Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Cell Sci ; 137(5)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38345099

ABSTRACT

Glycosylated mucin proteins contribute to the essential barrier function of the intestinal epithelium. The transmembrane mucin MUC13 is an abundant intestinal glycoprotein with important functions for mucosal maintenance that are not yet completely understood. We demonstrate that in human intestinal epithelial monolayers, MUC13 localized to both the apical surface and the tight junction (TJ) region on the lateral membrane. MUC13 deletion resulted in increased transepithelial resistance (TEER) and reduced translocation of small solutes. TEER buildup in ΔMUC13 cells could be prevented by addition of MLCK, ROCK or protein kinase C (PKC) inhibitors. The levels of TJ proteins including claudins and occludin were highly increased in membrane fractions of MUC13 knockout cells. Removal of the MUC13 cytoplasmic tail (CT) also altered TJ composition but did not affect TEER. The increased buildup of TJ complexes in ΔMUC13 and MUC13-ΔCT cells was dependent on PKC. The responsible PKC member might be PKCδ (or PRKCD) based on elevated protein levels in the absence of full-length MUC13. Our results demonstrate for the first time that a mucin protein can negatively regulate TJ function and stimulate intestinal barrier permeability.


Subject(s)
Protein Kinase C , Tight Junction Proteins , Humans , Tight Junction Proteins/metabolism , Protein Kinase C/metabolism , Intestines , Intestinal Mucosa/metabolism , Tight Junctions/metabolism , Occludin , Mucins/metabolism , Epithelial Cells/metabolism
2.
Front Cell Infect Microbiol ; 13: 1186936, 2023.
Article in English | MEDLINE | ID: mdl-37342248

ABSTRACT

Background: Despite the extensive use of silver ions or nanoparticles in research related to preventing implant-associated infections (IAI), their use in clinical practice has been debated. This is because the strong antibacterial properties of silver are counterbalanced by adverse effects on host cells. One of the reasons for this may be the lack of comprehensive in vitro models that are capable of analyzing host-bacteria and host-host interactions. Methods and results: In this study, we tested silver efficacy through multicellular in vitro models involving macrophages (immune system), mesenchymal stem cells (MSCs, bone cells), and S. aureus (pathogen). Our model showed to be capable of identifying each element of culture as well as tracking the intracellular survival of bacteria. Furthermore, the model enabled to find a therapeutic window for silver ions (AgNO3) and silver nanoparticles (AgNPs) where the viability of host cells was not compromised, and the antibacterial properties of silver were maintained. While AgNO3 between 0.00017 and 0.017 µg/mL retained antibacterial properties, host cell viability was not affected. The multicellular model, however, demonstrated that those concentrations had no effect on the survival of S. aureus, inside or outside host cells. Similarly, treatment with 20 nm AgNPs did not influence the phagocytic and killing capacity of macrophages or prevent S. aureus from invading MSCs. Moreover, exposure to 100 nm AgNPs elicited an inflammatory response by host cells as detected by the increased production of TNF-α and IL-6. This was visible only when macrophages and MSCs were cultured together. Conclusions: Multicellular in vitro models such as the one used here that simulate complex in vivo scenarios can be used to screen other therapeutic compounds or antibacterial biomaterials without the need to use animals.


Subject(s)
Metal Nanoparticles , Silver , Animals , Silver/pharmacology , Staphylococcus aureus , Anti-Bacterial Agents/pharmacology , Bacteria , Microbial Sensitivity Tests
3.
J Immunol ; 210(4): 389-397, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36637221

