Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Virulence ; 14(1): 2180228, 2023 12.
Article in English | MEDLINE | ID: mdl-36809931

ABSTRACT

Group A Streptococcus (GAS) is a globally important pathogen causing a broad range of human diseases. GAS pili are elongated proteins with a backbone comprised repeating T-antigen subunits, which extend from the cell surface and have important roles in adhesion and establishing infection. No GAS vaccines are currently available, but T-antigen-based candidates are in pre-clinical development. This study investigated antibody-T-antigen interactions to gain molecular insight into functional antibody responses to GAS pili. Large, chimeric mouse/human Fab-phage libraries generated from mice vaccinated with the complete T18.1 pilus were screened against recombinant T18.1, a representative two-domain T-antigen. Of the two Fab identified for further characterization, one (designated E3) was cross-reactive and also recognized T3.2 and T13, while the other (H3) was type-specific reacting with only T18.1/T18.2 within a T-antigen panel representative of the major GAS T-types. The epitopes for the two Fab, determined by x-ray crystallography and peptide tiling, overlapped and mapped to the N-terminal region of the T18.1 N-domain. This region is predicted to be buried in the polymerized pilus by the C-domain of the next T-antigen subunit. However, flow cytometry and opsonophagocytic assays showed that these epitopes were accessible in the polymerized pilus at 37°C, though not at lower temperature. This suggests that there is motion within the pilus at physiological temperature, with structural analysis of a covalently linked T18.1 dimer indicating "knee-joint" like bending occurs between T-antigen subunits to expose this immunodominant region. This temperature dependent, mechanistic flexing provides new insight into how antibodies interact with T-antigens during infection.


Subject(s)
Antigens, Viral, Tumor , Immunodominant Epitopes , Animals , Humans , Mice , Immunodominant Epitopes/metabolism , Antigens, Viral, Tumor/metabolism , Temperature , Fimbriae, Bacterial/metabolism , Fimbriae Proteins/metabolism , Bacterial Proteins/metabolism , Epitopes , Streptococcus
2.
Clin Transl Immunology ; 10(3): e1261, 2021.
Article in English | MEDLINE | ID: mdl-33747511

ABSTRACT

OBJECTIVES: Circulating antibodies are important markers of previous infection and immunity. Questions remain with respect to the durability and functionality of SARS-CoV-2 antibodies. This study explored antibody responses in recovered COVID-19 patients in a setting where the probability of re-exposure is effectively nil, owing to New Zealand's successful elimination strategy. METHODS: A triplex bead-based assay that detects antibody isotype (IgG, IgM and IgA) and subclass (IgG1, IgG2, IgG3 and IgG4) responses against Nucleocapsid (N) protein, the receptor binding domain (RBD) and Spike (S) protein of SARS-CoV-2 was developed. After establishing baseline levels with pre-pandemic control sera (n = 113), samples from PCR-confirmed COVID-19 patients with mild-moderate disease (n = 189) collected up to 8 months post-infection were examined. The relationship between antigen-specific antibodies and neutralising antibodies (NAbs) was explored with a surrogate neutralisation assay that quantifies inhibition of the RBD/hACE-2 interaction. RESULTS: While most individuals had broad isotype and subclass responses to each antigen shortly after infection, only RBD and S protein IgG, as well as NAbs, were relatively stable over the study period, with 99%, 96% and 90% of samples, respectively, having responses over baseline 4-8 months post-infection. Anti-RBD antibodies were strongly correlated with NAbs at all time points (Pearson's r ≥ 0.87), and feasibility of using finger prick sampling to accurately measure anti-RBD IgG was demonstrated. CONCLUSION: Antibodies to SARS-CoV-2 persist for up to 8 months following mild-to-moderate infection. This robust response can be attributed to the initial exposure without immune boosting given the lack of community transmission in our setting.

