Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Nature ; 609(7926): 335-340, 2022 09.
Article in English | MEDLINE | ID: mdl-35853476

ABSTRACT

Adhesive pili assembled through the chaperone-usher pathway are hair-like appendages that mediate host tissue colonization and biofilm formation of Gram-negative bacteria1-3. Archaic chaperone-usher pathway pili, the most diverse and widespread chaperone-usher pathway adhesins, are promising vaccine and drug targets owing to their prevalence in the most troublesome multidrug-resistant pathogens1,4,5. However, their architecture and assembly-secretion process remain unknown. Here, we present the cryo-electron microscopy structure of the prototypical archaic Csu pilus that mediates biofilm formation of Acinetobacter baumannii-a notorious multidrug-resistant nosocomial pathogen. In contrast to the thick helical tubes of the classical type 1 and P pili, archaic pili assemble into an ultrathin zigzag architecture secured by an elegant clinch mechanism. The molecular clinch provides the pilus with high mechanical stability as well as superelasticity, a property observed for the first time, to our knowledge, in biomolecules, while enabling a more economical and faster pilus production. Furthermore, we demonstrate that clinch formation at the cell surface drives pilus secretion through the outer membrane. These findings suggest that clinch-formation inhibitors might represent a new strategy to fight multidrug-resistant bacterial infections.


Subject(s)
Acinetobacter baumannii , Cryoelectron Microscopy , Fimbriae, Bacterial , Molecular Chaperones , Acinetobacter baumannii/cytology , Acinetobacter baumannii/ultrastructure , Elasticity , Fimbriae Proteins/chemistry , Fimbriae Proteins/metabolism , Fimbriae Proteins/ultrastructure , Fimbriae, Bacterial/chemistry , Fimbriae, Bacterial/metabolism , Fimbriae, Bacterial/ultrastructure , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Molecular Chaperones/ultrastructure
2.
Mol Biol Evol ; 35(12): 2851-2861, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30325443

ABSTRACT

Adenosine deaminases (ADAs) play a pivotal role in regulating the level of adenosine, an important signaling molecule that controls a variety of cellular responses. Two distinct ADAs, ADA1 and adenosine deaminase growth factor (ADGF aka ADA2), are known. Cytoplasmic ADA1 plays a key role in purine metabolism and is widely distributed from prokaryotes to mammals. On the other hand, secreted ADGF/ADA2 is a cell-signaling protein that was thought to be present only in multicellular organisms. Here, we discovered a bacterial homologue of ADGF/ADA2. Bacterial and eukaryotic ADGF/ADA2 possess the dimerization and PRB domains characteristic for the family, have nearly identical catalytic sites, and show similar catalytic characteristics. Most surprisingly, the bacterial enzyme has a signal sequence similar to that of eukaryotic ADGF/ADA2 and is specifically secreted into the extracellular space, where it may potentially control the level of extracellular adenosine. This finding provides the first example of evolution of an extracellular eukaryotic signaling protein from a secreted bacterial analogue with identical activity and suggests a potential role of ADGF/ADA2 in bacterial communication.


Subject(s)
Adenosine Deaminase/genetics , Bacterial Proteins/genetics , Evolution, Molecular , Amino Acid Sequence , Drosophila Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Multigene Family , Phylogeny
3.
J Biol Chem ; 293(44): 17070-17080, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30228191

ABSTRACT

Adhesive pili are external component of fibrous adhesive organelles and help bacteria attach to biotic or abiotic surfaces. The biogenesis of adhesive pili via the chaperone-usher pathway (CUP) is independent of external energy sources. In the classical CUP, chaperones transport assembly-competent pilins in a folded but expanded conformation. During donor-strand exchange, pilins subsequently collapse, producing a tightly packed hydrophobic core and releasing the necessary free energy to drive fiber formation. Here, we show that pilus biogenesis in non-classical, archaic, and alternative CUPs uses a different source of conformational energy. High-resolution structures of the archaic Csu-pili system from Acinetobacter baumannii revealed that non-classical chaperones employ a short donor strand motif that is insufficient to fully complement the pilin fold. This results in chaperone-bound pilins being trapped in a substantially unfolded intermediate. The exchange of this short motif with the longer donor strand from adjacent pilin provides the full steric information essential for folding, and thereby induces a large unfolded-to-folded conformational transition to drive assembly. Our findings may inform the development of anti-adhesion drugs (pilicides) to combat bacterial infections.


Subject(s)
Acinetobacter baumannii/metabolism , Fimbriae Proteins/chemistry , Fimbriae, Bacterial/chemistry , Molecular Chaperones/metabolism , Acinetobacter baumannii/chemistry , Acinetobacter baumannii/genetics , Fimbriae Proteins/genetics , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Models, Molecular , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Protein Folding
4.
Proc Natl Acad Sci U S A ; 115(21): 5558-5563, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29735695

ABSTRACT

Acinetobacter baumannii-a leading cause of nosocomial infections-has a remarkable capacity to persist in hospital environments and medical devices due to its ability to form biofilms. Biofilm formation is mediated by Csu pili, assembled via the "archaic" chaperone-usher pathway. The X-ray structure of the CsuC-CsuE chaperone-adhesin preassembly complex reveals the basis for bacterial attachment to abiotic surfaces. CsuE exposes three hydrophobic finger-like loops at the tip of the pilus. Decreasing the hydrophobicity of these abolishes bacterial attachment, suggesting that archaic pili use tip-fingers to detect and bind to hydrophobic cavities in substrates. Antitip antibody completely blocks biofilm formation, presenting a means to prevent the spread of the pathogen. The use of hydrophilic materials instead of hydrophobic plastics in medical devices may represent another simple and cheap solution to reduce pathogen spread. Phylogenetic analysis suggests that the tip-fingers binding mechanism is shared by all archaic pili carrying two-domain adhesins. The use of flexible fingers instead of classical receptor-binding cavities is presumably more advantageous for attachment to structurally variable substrates, such as abiotic surfaces.


Subject(s)
Acinetobacter baumannii/chemistry , Adhesins, Bacterial/chemistry , Bacterial Adhesion/physiology , Bacterial Proteins/chemistry , Biofilms/growth & development , Fimbriae, Bacterial/chemistry , Molecular Chaperones/chemistry , Acinetobacter baumannii/metabolism , Adhesins, Bacterial/metabolism , Amino Acid Sequence , Bacterial Proteins/metabolism , Crystallography, X-Ray , Fimbriae, Bacterial/metabolism , Molecular Chaperones/metabolism , Phylogeny , Sequence Homology
5.
Acta Crystallogr F Struct Biol Commun ; 73(Pt 8): 450-454, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28777087

ABSTRACT

Acinetobacter baumannii is one of the most difficult Gram-negative bacteria to control and treat. This pathogen forms biofilms on hospital surfaces and medical devices using Csu pili assembled via the archaic chaperone-usher pathway. To uncover the mechanism of bacterial attachment to abiotic surfaces, it was aimed to determine the crystal structure of the pilus tip adhesin CsuE. The CsuC-CsuE chaperone-subunit pre-assembly complex was purified from the periplasm of Escherichia coli overexpressing CsuC and CsuE. Despite the high purity of the complex, no crystals could be obtained. This challenge was solved by the methylation of lysine residues. The complex was crystallized in 0.1 M bis-tris pH 5.5, 17% PEG 3350 using the hanging-drop vapour-diffusion method. The crystals diffracted to a resolution of 2.31 Šand belonged to the triclinic space group P1, with unit-cell parameters a = 53.84, b = 63.85, c = 89.25 Å, α = 74.65, ß = 79.65, γ = 69.07°. Initial phases were derived from a single anomalous diffraction experiment using a selenomethionine derivative.


Subject(s)
Acinetobacter baumannii/chemistry , Adhesins, Bacterial/chemistry , Fimbriae, Bacterial/chemistry , Lysine/chemistry , Molecular Chaperones/chemistry , Protein Subunits/chemistry , Acinetobacter baumannii/metabolism , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Amino Acid Sequence , Cloning, Molecular , Crystallization , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Lysine/metabolism , Methylation , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Polyethylene Glycols/chemistry , Protein Binding , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , X-Ray Diffraction
6.
Nat Commun ; 8: 15017, 2017 05 12.
Article in English | MEDLINE | ID: mdl-28497795

ABSTRACT

Engineering light-sensitive protein regulators has been a tremendous multidisciplinary challenge. Optogenetic regulators of MAPKs, central nodes of cellular regulation, have not previously been described. Here we present OptoJNKi, a light-regulated JNK inhibitor based on the AsLOV2 light-sensor domain using the ubiquitous FMN chromophore. OptoJNKi gene-transfer allows optogenetic applications, whereas protein delivery allows optopharmacology. Development of OptoJNKi suggests a design principle for other optically regulated inhibitors. From this, we generate Optop38i, which inhibits p38MAPK in intact illuminated cells. Neurons are known for interpreting temporally-encoded inputs via interplay between ion channels, membrane potential and intracellular calcium. However, the consequences of temporal variation of JNK-regulating trophic inputs, potentially resulting from synaptic activity and reversible cellular protrusions, on downstream targets are unknown. Using OptoJNKi, we reveal maximal regulation of c-Jun transactivation can occur at unexpectedly slow periodicities of inhibition depending on the inhibitor's subcellular location. This provides evidence for resonance in metazoan JNK-signalling circuits.


Subject(s)
Neurons/drug effects , Optogenetics/methods , Protein Kinase Inhibitors/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Avena/genetics , Avena/metabolism , COS Cells , Cells, Cultured , Chlorocebus aethiops , Drug Design , Female , HEK293 Cells , Humans , Light , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/radiation effects , Male , Neurons/metabolism , Neurons/radiation effects , Phototropins/chemistry , Phototropins/genetics , Phototropins/metabolism , Protein Kinase Inhibitors/chemistry , Rats , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Mol Microbiol ; 102(4): 593-610, 2016 11.
Article in English | MEDLINE | ID: mdl-27507539

ABSTRACT

Three pathogenic species of the genus Yersinia assemble adhesive fimbriae via the FGL-chaperone/usher pathway. Closely related Y. pestis and Y. pseudotuberculosis elaborate the pH6 antigen (Psa), which mediates bacterial attachment to alveolar cells of the lung. Y. enterocolitica, instead, assembles the homologous fimbriae Myf of unknown function. Here, we discovered that Myf, like Psa, specifically recognizes ß1-3- or ß1-4-linked galactose in glycosphingolipids, but completely lacks affinity for phosphatidylcholine, the main receptor for Psa in alveolar cells. The crystal structure of a subunit of Psa (PsaA) complexed with choline together with mutagenesis experiments revealed that PsaA has four phosphatidylcholine binding pockets that enable super-high-avidity binding of Psa-fibres to cell membranes. The pockets are arranged as six tyrosine residues, which are all missing in the MyfA subunit of Myf. Conversely, the crystal structure of the MyfA-galactose complex revealed that the galactose-binding site is more extended in MyfA, enabling tighter binding to lactosyl moieties. Our results suggest that during evolution, Psa has acquired a tyrosine-rich surface that enables it to bind to phosphatidylcholine and mediate adhesion of Y. pestis/pseudotuberculosis to alveolar cells, whereas Myf has specialized as a carbohydrate-binding adhesin, facilitating the attachment of Y. enterocolitica to intestinal cells.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Fimbriae, Bacterial/metabolism , Yersinia/metabolism , Adhesins, Bacterial/metabolism , Amino Acid Sequence , Antigens, Bacterial/genetics , Antigens, Bacterial/ultrastructure , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/ultrastructure , Binding Sites , Fimbriae Proteins/metabolism , Molecular Chaperones/metabolism , Tropism/genetics , Virulence/genetics , Yersinia enterocolitica/metabolism , Yersinia pestis/metabolism , Yersinia pseudotuberculosis/metabolism
8.
PLoS Pathog ; 11(11): e1005269, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26587649

ABSTRACT

Gram-negative pathogens express fibrous adhesive organelles that mediate targeting to sites of infection. The major class of these organelles is assembled via the classical, alternative and archaic chaperone-usher pathways. Although non-classical systems share a wider phylogenetic distribution and are associated with a range of diseases, little is known about their assembly mechanisms. Here we report atomic-resolution insight into the structure and biogenesis of Acinetobacter baumannii Csu and Escherichia coli ECP biofilm-mediating pili. We show that the two non-classical systems are structurally related, but their assembly mechanism is strikingly different from the classical assembly pathway. Non-classical chaperones, unlike their classical counterparts, maintain subunits in a substantially disordered conformational state, akin to a molten globule. This is achieved by a unique binding mechanism involving the register-shifted donor strand complementation and a different subunit carboxylate anchor. The subunit lacks the classical pre-folded initiation site for donor strand exchange, suggesting that recognition of its exposed hydrophobic core starts the assembly process and provides fresh inspiration for the design of inhibitors targeting chaperone-usher systems.


Subject(s)
Acinetobacter baumannii/metabolism , Escherichia coli Proteins/metabolism , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/metabolism , Molecular Chaperones/metabolism , Amino Acid Sequence , Crystallography, X-Ray/methods , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Phylogeny , Protein Subunits/metabolism
9.
Acta Crystallogr F Struct Biol Commun ; 71(Pt 6): 770-4, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26057810

ABSTRACT

The attachment of many Gram-negative pathogens to biotic and abiotic surfaces is mediated by fimbrial adhesins, which are assembled via the classical, alternative and archaic chaperone-usher (CU) pathways. The archaic CU fimbrial adhesins have the widest phylogenetic distribution, yet very little is known about their structure and mechanism of assembly. To elucidate the biogenesis of archaic CU systems, structural analysis of the Csu fimbriae, which are used by Acinetobacter baumannii to form stable biofilms and cause nosocomial infection, was focused on. The major fimbriae subunit CsuA/B complexed with the CsuC chaperone was purified from the periplasm of Escherichia coli cells co-expressing CsuA/B and CsuC, and the complex was crystallized in PEG 3350 solution using the hanging-drop vapour-diffusion method. Selenomethionine-labelled CsuC-CsuA/B complex was purified and crystallized under the same conditions. The crystals diffracted to 2.40 Å resolution and belonged to the hexagonal space group P6(4)22, with unit-cell parameters a = b = 94.71, c = 187.05 Å, α = ß = 90, γ = 120°. Initial phases were derived from a single anomalous diffraction (SAD) experiment using the selenomethionine derivative.


Subject(s)
Acinetobacter baumannii/chemistry , Adhesins, Bacterial/chemistry , Fimbriae, Bacterial/chemistry , Molecular Chaperones/chemistry , Protein Subunits/chemistry , Acinetobacter baumannii/metabolism , Adhesins, Bacterial/genetics , Amino Acid Sequence , Cloning, Molecular , Crystallization , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Fimbriae, Bacterial/genetics , Gene Expression , Models, Molecular , Molecular Chaperones/genetics , Molecular Sequence Data , Polyethylene Glycols/chemistry , Protein Multimerization , Protein Subunits/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Selenomethionine/chemistry , X-Ray Diffraction
10.
PLoS Pathog ; 10(9): e1004404, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25232738

ABSTRACT

Enteroaggregative Escherichia coli (EAEC) is a leading cause of acute and persistent diarrhea worldwide. A recently emerged Shiga-toxin-producing strain of EAEC resulted in significant mortality and morbidity due to progressive development of hemolytic-uremic syndrome. The attachment of EAEC to the human intestinal mucosa is mediated by aggregative adherence fimbria (AAF). Using X-ray crystallography and NMR structures, we present new atomic resolution insight into the structure of AAF variant I from the strain that caused the deadly outbreak in Germany in 2011, and AAF variant II from archetype strain 042, and propose a mechanism for AAF-mediated adhesion and biofilm formation. Our work shows that major subunits of AAF assemble into linear polymers by donor strand complementation where a single minor subunit is inserted at the tip of the polymer by accepting the donor strand from the terminal major subunit. Whereas the minor subunits of AAF have a distinct conserved structure, AAF major subunits display large structural differences, affecting the overall pilus architecture. These structures suggest a mechanism for AAF-mediated adhesion and biofilm formation. Binding experiments using wild type and mutant subunits (NMR and SPR) and bacteria (ELISA) revealed that despite the structural differences AAF recognize a common receptor, fibronectin, by employing clusters of basic residues at the junction between subunits in the pilus. We show that AAF-fibronectin attachment is based primarily on electrostatic interactions, a mechanism not reported previously for bacterial adhesion to biotic surfaces.


Subject(s)
Adhesins, Escherichia coli/immunology , Bacterial Adhesion/immunology , Escherichia coli Infections/immunology , Escherichia coli Proteins/immunology , Escherichia coli/pathogenicity , Fimbriae, Bacterial/chemistry , Host-Pathogen Interactions/immunology , Adhesins, Escherichia coli/genetics , Amino Acid Sequence , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/immunology , Escherichia coli Infections/microbiology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Fibronectins/metabolism , Humans , Immunoblotting , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Magnetic Resonance Spectroscopy , Microscopy, Fluorescence , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation/genetics , Protein Conformation , Sequence Homology, Amino Acid
11.
Article in English | MEDLINE | ID: mdl-24316837

ABSTRACT

The outbreak of Shiga toxin-producing Escherichia coli O104:H4 infection in Germany in 2011 was associated with significant mortality and morbidity owing to the progressive development of haemolytic-uraemic syndrome. The outbreak strain emerged recently as a result of horizontal transfer events leading to the acquisition of a number of virulence factors. Among them, aggregative adherence fimbriae type I (AAF/I) are considered to be particularly important since they are involved in the initial attachment of bacteria to the intestinal mucosa. Here, the crystallization and preliminary X-ray diffraction analysis of the major subunit of AAF/I, AggA, are reported. Crystallization of recombinant donor-strand complemented AggA was performed by the vapour-diffusion method. The crystals diffracted to 1.55 Å resolution and belonged to the orthorhombic space group C222(1), with unit-cell parameters a = 77.83, b = 80.17, c = 91.42 Å. Despite a low sulfur content of the protein [0.57%(w/w)], sufficiently accurate initial phases were derived from a sulfur SAD experiment.


Subject(s)
Escherichia coli Proteins/chemistry , Fimbriae Proteins/chemistry , Protein Subunits/chemistry , Shiga-Toxigenic Escherichia coli/chemistry , Sulfur/chemistry , Amino Acid Sequence , Bacterial Adhesion , Crystallization , Crystallography, X-Ray , Disease Outbreaks , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Fimbriae Proteins/genetics , Fimbriae Proteins/metabolism , Gene Expression , Germany/epidemiology , Hemolytic-Uremic Syndrome/epidemiology , Hemolytic-Uremic Syndrome/microbiology , Humans , Molecular Sequence Data , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Shiga-Toxigenic Escherichia coli/genetics , Shiga-Toxigenic Escherichia coli/metabolism , Shiga-Toxigenic Escherichia coli/pathogenicity , X-Ray Diffraction
12.
J Neurosci ; 33(19): 8185-201, 2013 May 08.
Article in English | MEDLINE | ID: mdl-23658158

ABSTRACT

Neuronal nitric oxide synthase (nNOS) and p38MAPK are strongly implicated in excitotoxicity, a mechanism common to many neurodegenerative conditions, but the intermediary mechanism is unclear. NOS1AP is encoded by a gene recently associated with sudden cardiac death, diabetes-associated complications, and schizophrenia (Arking et al., 2006; Becker et al., 2008; Brzustowicz, 2008; Lehtinen et al., 2008). Here we find it interacts with p38MAPK-activating kinase MKK3. Excitotoxic stimulus induces recruitment of NOS1AP to nNOS in rat cortical neuron culture. Excitotoxic activation of p38MAPK and subsequent neuronal death are reduced by competing with the nNOS:NOS1AP interaction and by knockdown with NOS1AP-targeting siRNAs. We designed a cell-permeable peptide that competes for the unique PDZ domain of nNOS that interacts with NOS1AP. This peptide inhibits NMDA-induced recruitment of NOS1AP to nNOS and in vivo in rat, doubles surviving tissue in a severe model of neonatal hypoxia-ischemia, a major cause of neonatal death and pediatric disability. The highly unusual sequence specificity of the nNOS:NOS1AP interaction and involvement in excitotoxic signaling may provide future opportunities for generation of neuroprotectants with high specificity.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Neurons/physiology , Nitric Oxide Synthase Type I/metabolism , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Analysis of Variance , Animals , Animals, Newborn , Brain/cytology , Calcium/metabolism , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Hypoxia/pathology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , L-Lactate Dehydrogenase/metabolism , N-Methylaspartate/pharmacology , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Nitric Oxide Synthase Type I/genetics , Peptides/pharmacology , Protein Conformation , RNA, Small Interfering/pharmacology , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/drug effects , Transfection
13.
Mol Microbiol ; 86(5): 1100-15, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23046340

ABSTRACT

Coli surface antigen 6 (CS6) is a widely expressed enterotoxigenic Escherichia coli (ETEC) colonization factor that mediates bacterial attachment to the small intestinal epithelium. CS6 is a polymer of two protein subunits CssA and CssB, which are secreted and assembled on the cell surface via the CssC/CssD chaperone usher (CU) pathway. Here, we present an atomic resolution model for the structure of CS6 based on the results of X-ray crystallographic, spectroscopic and biochemical studies, and suggest a mechanism for CS6-mediated adhesion. We show that the CssA and CssB subunits are assembled alternately in linear fibres by the principle of donor strand complementation. This type of fibre assembly is novel for CU assembled adhesins. We also show that both subunits in the fibre bind to receptors on epithelial cells, and that CssB, but not CssA, specifically recognizes the extracellular matrix protein fibronectin. Taken together, structural and functional results suggest that CS6 is an adhesive organelle of a novel type, a hetero-polyadhesin that is capable of polyvalent attachment to different receptors.


Subject(s)
Antigens, Bacterial/chemistry , Antigens, Bacterial/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Adhesins, Bacterial/metabolism , Caco-2 Cells , Crystallography, X-Ray , Enterotoxigenic Escherichia coli/chemistry , Enterotoxigenic Escherichia coli/metabolism , Fibronectins/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Protein Subunits/chemistry , Protein Subunits/metabolism , Structure-Activity Relationship
14.
BMC Cell Biol ; 10: 5, 2009 Jan 26.
Article in English | MEDLINE | ID: mdl-19171023

ABSTRACT

BACKGROUND: Nuclear localization of proteolytically formed intracellular fragment of ErbB4 receptor tyrosine kinase has been shown to promote cell survival, and nuclear localization of ErbB4 receptor has been described in human breast cancer. Tumor necrosis factor alpha converting enzyme (TACE) initiates the proteolytic cascade leading to ErbB4 intracellular domain formation. Interactions between matrix metalloproteases and heparan sulfate have been described, but the effect of cell surface heparan sulfate on TACE activity has not been previously described. RESULTS: As indicated by immunodetection of increased ErbB4 intracellular domain formation and direct enzyme activity analysis, TACE activity was substantially amplified by enzymatic removal of cell surface heparan sulfate but not chondroitin sulfate. CONCLUSION: In this communication, we suggest a novel role for cell surface heparan sulfate. Removal of cell surface heparan sulfate led to increased formation of ErbB4 intracellular domain. As ErbB4 intracellular domain has previously been shown to promote cell survival this finding may indicate a novel mechanism how HS degradation active in tumor tissue may favor cell survival.


Subject(s)
ADAM Proteins/metabolism , Cell Membrane/chemistry , ErbB Receptors/metabolism , Heparitin Sulfate/metabolism , ADAM Proteins/genetics , ADAM17 Protein , Cell Line, Tumor , ErbB Receptors/chemistry , ErbB Receptors/genetics , Humans , Protein Structure, Tertiary , Receptor, ErbB-4
15.
Viral Immunol ; 17(2): 287-97, 2004.
Article in English | MEDLINE | ID: mdl-15279706

ABSTRACT

Tissue expression in viral infection of immunological effector molecules may depend on virus structure or replication or both. We analyzed cytokine mRNA expression in the central nervous system (CNS) of Balb/c mice during viral infection with Semliki Forest virus (SFV) clones, which varied either in structure or virulence or both. Highly neurovirulent SFV4 effectively induced IFN-gamma, TNF-alpha, IL-6 and TGF-beta, but its avirulent derivative V4-opal with nsP3 arginine-476 to opal mutation, elicited only weak induction of these cytokines. Structurally different, avirulent rA774, obtained by cloning from avirulent SFV A7(74) strain, did not induce synthesis of proinflammatory Th1 or Th2 cytokines in murine CNS, but increased synthesis of TGF-beta transcripts. In contrast, structurally identical but moderately virulent rA774-arg virus with sense codon at opal position in nsP3, markedly stimulated synthesis of IFN-gamma, TNF-alpha, and IL-10 transcripts, without, however, reaching the levels elicited by lethal SFV4. The rA774-arg clone was more potent in attracting peripheral immune cells into the CNS than the completely avirulent strains. In conclusion, induction of proinflammatory cytokine mRNA in the CNS by SFV infection seemed to correlate with the rate of viral replication and was not significantly influenced by the virus envelope or nonstructural protein primary structure. The results also have relevance for development of CNS gene therapy vectors as SFV4 and A774 display differences in CNS infection characteristics.


Subject(s)
Central Nervous System/metabolism , Central Nervous System/virology , Cytokines/metabolism , Gene Expression , Semliki forest virus/physiology , Animals , Cell Line , Cytokines/genetics , Mice , Mice, Inbred BALB C , RNA, Messenger/metabolism , RNA, Viral/biosynthesis , Semliki forest virus/genetics , Viral Proteins/biosynthesis , Virus Replication
16.
J Gen Virol ; 84(Pt 6): 1525-1533, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12771422

ABSTRACT

It has been shown previously that an avirulent Semliki Forest virus (SFV) clone, rA774, engineered to carry the nsP3 gene of the virulent clone SFV4 becomes highly neurovirulent and is lethal for adult BALB/c mice. rA774, like several other alphaviruses, has an opal termination codon close to the 5' end of nsP3 (aa 469), while SFV4 has an arginine residue at this position. Mutation of the opal codon to an arginine residue increases the virulence of rA774 but does not reconstruct the severe neurovirulence of SFV4. Additionally, nsP3 amino acid sequences differ between these two strains by eight amino acids and by a deletion of seven amino acids in the C-terminal third of rA774 nsP3. This study shows that neurovirulence can be reconstituted gradually by exchanging individual amino acids and is fully retained when combinations of two nsP3 mutations, V(11)-->I and L(201)-->F, V(11)-->I and D(249)-->N, A(48)-->E and G(70)-->A or T(435)-->A and F(442)-->L, are introduced into an rA774 derivative carrying R(469). The critical role of the arginine codon for neurovirulence was confirmed further by the acquisition of a fully lethal phenotype following the introduction of R(469) into a moderately virulent rA774 recombinant carrying the SFV4 nsP1 and nsP2 genes. In conclusion, virulence determinants in SFV are distributed over a wide region of the nonstructural genes.


Subject(s)
RNA-Binding Proteins/genetics , Semliki forest virus/genetics , Viral Nonstructural Proteins/genetics , Alphavirus Infections/virology , Amino Acid Sequence , Amino Acid Substitution , Animals , Cell Line , Cricetinae , Female , Genes, Viral , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutation , Phenotype , RNA-Dependent RNA Polymerase/genetics , Semliki forest virus/enzymology , Semliki forest virus/pathogenicity , Sequence Deletion , Sequence Homology, Amino Acid , Virulence/genetics
17.
J Neurovirol ; 9(1): 1-15, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12587064

ABSTRACT

Semliki Forest virus (SFV), an enveloped alphavirus of the family Togaviridae, infects a wide range of mammalian host cells. Most strains are neurotropic but differ in virulence. The authors took advantage of the nonpathogenic properties of SFV strain A7(74), cloned recently in their laboratory, and constructed a replication-proficient expression vector to target the central nervous system (CNS) for heterologous gene expression. The vector, termed VA7, was engineered to drive expression of foreign inserts through a second subgenomic promoter inserted in the viral 3' nontranslated region (NTR). Infectious virus was obtained by in vitro transcription and transfection into BHK cells, and was shown to direct synthesis of heterologous proteins in several mammalian cell lines. Although novel expression vehicle is not applicable for targeting specific cell populations within the CNS in its present form, in cultured rat hippocampal slices, VA7 encoding enhanced green fluorescent protein (EGFP) efficiently transduced pyramidal cells, interneurons, and glial cells. With prolonged time post infection, the number of EGFP-expressing neurons in hippocampal slices increased. Mice infected intraperitoneally with the recombinant virus remained completely asymptomatic but showed CNS expression of EGFP as evidenced by immunohistochemistry. SFV A7(74) is a nonintegrating virus, which gives rise to a randomly distributed, patchy infection of the adult CNS that is cleared within 10 days. With the advantage of noninvasive administration, the expression vector described in this work is thus applicable for short-term gene expression in the CNS.


Subject(s)
Genetic Vectors , Pyramidal Cells/virology , Semliki forest virus/genetics , Semliki forest virus/pathogenicity , Animals , CHO Cells , Cricetinae , Epithelial Cells/cytology , Epithelial Cells/virology , Female , Gene Expression Regulation, Viral , Glioma , Gliosarcoma , Green Fluorescent Proteins , Hippocampus/cytology , Hippocampus/virology , Indicators and Reagents/metabolism , Kidney/cytology , Luminescent Proteins/genetics , Melanoma , Mice , Mice, Inbred BALB C , Neuroblastoma , Pyramidal Cells/cytology , Rats , Semliki forest virus/growth & development , Transduction, Genetic , Tumor Cells, Cultured , Virulence , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...