ABSTRACT

Signal inhibitory receptor on leukocytes-1 (SIRL-1) is an immune inhibitory receptor expressed on human granulocytes and monocytes that dampens antimicrobial functions. We previously showed that sputum neutrophils from infants with severe respiratory syncytial virus (RSV) bronchiolitis have decreased SIRL-1 surface expression compared with blood neutrophils and that SIRL-1 surface expression is rapidly lost from in vitro activated neutrophils. This led us to hypothesize that activated neutrophils lose SIRL-1 by ectodomain shedding. Here, we developed an ELISA and measured the concentration of soluble SIRL-1 (sSIRL-1) in patients with RSV bronchiolitis and hospitalized patients with COVID-19, which are both characterized by neutrophilic inflammation. In line with our hypothesis, sSIRL-1 concentration was increased in sputum compared with plasma of patients with RSV bronchiolitis and in serum of hospitalized patients with COVID-19 compared with control serum. In addition, we show that in vitro activated neutrophils release sSIRL-1 by proteolytic cleavage and that this diminishes the ability to inhibit neutrophilic reactive oxygen species production via SIRL-1. Finally, we found that SIRL-1 shedding is prevented by proteinase 3 inhibition and by extracellular adherence protein from Staphylococcus aureus. Notably, we recently showed that SIRL-1 is activated by PSMα3 from S. aureus, suggesting that S. aureus may counteract SIRL-1 shedding to benefit from preserved inhibitory function of SIRL-1. In conclusion, we report that SIRL-1 is released from activated neutrophils by proteinase 3 cleavage and that endogenous sSIRL-1 protein is present in vivo.


Subject(s)
Bronchiolitis , COVID-19 , Respiratory Syncytial Virus Infections , Humans , Infant , Bronchiolitis/metabolism , COVID-19/metabolism , Myeloblastin , Neutrophils , Receptors, Immunologic , Staphylococcus aureus , Leukocytes/metabolism
5.
PLoS Pathog ; 17(11): e1010051, 2021 11.
Article in English | MEDLINE | ID: mdl-34752492

ABSTRACT

Complement proteins can form membrane attack complex (MAC) pores that directly kill Gram-negative bacteria. MAC pores assemble by stepwise binding of C5b, C6, C7, C8 and finally C9, which can polymerize into a transmembrane ring of up to 18 C9 monomers. It is still unclear if the assembly of a polymeric-C9 ring is necessary to sufficiently damage the bacterial cell envelope to kill bacteria. In this paper, polymerization of C9 was prevented without affecting binding of C9 to C5b-8, by locking the first transmembrane helix domain of C9. Using this system, we show that polymerization of C9 strongly enhanced damage to both the bacterial outer and inner membrane, resulting in more rapid killing of several Escherichia coli and Klebsiella strains in serum. By comparing binding of wildtype and 'locked' C9 by flow cytometry, we also show that polymerization of C9 is impaired when the amount of available C9 per C5b-8 is limited. This suggests that an excess of C9 is required to efficiently form polymeric-C9. Finally, we show that polymerization of C9 was impaired on complement-resistant E. coli strains that survive killing by MAC pores. This suggests that these bacteria can specifically block polymerization of C9. All tested complement-resistant E. coli expressed LPS O-antigen (O-Ag), compared to only one out of four complement-sensitive E. coli. By restoring O-Ag expression in an O-Ag negative strain, we show that the O-Ag impairs polymerization of C9 and results in complement-resistance. Altogether, these insights are important to understand how MAC pores kill bacteria and how bacterial pathogens can resist MAC-dependent killing.


Subject(s)
Blood Bactericidal Activity , Cell Wall/pathology , Complement C9/chemistry , Complement Membrane Attack Complex/pharmacology , Escherichia coli/growth & development , Klebsiella/growth & development , Polymerization , Cell Wall/drug effects , Escherichia coli/drug effects , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Humans , Klebsiella/drug effects , Klebsiella Infections/drug therapy , Klebsiella Infections/microbiology
6.
J Biol Chem ; 295(22): 7753-7762, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32303641

