 553-9, 2011.
Article in English | MEDLINE | ID: mdl-21185106

ABSTRACT

Wnt proteins are secreted, palmitoylated glycoproteins with multiple functions in cell proliferation and migration as well as tissue organization. They are best known for their role in embryonic development and tissue homeostasis. In the last years, Wnt signaling was also shown to be involved in the regulation of inflammatory processes: Wnt5a is induced in human macrophages in response to mycobacteria and conserved bacterial structures and contributes to the regulation of pro-inflammatory cytokines via its receptor Frizzled (Fzd) 5. Wnt5a is also induced in other infectious and inflammatory diseases such as tuberculosis, sepsis, psoriasis, rheumatoid arthritis and atherosclerosis. In contrast, Wnt3a, a ligand of Fzd1, is constitutively expressed by bronchial epithelial cells and mediates anti-inflammatory effects on mycobacteria-infected macrophages via the Wnt/beta-Catenin signaling pathway. This pathway suppresses the activity of GSK3beta, a well known regulator of NF-kappaB-dependent gene transcription. Here we review recent data on immunomodulatory activities of Wnt proteins. Additional experiments using exogenous Wnt homologs further support the notion that TLR/NF-kappaB and Wnt signaling are functionally interconnected.


Subject(s)
Macrophages/metabolism , Mycobacterium Infections/metabolism , Mycobacterium/metabolism , Wnt Proteins/metabolism , Animals , Humans , Macrophages/immunology , Mice , Mycobacterium Infections/blood , Mycobacterium Infections/immunology , Signal Transduction , Wnt Proteins/immunology
8.
FASEB J ; 24(11): 4599-612, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20667980

ABSTRACT

Wnt/Frizzled signaling, essential for embryonic development, has also recently been implicated in the modulation of inflammatory processes. In the current study, we observed a reciprocal regulation of the Toll-like receptor (TLR)/nuclear factor-κB (NF-κB) and the Wnt/ß-catenin pathway after aerosol infection of mice with Mycobacterium tuberculosis: whereas proinflammatory mediators were substantially increased, ß-catenin signaling was significantly reduced. A systematic screen of Fzd homologs in infected mice identified Fzd1 mRNA to be significantly up-regulated during the course of infection. In vitro infection of murine macrophages led to a strong induction of Fzd1 that was dependent on TLRs, the myeloid differentiation response gene 88 (MyD88), and a functional NF-κB pathway. Flow cytometry demonstrated an elevated Fzd1 expression on macrophages in response to M. tuberculosis that was synergistically enhanced in the presence of IFN-γ. Addition of the Fzd1 ligand Wnt3a induced Wnt/ß-catenin signaling in murine macrophages that was inhibited in the presence of a soluble Fzd1/Fc fusion protein. Furthermore, Wnt3a reduced TNF release, suggesting that Wnt3a promotes anti-inflammatory functions in murine macrophages. The current data support the notion that evolutionarily conserved Wnt/Fzd signaling is involved in balancing the inflammatory response to microbial stimulation of innate immune cells of vertebrate origin.


Subject(s)
Frizzled Receptors/immunology , Macrophages/immunology , Macrophages/microbiology , Receptors, G-Protein-Coupled/immunology , Tuberculosis/immunology , Wnt Proteins/immunology , Animals , Biomarkers/metabolism , Frizzled Receptors/genetics , Gene Expression Regulation , Ligands , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis/immunology , NF-kappa B/metabolism , RNA, Messenger/immunology , Receptors, G-Protein-Coupled/genetics , Recombinant Fusion Proteins/immunology , Signal Transduction , Tumor Necrosis Factors/immunology , Up-Regulation , Wnt Proteins/chemistry , Wnt3 Protein , Wnt3A Protein
SELECTION OF CITATIONS
SEARCH DETAIL