Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ries Langley is active.

Publication


Featured researches published by Ries Langley.


Journal of Immunology | 2005

The staphylococcal superantigen-like protein 7 binds IgA and complement C5 and inhibits IgA-Fc alpha RI binding and serum killing of bacteria.

Ries Langley; Bruce D. Wines; Natasha Willoughby; Indira Basu; Thomas Proft; John D. Fraser

The staphylococcal superantigen-like proteins (SSLs) are close relatives of the superantigens but are coded for by a separate gene cluster within a 19-kb region of the pathogenicity island SaPIn2. rSSL7 (formally known as SET1) bound with high affinity (KD, 1.1 nM) to the monomeric form of human IgA1 and IgA2 plus serum IgA from primate, pig, rat, and horse. SSL7 also bound the secretory form of IgA found in milk from human, cow, and sheep, and inhibited IgA binding to cell surface FcαRI (CD89) and to a soluble form of the FcαRI protein. In addition to IgA, SSL7 bound complement factor C5 from human (KD, 18 nM), primate, sheep, pig, and rabbit serum, and inhibited complement-mediated hemolysis and serum killing of a Gram-negative organism Escherichia coli. SSL7 is a superantigen-like protein secreted from Staphylococcus aureus that blocks IgA-FcR interactions and inhibits complement, leading to increased survival of a sensitive bacterium in blood.


Journal of Biological Chemistry | 2002

The Three-dimensional Structure of a Superantigen-like Protein, SET3, from a Pathogenicity Island of the Staphylococcus aureus Genome

Vickery L. Arcus; Ries Langley; Thomas Proft; John D. Fraser; Edward N. Baker

The staphylococcal enterotoxin-like toxins (SETs) are a family of proteins encoded within the Staphylococcus aureus genome that were identified by their similarity to the well described bacterial superantigens. The first crystal structure of a member of the SET family, SET3, has been determined to 1.9 Å (R = 0.205, R free = 0.240) and reveals a fold characteristic of the superantigen family but with significant differences. The SET proteins are secreted at varying levels by staphylococcal isolates, and seroconversion studies of normal individuals indicate that they are strongly antigenic to humans. Recombinant SETs do not exhibit any of the properties expected of superantigens such as major histocompatibility complex class II binding or broad T-cell activation, suggesting they have an entirely different function. The fact that the whole gene family is clustered within the pathogenicity island SaIn2 of theS. aureus genome suggests that they are involved in host/pathogen interactions.


Molecular Microbiology | 2007

The crystal structure of staphylococcal superantigen-like protein 11 in complex with sialyl Lewis X reveals the mechanism for cell binding and immune inhibition.

Matthew C. Chung; Bruce D. Wines; Heather M. Baker; Ries Langley; Edward N. Baker; John D. Fraser

Staphylococcus aureus is a major pathogen that produces a family of 14 staphylococcal superantigen‐like (SSL) proteins, which are structurally similar to superantigens but do not stimulate T cells. SSL11 is one member of the family that is found in all staphylococcal strains. Recombinant SSL11 bound to granulocytes and monocytes through a sialic acid‐dependent mechanism and was rapidly internalized. SSL11 also bound to sialic acid‐containing glycoproteins, such as the Fc receptor for IgA (FcαRI) and P‐selectin glycoprotein ligand‐1 (PSGL‐1), and inhibited neutrophil attachment to a P‐selectin‐coated surface. Biosensor analysis of two SSL11 alleles binding to sialyl Lewis X [sLex– Neu5Acα2‐3Galβ1‐4(Fuc1‐3)GlcNAc] coupled to bovine serum albumin gave dissociation constants of 0.7 and 7 μm respectively. Binding of SSL11 to a glycan array revealed specificity for glycans containing the trisaccharide sialyllactosamine (sLacNac – Neu5Acα2‐3Galβ1‐4GlcNAc). A 1.6 Å resolution crystal structure of SSL11 complexed with sLex revealed a discrete binding site in the C‐terminal β‐grasp domain, with predominant interactions with the sialic acid and galactose residues. A single amino acid mutation in the carbohydrate binding site abolished all SSL11 binding. Thus, SSL11 is a staphylococcal protein that targets myeloid cells by binding sialyllactosamine‐containing glycoproteins.