ABSTRACT

Members of the EAP family of Staphylococcus aureus immune evasion proteins potently inhibit the neutrophil serine proteases (NSPs) neutrophil elastase, cathepsin-G, and proteinase-3. Previously, we determined a 1.8 Å resolution crystal structure of the EAP family member EapH1 bound to neutrophil elastase. This structure revealed that EapH1 blocks access to the enzyme's active site by forming a noncovalent complex with this host protease. To determine how EapH1 inhibits other NSPs, we studied here the effects of EapH1 on cathepsin-G. We found that EapH1 inhibits cathepsin-G with a Ki of 9.8 ± 4.7 nm Although this Ki value is ∼466-fold weaker than the Ki for EapH1 inhibition of neutrophil elastase, the time dependence of inhibition was maintained. To define the physical basis for EapH1's inhibition of cathepsin-G, we crystallized EapH1 bound to this protease, solved the structure at 1.6 Å resolution, and refined the model to Rwork and Rfree values of 17.4% and 20.9%, respectively. This structure revealed a protease-binding mode for EapH1 with cathepsin-G that was globally similar to that seen in the previously determined EapH1-neutrophil elastase structure. The nature of the intermolecular interactions formed by EapH1 with cathepsin-G differed considerably from that with neutrophil elastase, however, with far greater contributions from the inhibitor backbone in the cathepsin-G-bound form. Together, these results reveal that EapH1's ability to form high-affinity interactions with multiple NSP targets is due to its remarkable level of local structural plasticity.


Subject(s)
Bacterial Proteins/chemistry , Cathepsin G/chemistry , Leukocyte Elastase/chemistry , Myeloblastin/chemistry , Neutrophils/enzymology , Serine Proteinase Inhibitors/chemistry , Staphylococcus aureus/chemistry , Crystallography, X-Ray , Humans , Protein Structure, Quaternary
7.
Cell Host Microbe ; 27(1): 41-53.e6, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31862381

ABSTRACT

Many Gram-negative bacterial pathogens antagonize anti-bacterial immunity through translocated effector proteins that inhibit pro-inflammatory signaling. In addition, the intracellular pathogen Salmonella enterica serovar Typhimurium initiates an anti-inflammatory transcriptional response in macrophages through its effector protein SteE. However, the target(s) and molecular mechanism of SteE remain unknown. Here, we demonstrate that SteE converts both the amino acid and substrate specificity of the host pleiotropic serine/threonine kinase GSK3. SteE itself is a substrate of GSK3, and phosphorylation of SteE is required for its activity. Remarkably, phosphorylated SteE then forces GSK3 to phosphorylate the non-canonical substrate signal transducer and activator of transcription 3 (STAT3) on tyrosine-705. This results in STAT3 activation, which along with GSK3 is required for SteE-mediated upregulation of the anti-inflammatory M2 macrophage marker interleukin-4Rα (IL-4Rα). Overall, the conversion of GSK3 to a tyrosine-directed kinase represents a tightly regulated event that enables a bacterial virulence protein to reprogram innate immune signaling and establish an anti-inflammatory environment.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Macrophages/microbiology , Protein Serine-Threonine Kinases/metabolism , STAT3 Transcription Factor/metabolism , Salmonella typhimurium , Animals , Bacterial Proteins/metabolism , HEK293 Cells , HeLa Cells , Host Microbial Interactions/immunology , Humans , Interleukin-4/metabolism , Macrophage Activation , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Protein-Tyrosine Kinases/metabolism , Salmonella typhimurium/immunology , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Virulence/immunology
8.
Science ; 362(6419): 1156-1160, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30523110

ABSTRACT

Many bacterial infections are hard to treat and tend to relapse, possibly due to the presence of antibiotic-tolerant persisters. In vitro, persister cells appear to be dormant. After uptake of Salmonella species by macrophages, nongrowing persisters also occur, but their physiological state is poorly understood. In this work, we show that Salmonella persisters arising during macrophage infection maintain a metabolically active state. Persisters reprogram macrophages by means of effectors secreted by the Salmonella pathogenicity island 2 type 3 secretion system. These effectors dampened proinflammatory innate immune responses and induced anti-inflammatory macrophage polarization. Such reprogramming allowed nongrowing Salmonella cells to survive for extended periods in their host. Persisters undermining host immune defenses might confer an advantage to the pathogen during relapse once antibiotic pressure is relieved.


Subject(s)
Drug Resistance, Bacterial , Host-Pathogen Interactions/immunology , Macrophages/immunology , Salmonella Infections/drug therapy , Salmonella Infections/immunology , Salmonella typhimurium/metabolism , Type III Secretion Systems/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Cells, Cultured , Female , Genomic Islands , Immunity, Innate , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Recurrence , Salmonella Infections/microbiology , Salmonella typhimurium/drug effects
9.
Protein Sci ; 27(2): 509-522, 2018 02.
Article in English | MEDLINE | ID: mdl-29114958

ABSTRACT

Neutrophils contain high levels of chymotrypsin-like serine proteases (NSPs) within their azurophilic granules that have a multitude of functions within the immune system. In response, the pathogen Staphylococcus aureus has evolved three potent inhibitors (Eap, EapH1, and EapH2) that protect the bacterium as well as several of its secreted virulence factors from the degradative action of NSPs. We previously showed that these so-called EAP domain proteins represent a novel class of NSP inhibitors characterized by a non-covalent inhibitory mechanism and a distinct target specificity profile. Based upon high levels of structural homology amongst the EAP proteins and the NSPs, as well as supporting biochemical data, we predicted that the inhibited complex would be similar for all EAP/NSP pairs. However, we present here evidence that EapH1 and EapH2 bind the canonical NSP, Neutrophil Elastase (NE), in distinct orientations. We discovered that alteration of EapH1 residues at the EapH1/NE interface caused a dramatic loss of affinity and inhibition of NE, while mutation of equivalent positions in EapH2 had no effect on NE binding or inhibition. Surprisingly, mutation of residues in an altogether different region of EapH2 severely impacted both the NE binding and inhibitory properties of EapH2. Even though EapH1 and EapH2 bind and inhibit NE and a second NSP, Cathepsin G, equally well, neither of these proteins interacts with the structurally related, but non-proteolytic granule protein, azurocidin. These studies expand our understanding of EAP/NSP interactions and suggest that members of this immune evasion protein family are capable of diverse target recognition modes.


Subject(s)
Leukocyte Elastase/metabolism , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/metabolism , Staphylococcus aureus/immunology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Cell Line , Humans , Immune Evasion , Models, Molecular , Mutation , Protein Binding , Protein Domains , Serine Proteinase Inhibitors/genetics , Staphylococcus aureus/enzymology
10.
Nat Microbiol ; 2: 16206, 2016 Nov 14.
Article in English | MEDLINE | ID: mdl-27841856

ABSTRACT

Intracellular bacterial pathogens can exhibit large heterogeneity in growth rate inside host cells, with major consequences for the infection outcome. If and how the host responds to this heterogeneity remains poorly understood. Here, we combined a fluorescent reporter of bacterial cell division with single-cell RNA-sequencing analysis to study the macrophage response to different intracellular states of the model pathogen Salmonella enterica serovar Typhimurium. The transcriptomes of individual infected macrophages revealed a spectrum of functional host response states to growing and non-growing bacteria. Intriguingly, macrophages harbouring non-growing Salmonella display hallmarks of the proinflammatory M1 polarization state and differ little from bystander cells, suggesting that non-growing bacteria evade recognition by intracellular immune receptors. By contrast, macrophages containing growing bacteria have turned into an anti-inflammatory, M2-like state, as if fast-growing intracellular Salmonella overcome host defence by reprogramming macrophage polarization. Additionally, our clustering approach reveals intermediate host functional states between these extremes. Altogether, our data suggest that gene expression variability in infected host cells shapes different cellular environments, some of which may favour a growth arrest of Salmonella facilitating immune evasion and the establishment of a long-term niche, while others allow Salmonella to escape intracellular antimicrobial activity and proliferate.


Subject(s)
Cell Differentiation , Gene Expression Profiling , Macrophages/microbiology , Macrophages/physiology , Salmonella typhimurium/growth & development , Animals , Biological Variation, Population , Cells, Cultured , Host-Pathogen Interactions , Mice, Inbred C57BL , Sequence Analysis, RNA , Single-Cell Analysis
11.
Microbiology (Reading) ; 162(7): 1185-1194, 2016 07.
Article in English | MEDLINE | ID: mdl-27112346

ABSTRACT

Staphylococcus aureus has developed many mechanisms to escape from human immune responses. To resist phagocytic clearance, S. aureus expresses a polysaccharide capsule, which effectively masks the bacterial surface and surface-associated proteins, such as opsonins, from recognition by phagocytic cells. Additionally, secretion of the extracellular fibrinogen binding protein (Efb) potently blocks phagocytic uptake of the pathogen. Efb creates a fibrinogen shield surrounding the bacteria by simultaneously binding complement C3b and fibrinogen at the bacterial surface. By means of neutrophil phagocytosis assays with fluorescently labelled encapsulated serotype 5 (CP5) and serotype 8 (CP8) strains we compare the immune-modulating function of these shielding mechanisms. The data indicate that, in highly encapsulated S. aureus strains, the polysaccharide capsule is able to prevent phagocytic uptake at plasma concentrations <10 %, but loses its protective ability at higher concentrations of plasma. Interestingly, Efb shows a strong inhibitory effect on both capsule-negative and encapsulated strains at all tested plasma concentrations. Furthermore, the results suggest that both shielding mechanisms can exist simultaneously and collaborate to provide optimal protection against phagocytosis at a broad range of plasma concentrations. As opsonizing antibodies will be shielded from recognition by either mechanism, incorporating both capsular polysaccharides and Efb in future vaccines could be of great importance.


Subject(s)
Bacterial Capsules/metabolism , Bacterial Proteins/metabolism , Fibrinogen/metabolism , Neutrophils/immunology , Phagocytosis/immunology , Polysaccharides, Bacterial/metabolism , Staphylococcus aureus/immunology , Antibodies, Bacterial/immunology , Complement C3b/metabolism , Fluorescent Dyes , Humans , Immunomodulation/immunology , Microscopy, Confocal , Opsonin Proteins/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/metabolism
12.
Curr Opin Microbiol ; 23: 42-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25461571

ABSTRACT

Neutrophil serine proteases (NSPs) are critical for the effective functioning of neutrophils and greatly contribute to immune protection against bacterial infections. Thanks to their broad substrate specificity, these chymotrypsin-like proteases trigger multiple reactions that are detrimental to bacterial survival such as direct bacterial killing, generation of antimicrobial peptides, inactivation of bacterial virulence factors and formation of neutrophil extracellular traps. Recently, the importance of NSPs in antibacterial defenses has been further underscored by discoveries of unique bacterial evasion strategies to combat these proteases. Bacteria can indirectly disarm NSPs by protecting bacterial substrates against NSP cleavage, but also produce inhibitory molecules that potently block NSPs. Here we review recent insights in the functional contribution of NSPs in host protection against bacterial infections and the elegant strategies that bacteria use to counteract these responses.


Subject(s)
Bacteria/immunology , Bacterial Infections/immunology , Host-Pathogen Interactions , Immune Evasion , Neutrophils/enzymology , Neutrophils/immunology , Serine Proteases/metabolism , Allergy and Immunology/trends , Animals , Biomedical Research/trends , Humans , Microbiology/trends , Neutrophils/microbiology
13.
J Immunol ; 193(12): 6161-6171, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25381436