3.
Clin Infect Dis ; 73(12): 2322-2325, 2021 12 16.
Article in English | MEDLINE | ID: mdl-33639619

ABSTRACT

Rheumatic fever is a serious post-infectious sequela of group A Streptococcus (GAS). Prior GAS exposures were mapped in sera using a large panel of M-type specific peptides. Rheumatic fever patients had serological evidence of significantly more GAS exposures than matched controls suggesting immune priming by repeat infections contributes to pathogenesis.


Subject(s)
Rheumatic Fever , Streptococcal Infections , Antigens, Bacterial , Humans , Rheumatic Fever/complications , Streptococcal Infections/complications , Streptococcus pyogenes
4.
J Leukoc Biol ; 107(1): 145-158, 2020 01.
Article in English | MEDLINE | ID: mdl-31667914

ABSTRACT

T cells play a key role in mounting an adaptive immune response. T cells are activated upon recognition of cognate Ag presented by an APC. Subsequently, T cells adhere to other activated T cells to form activation clusters, which lead to directed secretion of cytokines between communicating cells. T cell activation clusters have been implicated in regulating activation, proliferation, and memory formation in T cells. We previously reported the expression of the protease inhibitor neuroserpin by human T cells and showed that expression and intracellular localization is regulated following T cell activation. To gain a better understanding of neuroserpin in the proteolytic environment postactivation we assessed its role in human T cell clustering and proliferation. Neuroserpin knockdown increased T cell proliferation and cluster formation following T cell activation. This increased cluster formation was dependent on the proteases tissue plasminogen activator (tPA) and plasmin. Furthermore, neuroserpin knockdown or plasmin treatment of T cells increased the cleavage of annexin A2, a known plasmin target that regulates the actin cytoskeleton. Live cell imaging of activated T cells further indicated a role of the actin cytoskeleton in T cell clustering. The inhibition of actin regulators myosin ATPase and Rho-associated protein kinase signaling completely reversed the neuroserpin knockdown-induced effects. The results presented in this study reveal a novel role for neuroserpin and the proteolytic environment in the regulation of T cell activation biology.


Subject(s)
Cell Communication , Cell Proliferation , Lymphocyte Activation , Neuropeptides/pharmacology , Serine Proteinase Inhibitors/pharmacology , Serpins/pharmacology , T-Lymphocytes/cytology , Tissue Plasminogen Activator/antagonists & inhibitors , Actin Cytoskeleton/metabolism , Humans , Neuropeptides/antagonists & inhibitors , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Neuroserpin
5.
Immunol Cell Biol ; 98(1): 12-21, 2020 01.
Article in English | MEDLINE | ID: mdl-31742781

ABSTRACT

Acute rheumatic fever (ARF) and chronic rheumatic heart disease (RHD) are autoimmune sequelae of a Group A streptococcal infection with significant global mortality and poorly understood pathogenesis. Immunoglobulin and complement deposition were observed in ARF/RHD valve tissue over 50 years ago, yet contemporary investigations have been lacking. This study applied systems immunology to investigate the relationships between the complement system and immunoglobulin in ARF. Patients were stratified by C-reactive protein (CRP) concentration into high (≥10 µg mL-1 ) and low (<10 µg mL-1 ) groups to distinguish those with clinically significant inflammatory processes from those with abating inflammation. The circulating concentrations of 17 complement factors and six immunoglobulin isotypes and subclasses were measured in ARF patients and highly matched healthy controls using multiplex bead-based immunoassays. An integrative statistical approach combining feature selection and principal component analysis revealed a linked IgG3-C4 response in ARF patients with high CRP that was absent in controls. Strikingly, both IgG3 and C4 were elevated above clinical reference ranges, suggesting these features are a marker of ARF-associated inflammation. Humoral immunity in response to M protein, an antigen implicated in ARF pathogenesis, was completely polarized to IgG3 in the patient group. Furthermore, the anti-M-protein IgG3 response was correlated with circulating IgG3 concentration, highlighting a potential role for this potent immunoglobulin subclass in disease. In conclusion, a linked IgG3-C4 response appears important in the initial, inflammatory stage of ARF and may have immediate utility as a clinical biomarker given the lack of specific diagnostic tests currently available.


Subject(s)
Complement C4 , Immunity, Humoral , Immunoglobulin G , Rheumatic Fever , Adolescent , Child , Complement C4/immunology , Complement C4/metabolism , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Male , Rheumatic Fever/blood , Rheumatic Fever/immunology
6.
Sci Rep ; 9(1): 6975, 2019 May 01.
Article in English | MEDLINE | ID: mdl-31043651

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

7.
Sci Rep ; 7(1): 7174, 2017 08 03.
Article in English | MEDLINE | ID: mdl-28775292

ABSTRACT

The human pathogen Group A Streptococcus (GAS) produces pili that are involved in adhesion and colonisation of the host. These surface-exposed pili are immunogenic and therefore represent an attractive target for vaccine development. The pilus is encoded in the genomic region known as the fibronectin-collagen-T-antigen (FCT)-region, of which at least nine different types have been identified. In this study we investigate expressing two of the most common FCT-types (FCT-3 and FCT-4) in the food-grade bacteria Lactococcus lactis for use as a mucosal vaccine. We show that mucosally delivered L. lactis expressing GAS pili generates specific antibody responses in rabbits. Rabbit anti-pilus antibodies were shown to have both a neutralising effect on bacterial adhesion, and immunised rabbit antiserum was able to facilitate immune-mediated killing of bacteria via opsonophagocytosis. Furthermore, intranasal immunisation of mice improved clearance rates of GAS after nasopharyngeal challenge. These results demonstrate the potential for a novel, pilus-based vaccine to protect against GAS infections.


Subject(s)
Fimbriae Proteins/immunology , Lactococcus lactis/immunology , Streptococcal Vaccines/immunology , Streptococcus pyogenes/immunology , Animals , Antigens, Bacterial , Fibronectins , Fimbriae Proteins/genetics , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/immunology , Lactococcus lactis/genetics , Mice , Rabbits , Streptococcal Infections , Streptococcal Vaccines/pharmacology , Streptococcus pyogenes/genetics , Vaccination , Vaccines, Synthetic/immunology
8.
J Microbiol Methods ; 134: 58-61, 2017 03.
Article in English | MEDLINE | ID: mdl-28115206

ABSTRACT

The lack of standardised protocols for the assessment of functional antibodies has hindered Streptococcus pyogenes research and the development of vaccines. A robust, high throughput opsonophagocytic bactericidal assay to determine protective antibodies in human and rabbit serum has been developed that utilises bioluminescence as a rapid read out.


Subject(s)
Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , High-Throughput Screening Assays , Immunologic Tests/methods , Phagocytosis , Streptococcus pyogenes/immunology , Animals , Enzyme-Linked Immunosorbent Assay , Humans , Luminescent Measurements , Rabbits , Streptococcal Infections/diagnosis , Streptococcal Infections/microbiology
9.
Mol Microbiol ; 103(2): 282-298, 2017 01.
Article in English | MEDLINE | ID: mdl-27741558

ABSTRACT

Group A Streptococcus (GAS), or Streptococcus pyogenes, is a human pathogen that causes diseases ranging from skin and soft tissue infections to severe invasive diseases, such as toxic shock syndrome. Each GAS strain carries a particular pilus type encoded in the variable fibronectin-binding, collagen-binding, T antigen (FCT) genomic region. Here, we describe the functional analysis of the serotype M2 pilus encoded in the FCT-6 region. We found that, in contrast to other investigated GAS pili, the ancillary pilin 1 lacks adhesive properties. Instead, the backbone pilin is important for host cell adhesion and binds several host factors, including fibronectin and fibrinogen. Using a panel of recombinant pilus proteins, GAS gene deletion mutants and Lactococcus lactis gain-of-function mutants we show that, unlike other GAS pili, the FCT-6 pilus also contributes to immune evasion. This was demonstrated by a delay in blood clotting, increased intracellular survival of the bacteria in macrophages, higher bacterial survival rates in human whole blood and greater virulence in a Galleria mellonella infection model in the presence of fully assembled FCT-6 pili.


Subject(s)
Bacterial Adhesion/physiology , Fimbriae Proteins/physiology , Streptococcus pyogenes/physiology , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Antigens, Viral, Tumor , Bacterial Adhesion/genetics , Bacterial Adhesion/immunology , Bacterial Proteins/metabolism , Biofilms , Fibronectins/metabolism , Fimbriae Proteins/genetics , Fimbriae Proteins/immunology , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/metabolism , Humans , Immune Evasion , Mutation , Sequence Deletion , Serogroup , Streptococcal Infections/microbiology , Streptococcus pyogenes/genetics , Streptococcus pyogenes/immunology , Streptococcus pyogenes/metabolism , Virulence
10.
Immunol Cell Biol ; 94(10): 955-963, 2016 11.
Article in English | MEDLINE | ID: mdl-27301418

ABSTRACT

The homeostatic chemokine CCL21 has a pivotal role in lymphocyte homing and compartment localisation within the lymph node, and also affects adhesion between immune cells. The effects of CCL21 are modulated by its mode of presentation, with different cellular responses seen for surface-bound and soluble forms. Here we show that plasmin cleaves surface-bound CCL21 to release the C-terminal peptide responsible for CCL21 binding to glycosaminoglycans on the extracellular matrix and cell surfaces, thereby generating the soluble form. Loss of this anchoring peptide enabled the chemotactic activity of CCL21 and reduced cell tethering. Tissue plasminogen activator did not cleave CCL21 directly but enhanced CCL21 processing through generation of plasmin from plasminogen. The tissue plasminogen activator inhibitor neuroserpin prevented processing of CCL21 and blocked the effects of soluble CCL21 on cell migration. Similarly, the plasmin-specific inhibitor α2-antiplasmin inhibited CCL21-mediated migration of human T cells and dendritic cells and tethering of T cells to APCs. We conclude that the plasmin system proteins plasmin, tissue plasminogen activator and neuroserpin regulate CCL21 function in the immune system by controlling the balance of matrix- and cell-bound CCL21.


Subject(s)
Cell Movement/drug effects , Chemokine CCL21/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Plasminogen/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Amino Acid Sequence , Cell Adhesion/drug effects , Cell Communication/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Chemokine CCL21/chemistry , Dendritic Cells/drug effects , Humans , Neuropeptides/pharmacology , Protein Binding/drug effects , Recombinant Proteins/metabolism , Serpins/pharmacology , T-Lymphocytes/drug effects , Tissue Plasminogen Activator/pharmacology , alpha-2-Antiplasmin/pharmacology , Neuroserpin
11.
J Biol Chem ; 290(52): 31126-37, 2015 Dec 25.
Article in English | MEDLINE | ID: mdl-26527680

ABSTRACT

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 Mg(2+), Ca(2+), or Mn(2+). However, Zn(2+) 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.


Subject(s)
Blood Bactericidal Activity/immunology , Immune Evasion , N-Glycosyl Hydrolases/immunology , Streptococcus pyogenes/immunology , Streptococcus pyogenes/pathogenicity , Virulence Factors/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Humans , Lactococcus lactis/genetics , Lactococcus lactis/immunology , Macrophages , Microbial Viability/genetics , Microbial Viability/immunology , N-Glycosyl Hydrolases/genetics , Streptococcus pyogenes/genetics , Virulence Factors/genetics
12.
J Leukoc Biol ; 97(4): 699-710, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25670787

ABSTRACT

Contact between T cells and APCs and activation of an effective immune response trigger cellular polarization and the formation of a structured interface known as the immunological synapse. Interactions across the synapse and secretion of T cell and APC-derived factors into the perisynaptic compartment regulate synapse formation and activation of T cells. We report that the serine protease inhibitor neuroserpin, an axonally secreted protein thought to play roles in the formation of the neuronal synapse and refinement of synaptic activity, is expressed in human naïve effector memory and central memory subsets of CD4(+) and CD8(+) T cells, as well as monocytes, B cells, and NK cells. Neuroserpin partially colocalized with a TGN38/LFA-1-positive vesicle population in T cells and translocates to the immunological synapse upon activation with TCR antibodies or antigen-pulsed APCs. Activation of T cells triggered neuroserpin secretion, a rapid, 8.4-fold up-regulation of the serine protease tissue plasminogen activator, the protease target for neuroserpin, and a delayed, 6.25-fold down-regulation of neuroserpin expression. Evidence of polarization and regulated neuroserpin expression was also seen in ex vivo analyses of human lymph nodes and blood-derived T cells. Increased neuroserpin expression was seen in clusters of T cells in the paracortex of human lymph nodes, with some showing polarization to areas of cell:cell interaction. Our results support a role for neuroserpin and tissue plasminogen activator in activation-controlled proteolytic cleavage of proteins in the synaptic or perisynaptic space to modulate immune cell function.


Subject(s)
Immunological Synapses/physiology , Lymphocyte Activation/physiology , Neuropeptides/metabolism , Serpins/metabolism , T-Lymphocytes/immunology , Tissue Plasminogen Activator/metabolism , Adaptive Immunity/physiology , Antigen Presentation , Cell Communication , Cell Polarity , Humans , Immunologic Memory , Lymph Nodes/cytology , Lymphocyte Function-Associated Antigen-1/metabolism , Lymphocyte Subsets/metabolism , Membrane Glycoproteins/metabolism , Microscopy, Fluorescence , Monocytes/metabolism , Neuropeptides/genetics , Proteolysis , Receptors, Antigen, T-Cell/immunology , Secretory Vesicles/chemistry , Serpins/genetics , Subcellular Fractions/chemistry , T-Lymphocytes/metabolism , Tissue Plasminogen Activator/genetics , Up-Regulation , Neuroserpin
13.
J Transl Med ; 12: 292, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25331734

ABSTRACT

BACKGROUND: microRNAs (miRNAs) are emerging as key regulators of the immune system, but their role in CD8+ T cell differentiation is not well explored. Some evidence suggests that signals from cell surface receptors influence the expression of miRNAs in CD8+ T cells, and may have consequent effects on cell phenotype and function. We set out to investigate whether common gamma chain cytokines modulated human CD8+ T cell expression of miR-146a, which previous studies have associated with different stages of CD8+ differentiation. We also investigated how changes in miR-146a related to other miRNAs that alter with CD8+ differentiation status. METHODS: We treated human CD8+ T cells with the cytokines IL-2, IL-7 or IL-15 either at rest or after stimulation with anti-CD3 and anti-CD28. For some experiments we also purified human CD8+ T cell subsets ex vivo. Flow cytometry was used in parallel to assess cell surface memory marker expression. Total RNA from these cells was subjected to microarray analysis and real-time PCR for miRNA expression. Nucleofection studies were performed to assess potential mRNA targets of miR-146a. RESULTS: We find that miR-146a is up-regulated in naïve CD8+ T cells exposed to IL-2 or IL-15, even in the absence of an activating T cell receptor stimulus, but not when IL-7 is also present. miR-146a expression correlates with a memory phenotype in both ex vivo and in vitro cultured cells although in our hands overexpression of miR-146a was not sufficient alone to drive a full memory phenotype. In ex vivo analysis, miR-146a was one of a small number of miRNAs that was differentially expressed between naïve and memory CD8+ T cells. CONCLUSIONS: miR-146a is emerging as a critical regulator of immune system. Our data shows that miR-146a expression is strongly influenced by the cytokine milieu even in the absence of a T cell receptor stimulus. Our results have implications for studies designed to assess the function of miR-146a, help to define a fingerprint of miRNA expression in CD8+ T cell subsets and may be useful when designing optimal protocols for T cell expansion as efficacy of T cell immunotherapy is correlated with an 'early' memory phenotype.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Cytokines/pharmacology , Gene Expression Regulation/drug effects , MicroRNAs/genetics , T-Lymphocyte Subsets/metabolism , Antigens/metabolism , CD8-Positive T-Lymphocytes/drug effects , Fas-Associated Death Domain Protein/metabolism , Gene Expression Profiling , Humans , Immunologic Memory , Interleukin-15/pharmacology , Interleukin-2/pharmacology , Interleukin-7/pharmacology , MicroRNAs/metabolism , Phenotype , Receptors, Antigen, T-Cell/metabolism , Receptors, CCR7/metabolism , T-Lymphocyte Subsets/drug effects , TNF Receptor-Associated Factor 6/metabolism , Up-Regulation/drug effects
14.
Immunol Cell Biol ; 91(7): 469-76, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23797068

ABSTRACT

Staphylococcus aureus is an opportunistic bacterial pathogen responsible for a range of diseases, from local skin infections through to life-threatening illnesses such as toxic shock syndrome. S. aureus produces an assortment of molecules designed to evade or subvert the host immune system. One example is the 23 kDa staphylococcal superantigen-like protein 7 (SSL7) that simultaneously binds immunoglobulin A (IgA) and complement C5 to inhibit complement-mediated hemolytic and bactericidal activity. The avirulent bacterium Lactococcus lactis was engineered to express SSL7 so that its role in bacterial survival could be assessed without interference from other virulence factors. Expression of SSL7 by L. lactis led to significantly enhanced bacterial survival in whole human blood and prevented the membrane attack complex (C5b-9) forming on the cell wall. To further understand the mechanism of action of SSL7, the activity of wild-type SSL7 protein was compared with a panel of mutant proteins lacking the capacity to bind IgA, C5, or both IgA and C5. SSL7 potently inhibited in vitro chemotaxis of inflammatory myeloid cells in response to a pathogenic stimulus and when injected into mice, SSL7 blocked the migration of neutrophils into the peritoneum in response to an inoculum of heat-killed S. aureus. Mutagenesis of the C5-binding site on SSL7 abolished all inhibitory activity, while mutation of the IgA-binding site had only partial effects, indicating that while IgA binding enhances activity it is not essential. SSL7 is an important staphylococcal virulence factor with potent anti-inflammatory properties, which are mediated by targeting complement C5 and IgA.


Subject(s)
Complement C5/metabolism , Exotoxins/metabolism , Immunoglobulin A/metabolism , Lactococcus lactis/genetics , Neutrophils/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Virulence Factors/metabolism , Animals , Blood Bactericidal Activity/genetics , Cell Movement/genetics , Cells, Cultured , Exotoxins/genetics , Genetic Engineering , Humans , Immune Evasion , Mice , Mice, Inbred BALB C , Mutation/genetics , Protein Binding/genetics , Staphylococcus aureus/pathogenicity , Transgenes/genetics , Virulence Factors/genetics
15.
Proc Natl Acad Sci U S A ; 107(8): 3681-6, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20133685

ABSTRACT

Staphylococcus aureus secretes the SSL7 protein as part of its immune evasion strategy. The protein binds both complement C5 and IgA, yet it is unclear whether SSL7 cross-links these two proteins and, if so, what purpose this serves the pathogen. We have isolated a stable IgA-SSL7-C5 complex, and our crystal structure of the C5-SSL7 complex confirms that binding to C5 occurs exclusively through the C-terminal beta-grasp domain of SSL7 leaving the OB domain free to interact with IgA. SSL7 interacts with C5 >70 A from the C5a cleavage site without inducing significant conformational changes in C5, and efficient inhibition of convertase cleavage of C5 is shown to be IgA dependent. Inhibition of C5a production and bacteriolysis are all shown to require C5 and IgA binding while inhibition of hemolysis is achieved by the C5 binding SSL7 beta-grasp domain alone. These results provide a conceptual and structural basis for the development of a highly specific complement inhibitor preventing only the formation of the lytic membrane attack complex without affecting the important signaling functions of C5a.


Subject(s)
Complement C5/antagonists & inhibitors , Complement C5/chemistry , Exotoxins/immunology , Staphylococcus aureus/immunology , Animals , Complement C5/immunology , Crystallography, X-Ray , Humans , Mutation , Protein Structure, Tertiary , Rabbits , Staphylococcus aureus/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL
...