Journal of Immunology | 2010

Specificity of staphylococcal superantigen-like protein 10 toward the human IgG1 Fc domain.

Deepa Patel; Bruce D. Wines; Ries Langley; John D. Fraser

Staphylococcal superantigen-like protein 10 (SSL10) is a highly conserved member of the SSL family secreted by Staphylococcus aureus that displays structural but not functional similarity to superantigens. SSL10 bound to fibrinogen and fibronectin from plasma and in addition displayed striking specificity toward the γ-1 subclass of human Igs. SSL10 also bound strongly to primate IgG but not to any other species tested, including rabbit, pig, guinea pig, cow, sheep, or mouse. A soluble form of the 12-kDa β-grasp C-terminal domain of SSL10 (SSL1095–197) retained fibrinogen and fibronectin binding but lost the ability to bind IgG1, indicating that SSL10 bound to IgG1 primarily through its N-terminal oligonucleotide binding fold domain. SSL10 blocked the binding of IgG1 to FcγRs on monocytes and neutrophil phagocytosis of IgG1-opsonized bacteria. Mutagenesis of human IgG1 at key sites significantly reduced SSL10 binding including Lys322 that is important for C1q binding, a combination of Leu234 and Leu235 that are important for FcγR binding, and a combination of Lys274 and Asp276 that together are unique to IgG1. These mutations suggest that the most likely site bound by SSL10 is the outer face of the Cγ2 domain in close proximity to both the FcγR and C1q binding sites. SSL10 is a potential virulence factor for S. aureus targeting IgG1-mediated immunity.


Infection and Immunity | 2012

Structural and Functional Properties of Staphylococcal Superantigen- Like Protein 4

Stefan J. Hermans; Heather M. Baker; Richard P. Sequeira; Ries Langley; Edward N. Baker; John D. Fraser

ABSTRACT Staphylococcus aureus is a prevalent and significant human pathogen. Among the repertoire of virulence factors produced by this bacterium are the 14 staphylococcal superantigen-like (SSL) proteins. SSL protein 4 (SSL4) is one member of this family and contains a highly conserved carbohydrate binding site also found in SSL2, SSL3, SSL5, SSL6, and SSL11. Recombinant SSL4t, comprising amino acids 109 to 309 of Newman strain SSL4 (SSL4-Newman), has been shown to bind and be internalized by human granulocytes and macrophages in a sialic-acid (Sia)-dependent manner. SSL4t can compete with itself for cell binding, indicating that binding is target specific. A 2.5-Å-resolution crystal structure of SSL4t complexed with sialyl Lewis X (sLex) [sLex-Neu5Acα2-3Galβ1-4(Fucα1-3)GlcNAc] revealed a similar binding site to SSL5 and SSL11. These data, along with data on SSL4t binding to a glycan array and biosensor analysis of sLex and sialyllactosamine (sLacNac) binding are compared with those for SSL11. Although these proteins show great similarity in their carbohydrate binding sites, with a root mean square (RMS) difference between main chain atom positions of only 0.34 Å, these proteins differ in detail in their affinity for sLex and sLacNac, as well as their glycan preference. Together with cell binding data, this shows how S. aureus produces multiple related proteins that target myeloid cells through specific sialyllactosamine-containing glycoproteins.


Immunogenetics | 1997

Genomic organization, chromosomal mapping, and analysis of the 5’ promoter region of the human MAdCAM-1 gene

Euphemia Leung; Randal W. Berg; Ries Langley; John M. Greene; Lisa A. Raymond; Meena Augustus; Jian Ni; Kenneth C. Carter; Nigel K. Spurr; K. H. Andy Choo; Geoff Krissansen

Abstract MAdCAM-1, the endothelial addressin cell adhesion molecule-1, interacts preferentially with the leukocyte β7 integrin LPAM-1 (α4β7), but also with L-selectin, and with VLA-4 (α4β1) on myeloid cells, and serves to direct leukocytes into mucosal and inflamed tissues. Overlapping cosmid and phage λ genomic clones were isolated, revealing that the human MAdCAM-1 gene contains five exons where the signal peptide, two Ig domains, and mucin domain are each encoded by separate exons. The transmembrane domain, cytoplasmic domain, and 3′ untranslated region are encoded together on exon 5. The mucin domain contains eight repeats in total that are subject to alternative splicing. Despite the absence of a human counterpart of the third IgA-homologous domain and lack of sequence conservation of the mucin domain, the genomic organizations of the human and mouse MAdCAM-1 genes are similar. An alternatively spliced MAdCAM-1 variant was identified that lacks exon 4 encoding the mucin domain, and may mediate leukocyte adhesion to LPAM-1 without adhesion to the alternate receptor, L-selectin. The MAdCAM-1 gene was located at p13.3 on chromosome 19, in close proximity to the ICAM-1 and ICAM-3 genes (p13.2-p13.3). PMA-inducible promotor activity was contained in a 700 base pair 5’ flanking fragment conserved with the mouse MAdCAM-1 gene including tandem NF-kB sites, and an Sp1 site; and in addition multiple potential AP2, Adh1 (ETF), PEA3, and Sp1 sites. In summary, the data establish that the previously reported human MAdCAM-1 cDNA does indeed encode the human homologue of mouse MAdCAM-1, despite gross dissimilarities in the MAdCAM-1 C-terminal structures.


Cytogenetic and Genome Research | 1999

The integrin α10 subunit: expression pattern, partial gene structure, and chromosomal localization

K. Lehnert; J. Ni; Euphemia Leung; Sheryl M. Gough; Christine M. Morris; D. Liu; S.-X. Wang; Ries Langley; Geoffrey W. Krissansen

Herein we report the cloning of cDNAs and incompletely processed hnRNAs from endothelia and heart that encode the α10 subunit forming part of the novel collagen type II-binding integrin α10β1 of chondrocytes. Analysis of hnRNA clones and reported expressed sequence tags revealed the positions of 17 putative intron-exon splice junctions shared with those of the p150,95 (ITGAX) gene. Human α10 transcripts of 5.4 and 1.8 kb were not restricted to chondrocytes but, instead, were widely expressed in a panel of 24 tissue types, where the highest expression was found in muscle and heart. The human α10 subunit gene (ITGA10) was localized to band q21 of chromosome 1.


Journal of Biological Chemistry | 2015

Streptococcal 5'-Nucleotidase A (S5nA), a Novel Streptococcus pyogenes Virulence Factor That Facilitates Immune Evasion.

Lisa Zheng; Adrina Khemlani; Natalie Lorenz; Jacelyn M. S. Loh; Ries Langley; Thomas Proft

Background: 5′-Nucleotidases are important virulence factors found in several bacterial pathogens. Results: Streptococcal 5′-nucleotidase A (S5nA) generated immunomodulatory molecules adenosine and deoxyadenosine and rescued Lactococcus lactis in a blood killing assay. Conclusion: S5nA is a novel Streptococcus pyogenes virulence factor that facilitates immune evasion from the host. Significance: S5nA might be a target for developing new therapeutics or vaccines. Streptococcus pyogenes is an important human pathogen that causes a wide range of diseases. Using bioinformatics analysis of the complete S. pyogenes strain SF370 genome, we have identified a novel S. pyogenes virulence factor, which we termed streptococcal 5′-nucleotidase A (S5nA). A recombinant form of S5nA hydrolyzed AMP and ADP, but not ATP, to generate the immunomodulatory molecule adenosine. Michaelis-Menten kinetics revealed a Km of 169 μm and a Vmax of 7550 nmol/mg/min for the substrate AMP. Furthermore, recombinant S5nA acted synergistically with S. pyogenes nuclease A to generate macrophage-toxic deoxyadenosine from DNA. The enzyme showed optimal activity between pH 5 and pH 6.5 and between 37 and 47 °C. Like other 5′-nucleotidases, S5nA requires divalent cations and was active in the presence of Mg2+, Ca2+, or Mn2+. However, Zn2+ inhibited the enzymatic activity. Structural modeling combined with mutational analysis revealed a highly conserved catalytic dyad as well as conserved substrate and cation-binding sites. Recombinant S5nA significantly increased the survival of the non-pathogenic bacterium Lactococcus lactis during a human whole blood killing assay in a dose-dependent manner, suggesting a role as an S. pyogenes virulence factor. In conclusion, we have identified a novel S. pyogenes enzyme with 5′-nucleotidase activity and immune evasion properties.


The Comprehensive Sourcebook of Bacterial Protein Toxins (Fourth Edition) | 2015

Bacterial superantigens and superantigen-like toxins

Ries Langley; John D. Fraser; Thomas Proft

Superantigens (SAgs) are the most powerful T-cell mitogens ever discovered. By simultaneously binding to the major histocompatibility class II molecule on the surface of antigen-presenting cells and to the T cell receptor (TcR) on T cells, SAgs trigger a strong oligoclonal expansion of T cells and the release of massive amounts of proinflammatory cytokines. Thus far, bacterial SAgs have been found in Streptococcus pyogenes (Group A Streptococcus), Group C Streptococcus, Group G Streptococcus, several species of Staphylococcus, Yersinia pseudotuberculosis, and Mycoplasma arthritidis. The streptococcal and staphylococcal SAgs form a large family of structurally related toxins that appear to have a common ancestor. This chapter gives a detailed overview of the structure, biological function, and clinical associations of bacterial SAgs. In addition, we discuss the staphylococcal superantigen-like (SSL) proteins, a novel toxin family of Staphylococcus aureus that are structurally related to SAgs, but target different parts of the innate immune response.


PLOS Pathogens | 2017

Staphylococcal enterotoxin-like X (SElX) is a unique superantigen with functional features of two major families of staphylococcal virulence factors

Ries Langley; Yi Tian Ting; Fiona Clow; Paul G. Young; Fiona J. Radcliff; Jeong Min Choi; Richard P. Sequeira; Silva Holtfreter; Heather M. Baker; John D. Fraser

Staphylococcus aureus is an opportunistic pathogen that produces many virulence factors. Two major families of which are the staphylococcal superantigens (SAgs) and the Staphylococcal Superantigen-Like (SSL) exoproteins. The former are immunomodulatory toxins that induce a Vβ-specific activation of T cells, while the latter are immune evasion molecules that interfere with a wide range of innate immune defences. The superantigenic properties of Staphylococcal enterotoxin-like X (SElX) have recently been established. We now reveal that SElX also possesses functional characteristics of the SSLs. A region of SElX displays high homology to the sialyl-lactosamine (sLacNac)-specific binding site present in a sub-family of SSLs. By analysing the interaction of SElX with sLacNac-containing glycans we show that SElX has an equivalent specificity and host cell binding range to the SSLs. Mutation of key amino acids in this conserved region affects the ability of SElX to bind to cells of myeloid origin and significantly reduces its ability to protect S. aureus from destruction in a whole blood killing (WBK) assay. Like the SSLs, SElX is up-regulated early during infection and is under the control of the S. aureus exotoxin expression (Sae) two component gene regulatory system. Additionally, the structure of SElX in complex with the sLacNac-containing tetrasaccharide sialyl Lewis X (sLeX) reveals that SElX is a unique single-domain SAg. In summary, SElX is an ‘SSL-like’ SAg.

Collaboration


Dive into the Ries Langley's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jian Ni

Human Genome Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge