Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters

Publication year range
1.
J Biol Chem ; 299(3): 102936, 2023 03.
Article in English | MEDLINE | ID: mdl-36702253

ABSTRACT

Staphylococcus aureus and Staphylococcus epidermidis are frequently associated with medical device infections that involve establishment of a bacterial biofilm on the device surface. Staphylococcal surface proteins Aap, SasG, and Pls are members of the Periscope Protein class and have been implicated in biofilm formation and host colonization; they comprise a repetitive region ("B region") and an N-terminal host colonization domain within the "A region," predicted to be a lectin domain. Repetitive E-G5 domains (as found in Aap, SasG, and Pls) form elongated "stalks" that would vary in length with repeat number, resulting in projection of the N-terminal A domain variable distances from the bacterial cell surface. Here, we present the structures of the lectin domains within A regions of SasG, Aap, and Pls and a structure of the Aap lectin domain attached to contiguous E-G5 repeats, suggesting the lectin domains will sit at the tip of the variable length rod. We demonstrate that these isolated domains (Aap, SasG) are sufficient to bind to human host desquamated nasal epithelial cells. Previously, proteolytic cleavage or a deletion within the A domain had been reported to induce biofilm formation; the structures suggest a potential link between these observations. Intriguingly, while the Aap, SasG, and Pls lectin domains bind a metal ion, they lack the nonproline cis peptide bond thought to be key for carbohydrate binding by the lectin fold. This suggestion of noncanonical ligand binding should be a key consideration when investigating the host cell interactions of these bacterial surface proteins.


Subject(s)
Bacterial Proteins , Models, Molecular , Protein Domains , Staphylococcus aureus , Humans , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Lectins/chemistry , Lectins/metabolism , Staphylococcal Infections/microbiology , Staphylococcus epidermidis/chemistry , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/metabolism , Protein Domains/physiology , Protein Structure, Tertiary , Protein Binding , Staphylococcus aureus/chemistry , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Escherichia coli , Epithelial Cells/microbiology
2.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33361150

ABSTRACT

Staphylococcus aureus colonizes the skin of the majority of patients with atopic dermatitis (AD), and its presence increases disease severity. Adhesion of S. aureus to corneocytes in the stratum corneum is a key initial event in colonization, but the bacterial and host factors contributing to this process have not been defined. Here, we show that S. aureus interacts with the host protein corneodesmosin. Corneodesmosin is aberrantly displayed on the tips of villus-like projections that occur on the surface of AD corneocytes as a result of low levels of skin humectants known as natural moisturizing factor (NMF). An S. aureus mutant deficient in fibronectin binding protein B (FnBPB) and clumping factor B (ClfB) did not bind to corneodesmosin in vitro. Using surface plasmon resonance, we found that FnBPB and ClfB proteins bound with similar affinities. The S. aureus binding site was localized to the N-terminal glycine-serine-rich region of corneodesmosin. Atomic force microscopy showed that the N-terminal region was present on corneocytes containing low levels of NMF and that blocking it with an antibody inhibited binding of individual S. aureus cells to corneocytes. Finally, we found that S. aureus mutants deficient in FnBPB or ClfB have a reduced ability to adhere to low-NMF corneocytes from patients. In summary, we show that FnBPB and ClfB interact with the accessible N-terminal region of corneodesmosin on AD corneocytes, allowing S. aureus to take advantage of the aberrant display of corneodesmosin that accompanies low NMF in AD. This interaction facilitates the characteristic strong binding of S. aureus to AD corneocytes.


Subject(s)
Dermatitis, Atopic/microbiology , Intercellular Signaling Peptides and Proteins/metabolism , Staphylococcus aureus/metabolism , Adhesins, Bacterial/metabolism , Bacterial Adhesion/physiology , Coagulase/metabolism , Dermatitis, Atopic/metabolism , Epidermis , Epithelial Cells/metabolism , Humans , Microscopy, Atomic Force , Skin/metabolism , Skin/microbiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/pathogenicity
3.
Microbiology (Reading) ; 169(6)2023 06.
Article in English | MEDLINE | ID: mdl-37326621

ABSTRACT

Adhesive interactions between Staphylococcus aureus and the host rely on cell-wall-anchored proteins such as fibronectin-binding protein B (FnBPB). Recently we showed that the FnBPB protein expressed by clonal complex (CC) 1 isolates of S. aureus mediates bacterial adhesion to corneodesmosin. The proposed ligand-binding region of CC1-type FnBPB shares just 60 % amino acid identity with the archetypal FnBPB protein from CC8. Here we investigated ligand binding and biofilm formation by CC1-type FnBPB. We found that the A domain of FnBPB binds to fibrinogen and corneodesmosin and identified residues within the hydrophobic ligand trench in the A domain that are essential for the binding of CC1-type FnBPB to ligands and during biofilm formation. We further investigated the interplay between different ligands and the influence of ligand binding on biofilm formation. Overall, our study provides new insights into the requirements for CC1-type FnBPB-mediated adhesion to host proteins and FnBPB-mediated biofilm formation in S. aureus.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Humans , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Ligands , Adhesins, Bacterial/genetics , Adhesins, Bacterial/chemistry , Adhesins, Bacterial/metabolism , Bacterial Adhesion , Carrier Proteins/metabolism , Protein Binding , Staphylococcal Infections/microbiology , Fibronectins/metabolism , Biofilms , Bacterial Proteins/metabolism
4.
Microbiology (Reading) ; 169(9)2023 09.
Article in English | MEDLINE | ID: mdl-37668351

ABSTRACT

Complement offers a first line of defence against infection through the opsonization of microbial pathogens, recruitment of professional phagocytes to the infection site and the coordination of inflammatory responses required for the resolution of infection. Staphylococcus aureus is a successful pathogen that has developed multiple mechanisms to thwart host immune responses. Understanding the precise strategies employed by S. aureus to bypass host immunity will be paramount for the development of vaccines and or immunotherapies designed to prevent or limit infection. To gain a better insight into the specific immune evasion mechanisms used by S. aureus we examined the pathogen's interaction with the soluble complement inhibitor, C4b-binding protein (C4BP). Previous studies indicated that S. aureus recruits C4BP using a specific cell-wall-anchored surface protein and that bound C4BP limits complement deposition on the staphylococcal surface. Using flow-cytometric-based bacterial-protein binding assays we observed no interaction between S. aureus and C4BP. Moreover, we offer a precautionary warning that C4BP isolated from plasma can be co-purified with minute quantities of human IgG, which can distort binding analysis between S. aureus and human-derived proteins. Combined our data indicates that recruitment of C4BP is not a complement evasion strategy employed by S. aureus.


Subject(s)
Complement C4b-Binding Protein , Staphylococcal Infections , Humans , Staphylococcus aureus/genetics , Complement System Proteins , Staphylococcus , Membrane Proteins
5.
PLoS Pathog ; 15(4): e1007713, 2019 04.
Article in English | MEDLINE | ID: mdl-31009507

ABSTRACT

Staphylococcus aureus expresses a number of cell wall-anchored proteins that mediate adhesion and invasion of host cells and tissues and promote immune evasion, consequently contributing to the virulence of this organism. The cell wall-anchored protein clumping factor B (ClfB) has previously been shown to facilitate S. aureus nasal colonization through high affinity interactions with the cornified envelope in the anterior nares. However, the role of ClfB during skin and soft tissue infection (SSTI) has never been investigated. This study reveals a novel role for ClfB during SSTIs. ClfB is crucial in determining the abscess structure and bacterial burden early in infection and this is dependent upon a specific interaction with the ligand loricrin which is expressed within the abscess tissue. Targeting ClfB using a model vaccine that induced both protective humoral and cellular responses, leads to protection during S. aureus skin infection. This study therefore identifies ClfB as an important antigen for future SSTI vaccines.


Subject(s)
Adhesins, Bacterial/metabolism , Staphylococcal Skin Infections/microbiology , Staphylococcus aureus/immunology , Vaccines/immunology , Virulence Factors/metabolism , Virulence , Adhesins, Bacterial/genetics , Adhesins, Bacterial/immunology , Animals , Bacterial Adhesion , Female , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/metabolism , Vaccines/administration & dosage , Virulence Factors/genetics , Virulence Factors/immunology
6.
PLoS Pathog ; 15(6): e1007816, 2019 06.
Article in English | MEDLINE | ID: mdl-31216354

ABSTRACT

Fibrinogen is an essential part of the blood coagulation cascade and a major component of the extracellular matrix in mammals. The interface between fibrinogen and bacterial pathogens is an important determinant of the outcome of infection. Here, we demonstrate that a canine host-restricted skin pathogen, Staphylococcus pseudintermedius, produces a cell wall-associated protein (SpsL) that has evolved the capacity for high strength binding to canine fibrinogen, with reduced binding to fibrinogen of other mammalian species including humans. Binding occurs via the surface-expressed N2N3 subdomains, of the SpsL A-domain, to multiple sites in the fibrinogen α-chain C-domain by a mechanism analogous to the classical dock, lock, and latch binding model. Host-specific binding is dependent on a tandem repeat region of the fibrinogen α-chain, a region highly divergent between mammals. Of note, we discovered that the tandem repeat region is also polymorphic in different canine breeds suggesting a potential influence on canine host susceptibility to S. pseudintermedius infection. Importantly, the strong host-specific fibrinogen-binding interaction of SpsL to canine fibrinogen is essential for bacterial aggregation and biofilm formation, and promotes resistance to neutrophil phagocytosis, suggesting a key role for the interaction during pathogenesis. Taken together, we have dissected a bacterial surface protein-ligand interaction resulting from the co-evolution of host and pathogen that promotes host-specific innate immune evasion and may contribute to its host-restricted ecology.


Subject(s)
Bacterial Proteins/immunology , Biofilms/growth & development , Fibrinogen/immunology , Immune Evasion , Immunity, Innate , Staphylococcus/physiology , Animals , Bacterial Proteins/genetics , Chickens , Dogs , Fibrinogen/genetics , Humans
7.
Proc Natl Acad Sci U S A ; 115(21): 5564-5569, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29735708

ABSTRACT

Clumping factor A (ClfA), a cell-wall-anchored protein from Staphylococcus aureus, is a virulence factor in various infections and facilitates the colonization of protein-coated biomaterials. ClfA promotes bacterial adhesion to the blood plasma protein fibrinogen (Fg) via molecular forces that have not been studied so far. A unique, yet poorly understood, feature of ClfA is its ability to favor adhesion to Fg at high shear stress. Unraveling the strength and dynamics of the ClfA-Fg interaction would help us better understand how S. aureus colonizes implanted devices and withstands physiological shear stress. By means of single-molecule experiments, we show that ClfA behaves as a force-sensitive molecular switch that potentiates staphylococcal adhesion under mechanical stress. The bond between ClfA and immobilized Fg is weak (∼0.1 nN) at low tensile force, but is dramatically enhanced (∼1.5 nN) by mechanical tension, as observed with catch bonds. Strong bonds, but not weak ones, are inhibited by a peptide mimicking the C-terminal segment of the Fg γ-chain. These results point to a model whereby ClfA interacts with Fg via two distinct binding sites, the adhesive function of which is regulated by mechanical tension. This force-activated mechanism is of biological significance because it explains at the molecular level the ability of ClfA to promote bacterial attachment under high physiological shear stress.


Subject(s)
Bacterial Adhesion/physiology , Coagulase/metabolism , Fibrinogen/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology , Binding Sites , Biomechanical Phenomena , Cells, Cultured , Coagulase/genetics , Fibrinogen/genetics , Molecular Dynamics Simulation , Protein Binding
8.
J Biol Chem ; 294(10): 3588-3602, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30622139

ABSTRACT

Staphylococcus aureus is a Gram-positive bacterium that can cause both superficial and deep-seated infections. Histones released by neutrophils kill bacteria by binding to the bacterial cell surface and causing membrane damage. We postulated that cell wall-anchored proteins protect S. aureus from the bactericidal effects of histones by binding to and sequestering histones away from the cell envelope. Here, we focused on S. aureus strain LAC and by using an array of biochemical assays, including surface plasmon resonance and ELISA, discovered that fibronectin-binding protein B (FnBPB) is the main histone receptor. FnBPB bound all types of histones, but histone H3 displayed the highest affinity and bactericidal activity and was therefore investigated further. H3 bound specifically to the A domain of recombinant FnBPB with a KD of 86 nm, ∼20-fold lower than that for fibrinogen. Binding apparently occurred by the same mechanism by which FnBPB binds to fibrinogen, because FnBPB variants defective in fibrinogen binding also did not bind H3. An FnBPB-deletion mutant of S. aureus LAC bound less H3 and was more susceptible to its bactericidal activity and to neutrophil extracellular traps, whereas an FnBPB-overexpressing mutant bound more H3 and was more resistant than the WT. FnBPB bound simultaneously to H3 and plasminogen, which after activation by tissue plasminogen activator cleaved the bound histone. We conclude that FnBPB provides a dual immune-evasion function that captures histones and prevents them from reaching the bacterial membrane and simultaneously binds plasminogen, thereby promoting its conversion to plasmin to destroy the bound histone.


Subject(s)
Adhesins, Bacterial/metabolism , Anti-Infective Agents/pharmacology , Histones/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Anti-Infective Agents/metabolism , Cell Wall/drug effects , Cell Wall/metabolism , Histones/metabolism , Osmolar Concentration , Plasminogen/metabolism , Protein Binding , Staphylococcus aureus/cytology
9.
Allergy ; 75(12): 3216-3227, 2020 12.
Article in English | MEDLINE | ID: mdl-32644214

ABSTRACT

BACKGROUND: Atopic dermatitis (AD) is associated with a dysregulation of the skin barrier and may predispose to the development of secondary allergic conditions, such as asthma. Tmem79ma/ma mice harbor a mutation in the gene encoding Transmembrane Protein 79 (or Mattrin), which has previously been associated with AD. As a result of the Tmem79 gene mutation, these mice have a defective skin barrier and develop spontaneous skin inflammation. In this study, Tmem79ma/ma mice were assessed for the underlying immunological response in the development of spontaneous skin and lung inflammation. METHODS: Development of spontaneous skin and lung inflammation in Tmem79ma/ma mice was analyzed. We further investigated susceptibility to cutaneous Staphylococcus aureus infection. Tmem79ma/ma were crossed to IL-17A-deficient mice to address the contribution of IL-17A to spontaneous skin and lung disease. RESULTS: Tmem79ma/ma mice developed IL-17A-dependent spontaneous AD-like inflammation and were refractory to S aureus infection. Mutant mice progressed to airway inflammation subsequent to the occurrence of dermatitis. The progression from skin to lung disease is dependent on adaptive immunity and is facilitated by cutaneous expansion of Th17 and TCRγδ T cells. CONCLUSION: Mice lacking Tmem79/Mattrin expression have a defective skin barrier. In adulthood, these mice develop dermatitis with secondary progression to lung inflammation. The development of skin and lung inflammation is IL-17A-dependent and mediated by TCRγδ T cells.


Subject(s)
Dermatitis, Atopic , Interleukin-17 , Pneumonia , Animals , Dermatitis, Atopic/genetics , Disease Models, Animal , Interleukin-17/genetics , Membrane Proteins/genetics , Mice , Pneumonia/genetics , Skin
10.
Proc Natl Acad Sci U S A ; 114(14): 3738-3743, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28320940

ABSTRACT

Staphylococcus aureus forms biofilms on indwelling medical devices using a variety of cell-surface proteins. There is growing evidence that specific homophilic interactions between these proteins represent an important mechanism of cell accumulation during biofilm formation, but the underlying molecular mechanisms are still not well-understood. Here we report the direct measurement of homophilic binding forces by the serine-aspartate repeat protein SdrC and their inhibition by a peptide. Using single-cell and single-molecule force measurements, we find that SdrC is engaged in low-affinity homophilic bonds that promote cell-cell adhesion. Low-affinity intercellular adhesion may play a role in favoring biofilm dynamics. We show that SdrC also mediates strong cellular interactions with hydrophobic surfaces, which are likely to be involved in the initial attachment to biomaterials, the first stage of biofilm formation. Furthermore, we demonstrate that a peptide derived from ß-neurexin is a powerful competitive inhibitor capable of efficiently blocking surface attachment, homophilic adhesion, and biofilm accumulation. Molecular modeling suggests that this blocking activity may originate from binding of the peptide to a sequence of SdrC involved in homophilic interactions. Our study opens up avenues for understanding the role of homophilic interactions in staphylococcal adhesion, and for the design of new molecules to prevent biofilm formation during infection.


Subject(s)
Bacterial Proteins/metabolism , Biofilms , Nerve Tissue Proteins/chemistry , Peptides/pharmacology , Staphylococcus aureus/physiology , Bacterial Adhesion , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Binding Sites , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Peptides/chemistry , Protein Binding , Single-Cell Analysis
11.
Proc Natl Acad Sci U S A ; 113(2): 410-5, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26715750

ABSTRACT

Staphylococcus aureus surface protein SasG promotes cell-cell adhesion during the accumulation phase of biofilm formation, but the molecular basis of this interaction remains poorly understood. Here, we unravel the mechanical properties of SasG on the surface of living bacteria, that is, in its native cellular environment. Nanoscale multiparametric imaging of living bacteria reveals that Zn(2+) strongly increases cell wall rigidity and activates the adhesive function of SasG. Single-cell force measurements show that SasG mediates cell-cell adhesion via specific Zn(2+)-dependent homophilic bonds between ß-sheet-rich G5-E domains on neighboring cells. The force required to unfold individual domains is remarkably strong, up to ∼500 pN, thus explaining how SasG can withstand physiological shear forces. We also observe that SasG forms homophilic bonds with the structurally related accumulation-associated protein of Staphylococcus epidermidis, suggesting the possibility of multispecies biofilms during host colonization and infection. Collectively, our findings support a model in which zinc plays a dual role in activating cell-cell adhesion: adsorption of zinc ions to the bacterial cell surface increases cell wall cohesion and favors the projection of elongated SasG proteins away from the cell surface, thereby enabling zinc-dependent homophilic bonds between opposing cells. This work demonstrates an unexpected relationship between mechanics and adhesion in a staphylococcal surface protein, which may represent a general mechanism among bacterial pathogens for activating cell association.


Subject(s)
Bacterial Proteins/metabolism , Biofilms/drug effects , Membrane Proteins/metabolism , Staphylococcus aureus/physiology , Zinc/pharmacology , Bacterial Adhesion/drug effects , Bacterial Proteins/chemistry , Biomechanical Phenomena/drug effects , Biophysical Phenomena/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Wall/drug effects , Cell Wall/metabolism , Membrane Proteins/chemistry , Microscopy, Atomic Force , Models, Biological , Protein Structure, Tertiary , Staphylococcus aureus/drug effects
12.
Environ Microbiol ; 20(4): 1576-1589, 2018 04.
Article in English | MEDLINE | ID: mdl-29521441

ABSTRACT

Excess copper is highly toxic and forms part of the host innate immune system's antibacterial arsenal, accumulating at sites of infection and acting within macrophages to kill engulfed pathogens. We show for the first time that a novel, horizontally gene transferred copper resistance locus (copXL), uniquely associated with the SCCmec elements of the highly virulent, epidemic, community acquired methicillin resistant Staphylococcus aureus (CA-MRSA) USA300, confers copper hyper-resistance. These genes are additional to existing core genome copper resistance mechanisms, and are not found in typical S. aureus lineages, but are increasingly identified in emerging pathogenic isolates. Our data show that CopX, a putative P1B-3 -ATPase efflux transporter, and CopL, a novel lipoprotein, confer copper hyper-resistance compared to typical S. aureus strains. The copXL genes form an operon that is tightly repressed in low copper environments by the copper regulator CsoR. Significantly, CopX and CopL are important for S. aureus USA300 intracellular survival within macrophages. Therefore, the emergence of new S. aureus clones with the copXL locus has significant implications for public health because these genes confer increased resistance to antibacterial copper toxicity, enhancing bacterial fitness by altering S. aureus interaction with innate immunity.


Subject(s)
Anti-Bacterial Agents/toxicity , Copper/toxicity , Drug Resistance, Bacterial/genetics , Macrophages/microbiology , Membrane Transport Proteins/genetics , Methicillin-Resistant Staphylococcus aureus , Gene Transfer, Horizontal/genetics , Humans , Immunity, Innate/immunology , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/growth & development , Operon , Staphylococcal Infections/microbiology
13.
Curr Top Microbiol Immunol ; 409: 95-120, 2017.
Article in English | MEDLINE | ID: mdl-26667044

ABSTRACT

Staphylococcus aureus persistently colonizes about 20 % of the population and is intermittently associated with the remainder. The organism can cause superficial skin infections and life-threatening invasive diseases. The surface of the bacterial cell displays a variety of proteins that are covalently anchored to peptidoglycan. They perform many functions including adhesion to host cells and tissues, invasion of non-phagocytic cells, and evasion of innate immune responses. The proteins have been categorized into distinct classes based on structural and functional analysis. Many surface proteins are multifunctional. Cell wall-anchored proteins perform essential functions supporting survival and proliferation during the commensal state and during invasive infections. The ability of cell wall-anchored proteins to bind to desquamated epithelial cells is important during colonization, and the binding to fibrinogen is of particular significance in pathogenesis.


Subject(s)
Staphylococcus aureus , Bacterial Adhesion , Bacterial Proteins , Cell Wall , Membrane Proteins
14.
J Biol Chem ; 291(35): 18148-62, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27387503

ABSTRACT

Staphylococcus aureus is a commensal bacterium that has the ability to cause superficial and deep-seated infections. Like several other invasive pathogens, S. aureus can capture plasminogen from the human host where it can be converted to plasmin by host plasminogen activators or by endogenously expressed staphylokinase. This study demonstrates that sortase-anchored cell wall-associated proteins are responsible for capturing the bulk of bound plasminogen. Two cell wall-associated proteins, the fibrinogen- and fibronectin-binding proteins A and B, were found to bind plasminogen, and one of them, FnBPB, was studied in detail. Plasminogen captured on the surface of S. aureus- or Lactococcus lactis-expressing FnBPB could be activated to the potent serine protease plasmin by staphylokinase and tissue plasminogen activator. Plasminogen bound to recombinant FnBPB with a KD of 0.532 µm as determined by surface plasmon resonance. Plasminogen binding did not to occur by the same mechanism through which FnBPB binds to fibrinogen. Indeed, FnBPB could bind both ligands simultaneously indicating that their binding sites do not overlap. The N3 subdomain of FnBPB contains the full plasminogen-binding site, and this includes, at least in part, two conserved patches of surface-located lysine residues that were recognized by kringle 4 of the host protein.


Subject(s)
Adhesins, Bacterial/chemistry , Bacterial Proteins/chemistry , Plasminogen/chemistry , Staphylococcus aureus/chemistry , Adhesins, Bacterial/metabolism , Bacterial Proteins/metabolism , Humans , Plasminogen/metabolism , Protein Binding , Protein Domains , Staphylococcus aureus/metabolism
15.
Infect Immun ; 85(12)2017 12.
Article in English | MEDLINE | ID: mdl-28947645

ABSTRACT

Staphylococcus aureus has become increasingly resistant to antibiotics, and vaccines offer a potential solution to this epidemic of antimicrobial resistance. Targeting of specific T cell subsets is now considered crucial for next-generation anti-S. aureus vaccines; however, there is a paucity of information regarding T cell antigens of S. aureus This study highlights the importance of cell wall-anchored proteins as human CD4+ T cell activators capable of driving antigen-specific Th1 and Th17 cell activation. Clumping factor A (ClfA), which contains N1, N2, and N3 binding domains, was found to be a potent human T cell activator. We further investigated which subdomains of ClfA were involved in T cell activation and found that the full-length ClfA N123 and N23 were potent Th1 and Th17 activators. Interestingly, the N1 subdomain was capable of exclusively activating Th1 cells. Furthermore, when these subdomains were used in a model vaccine, N23 and N1 offered Th1- and Th17-mediated systemic protection in mice upon intraperitoneal challenge. Overall, however, full-length ClfA N123 is required for maximal protection both locally and systemically.


Subject(s)
Antigens, Bacterial/immunology , Coagulase/immunology , Staphylococcus aureus/immunology , T-Lymphocytes/immunology , Animals , Cells, Cultured , Disease Models, Animal , Humans , Mice, Inbred C57BL , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/administration & dosage , Staphylococcal Vaccines/immunology , Survival Analysis
16.
Infect Immun ; 85(6)2017 06.
Article in English | MEDLINE | ID: mdl-28373353

ABSTRACT

Staphylococcus aureus skin infection is a frequent and recurrent problem in children with the common inflammatory skin disease atopic dermatitis (AD). S. aureus colonizes the skin of the majority of children with AD and exacerbates the disease. The first step during colonization and infection is bacterial adhesion to the cornified envelope of corneocytes in the outer layer, the stratum corneum. Corneocytes from AD skin are structurally different from corneocytes from normal healthy skin. The objective of this study was to identify bacterial proteins that promote the adherence of S. aureus to AD corneocytes. S. aureus strains from clonal complexes 1 and 8 were more frequently isolated from infected AD skin than from the nasal cavity of healthy children. AD strains had increased ClfB ligand binding activity compared to normal nasal carriage strains. Adherence of single S. aureus bacteria to corneocytes from AD patients ex vivo was studied using atomic force microscopy. Bacteria expressing ClfB recognized ligands distributed over the entire corneocyte surface. The ability of an isogenic ClfB-deficient mutant to adhere to AD corneocytes compared to that of its parent clonal complex 1 clinical strain was greatly reduced. ClfB from clonal complex 1 strains had a slightly higher binding affinity for its ligand than ClfB from strains from other clonal complexes. Our results provide new insights into the first step in the establishment of S. aureus colonization in AD patients. ClfB is a key adhesion molecule for the interaction of S. aureus with AD corneocytes and represents a target for intervention.


Subject(s)
Adhesins, Bacterial/metabolism , Dermatitis, Atopic/microbiology , Epithelial Cells/microbiology , Staphylococcal Skin Infections/microbiology , Staphylococcus aureus/metabolism , Adhesins, Bacterial/genetics , Bacterial Adhesion , Child, Preschool , Female , Filaggrin Proteins , Humans , Male , Nasal Cavity/microbiology , Sequence Deletion , Skin/cytology , Skin/microbiology , Staphylococcus aureus/genetics
17.
PLoS Pathog ; 11(11): e1005226, 2015.
Article in English | MEDLINE | ID: mdl-26539822

ABSTRACT

Mechanisms of protective immunity to Staphylococcus aureus infection in humans remain elusive. While the importance of cellular immunity has been shown in mice, T cell responses in humans have not been characterised. Using a murine model of recurrent S. aureus peritonitis, we demonstrated that prior exposure to S. aureus enhanced IFNγ responses upon subsequent infection, while adoptive transfer of S. aureus antigen-specific Th1 cells was protective in naïve mice. Translating these findings, we found that S. aureus antigen-specific Th1 cells were also significantly expanded during human S. aureus bloodstream infection (BSI). These Th1 cells were CD45RO+, indicative of a memory phenotype. Thus, exposure to S. aureus induces memory Th1 cells in mice and humans, identifying Th1 cells as potential S. aureus vaccine targets. Consequently, we developed a model vaccine comprising staphylococcal clumping factor A, which we demonstrate to be an effective human T cell antigen, combined with the Th1-driving adjuvant CpG. This novel Th1-inducing vaccine conferred significant protection during S. aureus infection in mice. This study notably advances our understanding of S. aureus cellular immunity, and demonstrates for the first time that a correlate of S. aureus protective immunity identified in mice may be relevant in humans.


Subject(s)
Immunologic Memory , Staphylococcal Infections/immunology , Staphylococcal Skin Infections/microbiology , Staphylococcus aureus/immunology , Th1 Cells/immunology , Adjuvants, Immunologic/pharmacology , Adoptive Transfer , Adult , Aged , Animals , Antigens/immunology , Female , Humans , Interleukin-17/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Staphylococcal Skin Infections/immunology , Th1 Cells/drug effects
18.
Bioconjug Chem ; 28(6): 1684-1692, 2017 06 21.
Article in English | MEDLINE | ID: mdl-28489355

ABSTRACT

Numerous naturally occurring toxins can perturb biological systems when they invade susceptible cells. Coupling of pertinent targeting ligands to the active domains of such proteins provides a strategy for directing these to particular cellular populations implicated in disease. A novel approach described herein involved fusion of one mutated immunoglobulin G (IgG) binding moiety of staphylococcal protein A to the SNARE protease and translocation domain of botulinum neurotoxin A (BoNT/A). This chimera could be monovalently coupled to IgG or via its Fc region to recombinant targeting ligands. The utility of the resulting conjugates is demonstrated by the delivery of a SNARE protease into a cell line expressing tropomyosin receptor kinase A (TrkA) through coupling to anti-TrkA IgG or a fusion of Fc and nerve-growth factor. Thus, this is a versitile and innovative technology for conjugating toxins to diverse ligands for retargeted cell delivery of potential therapeutics.


Subject(s)
Botulinum Toxins, Type A/chemistry , Immunoglobulin G/chemistry , SNARE Proteins/metabolism , Binding Sites , Drug Delivery Systems , Immunoglobulin Fc Fragments , Immunoglobulin G/metabolism , Nerve Growth Factor/immunology , Receptor, trkA/immunology , Vaccines
19.
J Infect Dis ; 213(1): 139-48, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26136471

ABSTRACT

Staphylococcus aureus biofilms, a leading cause of persistent infections, are highly resistant to immune defenses and antimicrobial therapies. In the present study, we investigated the contribution of fibrin and staphylokinase (Sak) to biofilm formation. In both clinical S. aureus isolates and laboratory strains, high Sak-producing strains formed less biofilm than strains that lacked Sak, suggesting that Sak prevents biofilm formation. In addition, Sak induced detachment of mature biofilms. This effect depended on plasminogen activation by Sak. Host-derived fibrin, the main substrate cleaved by Sak-activated plasminogen, was a major component of biofilm matrix, and dissolution of this fibrin scaffold greatly increased susceptibility of biofilms to antibiotics and neutrophil phagocytosis. Sak also attenuated biofilm-associated catheter infections in mouse models. In conclusion, our results reveal a novel role for Sak-induced plasminogen activation that prevents S. aureus biofilm formation and induces detachment of existing biofilms through proteolytic cleavage of biofilm matrix components.


Subject(s)
Biofilms/drug effects , Metalloendopeptidases/metabolism , Staphylococcus aureus/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Female , Fibrin/metabolism , Male , Metalloendopeptidases/pharmacology , Mice , Mice, Inbred C57BL , Plasminogen/metabolism , Staphylococcus aureus/drug effects
20.
Infect Immun ; 83(4): 1598-609, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25644005

ABSTRACT

The immunoglobulin binding protein A (SpA) of Staphylococcus aureus is synthesized as a precursor with a C-terminal sorting signal. The sortase A enzyme mediates covalent attachment to peptidoglycan so that SpA is displayed on the surface of the bacterium. Protein A is also found in the extracellular medium, but the processes involved in its release are not fully understood. Here, we show that a portion of SpA is released into the supernatant with an intact sorting signal, indicating that it has not been processed by sortase A. Release of SpA was reduced when the native sorting signal of SpA was replaced with the corresponding region of another sortase-anchored protein (SdrE). Similarly, a reporter protein fused to the sorting signal of SpA was released to a greater extent than the same polypeptide fused to the SdrE sorting signal. Released SpA protected bacteria from killing in human blood, indicating that it contributes to immune evasion.


Subject(s)
Aminoacyltransferases/immunology , Bacterial Proteins/immunology , Cell Wall/immunology , Cysteine Endopeptidases/immunology , Immune Evasion/immunology , Staphylococcal Protein A/immunology , Staphylococcus aureus/immunology , Aminoacyltransferases/biosynthesis , Aminoacyltransferases/genetics , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cysteine Endopeptidases/biosynthesis , Cysteine Endopeptidases/genetics , Endopeptidases/metabolism , Humans , Peptidoglycan/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Signal Transduction , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Protein A/biosynthesis , Staphylococcal Protein A/genetics , Staphylococcus aureus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL