Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Genome Biol ; 17(1): 201, 2016 09 29.
Article in English | MEDLINE | ID: mdl-27687735

ABSTRACT

We present a sensitive approach to predict genes expressed selectively in specific cell types, by searching publicly available expression data for genes with a similar expression profile to known cell-specific markers. Our method, CellMapper, strongly outperforms previous computational algorithms to predict cell type-specific expression, especially for rare and difficult-to-isolate cell types. Furthermore, CellMapper makes accurate predictions for human brain cell types that have never been isolated, and can be rapidly applied to diverse cell types from many tissues. We demonstrate a clinically relevant application to prioritize candidate genes in disease susceptibility loci identified by GWAS.


Subject(s)
Algorithms , Computational Biology/methods , Gene Expression Profiling/methods , Genome-Wide Association Study/methods , Animals , Brain/cytology , Brain/metabolism , Caco-2 Cells , Cell Line , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Genetic Predisposition to Disease/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Immune System/cytology , Immune System/metabolism , Mice, Inbred C57BL , Neurons/cytology , Neurons/metabolism , Polymorphism, Single Nucleotide , Reproducibility of Results , Spleen/cytology , Spleen/metabolism
2.
Mol Biol Cell ; 24(15): 2398-405, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23741050

ABSTRACT

The neonatal receptor for immunoglobulin G (IgG; FcRn) prevents IgG degradation by efficiently sorting IgG into recycling endosomes and away from lysosomes. When bound to IgG-opsonized antigen complexes, however, FcRn traffics cargo into lysosomes, where antigen processing can occur. Here we address the mechanism of sorting when FcRn is bound to multivalent IgG-opsonized antigens. We find that only the unbound receptor or FcRn bound to monomeric IgG is sorted into recycling tubules emerging from early endosomes. Cross-linked FcRn is never visualized in tubules containing the unbound receptor. Similar results are found for transferrin receptor, suggesting a general mechanism of action. Deletion or replacement of the FcRn cytoplasmic tail does not prevent diversion of trafficking to lysosomes upon cross-linking. Thus physical properties of the lumenal ligand-receptor complex appear to act as key determinants for sorting between the recycling and lysosomal pathways by regulating FcRn entry into recycling tubules.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Lysosomes/metabolism , Receptors, Fc/metabolism , Receptors, Polymeric Immunoglobulin/metabolism , Animals , Cell Line , Cross-Linking Reagents/chemistry , Endosomes/metabolism , Green Fluorescent Proteins/metabolism , Hemagglutinins/metabolism , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/genetics , Humans , Immunoglobulin G/metabolism , Mice , Protein Binding , Protein Structure, Tertiary , Protein Transport , Receptors, Fc/chemistry , Receptors, Fc/genetics , Receptors, Transferrin/metabolism , Recombinant Fusion Proteins/metabolism , beta 2-Microglobulin/metabolism
3.
Mol Microbiol ; 78(5): 1232-45, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21091507

ABSTRACT

Enteric bacterial pathogens have evolved sophisticated strategies to evade host immune defences. Some pathogens deliver anti-inflammatory effector molecules into the host cell cytoplasm via a type III secretion system (T3SS). Enteropathogenic Escherichia coli (EPEC) inhibits inflammation by an undefined, T3SS-dependent mechanism. Two proteins encoded outside of the EPEC locus of enterocyte effacement (LEE) pathogenicity island, non-LEE-encoded effector H1 (NleH1) and H2 (NleH2), display sequence similarity to Shigella flexneri OspG, which inhibits activation of the pro-inflammatory transcription factor NF-κB. We hypothesized that the anti-inflammatory effects of EPEC were mediated by NleH1 and NleH2. In this study, we examined the effect of NleH1/H2 on the NF-κB pathway. We show that NleH1/H2 are secreted via the T3SS and that transfection of cells with plasmids harbouring nleH1 or nleH2 decreased IKK-ß-induced NF-κB activity and attenuated TNF-α-induced degradation of phospho-IκBα by preventing ubiquitination. Serum KC levels were higher in mice infected with ΔnleH1H2 than those infected with WT EPEC, indicating that NleH1/H2 dampen pro-inflammatory cytokine expression. ΔnleH1H2 was cleared more rapidly than WT EPEC while complementation of ΔnleH1H2 with either NleH1 or NleH2 prolonged colonization. Together, these data show that NleH1 and NleH2 function to dampen host inflammation and facilitate EPEC colonization during pathogenesis.


Subject(s)
Enteropathogenic Escherichia coli/immunology , Escherichia coli Infections/immunology , Escherichia coli Proteins/immunology , NF-kappa B/immunology , Animals , Cell Line , Enteropathogenic Escherichia coli/genetics , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , HEK293 Cells , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/immunology , Male , Mice , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha , NF-kappa B/genetics
4.
Cell Microbiol ; 12(7): 919-29, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20088948

ABSTRACT

Enteropathogenic Escherichia coli (EPEC) infection requires the injection of effector proteins into intestinal epithelial cells (IECs) via type 3 secretion. Type 3-secreted effectors can interfere with IEC signalling pathways via specific protein-protein interactions. For example, E. coli secreted protein F (EspF) binds sorting nexin 9 (SNX9), an endocytic regulator, resulting in tubulation of the plasma membrane. Our aim was to determine the mechanism of EspF/SNX9-induced membrane tubulation. Point mutation of the SNX9 lipid binding domains or truncation of the EspF SNX9 binding domains significantly inhibited tubulation, as did inhibition of clathrin coated pit (CCP) assembly. Although characterized as non-invasive, EPEC are known to invade IECs in vitro and in vivo. Indeed, we found significant invasion of Caco-2 cells by EPEC, which, like tubulation, was blocked by pharmacological inhibition of CCPs. Interestingly, however, inhibition of dynamin activity did not prevent tubulation or EPEC invasion, which is in contrast to Salmonella invasion, which requires dynamin activity. Our data also indicate that EPEC invasion is dependent on EspF and its interaction with SNX9. Together, these findings suggest that EspF promotes EPEC invasion of IECs by harnessing the membrane-deforming activity of SNX9.


Subject(s)
Carrier Proteins/metabolism , Cell Membrane/metabolism , Enteropathogenic Escherichia coli/metabolism , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Proteins/metabolism , Vesicular Transport Proteins/metabolism , Blotting, Western , Caco-2 Cells , Carrier Proteins/genetics , Cell Membrane/drug effects , Chlorpromazine/pharmacology , Enteropathogenic Escherichia coli/drug effects , Escherichia coli Proteins/genetics , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Protein Binding/genetics , Protein Binding/physiology , Sorting Nexins , Vesicular Transport Proteins/genetics
5.
Ann N Y Acad Sci ; 1165: 169-74, 2009 May.
Article in English | MEDLINE | ID: mdl-19538303

ABSTRACT

Enteropathogenic E. coli (EPEC) are a leading cause of infantile diarrhea in developing countries, resulting in millions of deaths each year. EPEC secrete virulence factors, also called effectors, directly into host intestinal epithelial cells via type three secretion systems. Secreted effectors then affect host signaling pathways to induce several phenotypes, which ultimately lead to disease. Among the over 20 secreted effectors is E. coli secreted protein F (EspF), a 206 amino acid protein believed to be central to EPEC pathogenesis, as it disrupts tight junction structure and function. Although the mechanism by which this occurs is unknown, EspF has recently been found to contain several protein-protein interaction domains that may be involved. We have shown EspF to interact with the endocytic regulators sorting nexin 9 (SNX9) and N-WASP via non-exclusive binding sites. These interactions induce actin polymerization in vitro, and interaction with SNX9 alters its endocytic activity, as EspF induces the formation of tubular vesicles in a manner dependent upon its interaction with SNX9. EspF, therefore, appears to hijack endocytic regulation via SNX9 and possibly N-WASP interaction, to affect an as yet unidentified pathogenic phenotype.


Subject(s)
Enteropathogenic Escherichia coli/metabolism , Tight Junctions/metabolism , Amino Acid Sequence , Animals , Binding Sites , Carrier Proteins/metabolism , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Mice , Models, Biological , Molecular Sequence Data , NIH 3T3 Cells , Vesicular Transport Proteins/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism
6.
J Cell Biol ; 178(7): 1265-78, 2007 Sep 24.
Article in English | MEDLINE | ID: mdl-17893247

ABSTRACT

Bacterial toxins and effector proteins hijack eukaryotic enzymes that are spatially localized and display rapid signaling kinetics. However, the molecular mechanisms by which virulence factors engage highly dynamic substrates in the host cell environment are poorly understood. Here, we demonstrate that the enteropathogenic Escherichia coli (EPEC) type III effector protein EspF nucleates a multiprotein signaling complex composed of eukaryotic sorting nexin 9 (SNX9) and neuronal Wiskott-Aldrich syndrome protein (N-WASP). We demonstrate that a specific and high affinity association between EspF and SNX9 induces membrane remodeling in host cells. These membrane-remodeling events are directly coupled to N-WASP/Arp2/3-mediated actin nucleation. In addition to providing a biochemical mechanism of EspF function, we find that EspF dynamically localizes to membrane-trafficking organelles in a spatiotemporal pattern that correlates with SNX9 and N-WASP activity in living cells. Thus, our findings suggest that the EspF-dependent assembly of SNX9 and N-WASP represents a novel form of signaling mimicry used to promote EPEC pathogenesis and gastrointestinal disease.


Subject(s)
Carrier Proteins/metabolism , Cell Membrane/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Eukaryotic Cells/metabolism , Signal Transduction , Actin-Related Protein 2-3 Complex/metabolism , Actins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Cell Polarity , Cell Survival , Dogs , Epithelial Cells/cytology , Escherichia coli Proteins/chemistry , Evolution, Molecular , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Ligands , Mice , Molecular Sequence Data , Protein Binding , Protein Transport , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , src Homology Domains
SELECTION OF CITATIONS
SEARCH DETAIL