ABSTRACT

The pathogenic bacterium Staphylococcus aureus actively evades many aspects of human innate immunity by expressing a series of small inhibitory proteins. A number of these proteins inhibit the complement system, which labels bacteria for phagocytosis and generates inflammatory chemoattractants. Although the majority of staphylococcal complement inhibitors act on the alternative pathway to block the amplification loop, only a few proteins act on the initial recognition cascades that constitute the classical pathway (CP) and lectin pathway (LP). We screened a collection of recombinant, secreted staphylococcal proteins to determine whether S. aureus produces other molecules that inhibit the CP and/or LP. Using this approach, we identified the extracellular adherence protein (Eap) as a potent, specific inhibitor of both the CP and LP. We found that Eap blocked CP/LP-dependent activation of C3, but not C4, and that Eap likewise inhibited deposition of C3b on the surface of S. aureus cells. In turn, this significantly diminished the extent of S. aureus opsonophagocytosis and killing by neutrophils. This combination of functional properties suggested that Eap acts specifically at the level of the CP/LP C3 convertase (C4b2a). Indeed, we demonstrated a direct, nanomolar-affinity interaction of Eap with C4b. Eap binding to C4b inhibited binding of both full-length C2 and its C2b fragment, which indicated that Eap disrupts formation of the CP/LP C3 proconvertase (C4b2). As a whole, our results demonstrate that S. aureus inhibits two initiation routes of complement by expression of the Eap protein, and thereby define a novel mechanism of immune evasion.


Subject(s)
Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Complement C3 Convertase, Alternative Pathway/antagonists & inhibitors , Complement Pathway, Classical/immunology , Complement Pathway, Mannose-Binding Lectin/immunology , RNA-Binding Proteins/immunology , RNA-Binding Proteins/metabolism , Staphylococcus aureus/immunology , Staphylococcus aureus/metabolism , Bacterial Proteins/chemistry , Binding Sites , Complement C2/immunology , Complement C2/metabolism , Complement C3b/immunology , Complement C3b/metabolism , Complement C4b/immunology , Complement C4b/metabolism , Cytotoxicity, Immunologic , Humans , Models, Immunological , Neutrophils/immunology , Phagocytosis/immunology , Protein Binding , Protein Interaction Domains and Motifs , RNA-Binding Proteins/chemistry , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism
14.
J Immunol ; 193(11): 5506-14, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25355925

ABSTRACT

The uptake of Ag-Ab immune complexes (IC) after the ligation of activating FcγR on dendritic cells (DC) leads to 100 times more efficient Ag presentation than the uptake of free Ags. FcγRs were reported to facilitate IC uptake and simultaneously induce cellular activation that drives DC maturation and mediates efficient T cell activation. Activating FcγRs elicit intracellular signaling via the ITAM domain of the associated FcRγ-chain. Studies with FcRγ-chain knockout (FcRγ(-/-)) mice reported FcRγ-chain ITAM signaling to be responsible for enhancing both IC uptake and DC maturation. However, FcRγ-chain is also required for surface expression of activating FcγRs, hampering the dissection of ITAM-dependent and independent FcγR functions in FcRγ(-/-) DCs. In this work, we studied the role of FcRγ-chain ITAM signaling using DCs from NOTAM mice that express normal surface levels of activating FcγR, but lack functional ITAM signaling. IC uptake by bone marrow-derived NOTAM DCs was reduced compared with wild-type DCs, but was not completely absent as in FcRγ(-/-) DCs. In NOTAM DCs, despite the uptake of ICs, both MHC class I and MHC class II Ag presentation was completely abrogated similar to FcRγ(-/-) DCs. Secretion of cytokines, upregulation of costimulatory molecules, and Ag degradation were abrogated in NOTAM DCs in response to FcγR ligation. Cross-presentation using splenic NOTAM DCs and prolonged incubation with OVA-IC was also abrogated. Interestingly, in this setup, proliferation of CD4(+) OT-II cells was induced by NOTAM DCs. We conclude that FcRγ-chain ITAM signaling facilitates IC uptake and is essentially required for cross-presentation, but not for MHC class II Ag presentation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Receptors, IgG/metabolism , Animals , Antigen-Antibody Complex/immunology , Antigens, CD/metabolism , Cell Differentiation/genetics , Cells, Cultured , Cross-Priming/genetics , Endocytosis/genetics , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/metabolism , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Structure, Tertiary/genetics , Receptors, IgG/genetics , Signal Transduction
15.
Proc Natl Acad Sci U S A ; 111(36): 13187-92, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25161283

ABSTRACT

Neutrophils are indispensable for clearing infections with the prominent human pathogen Staphylococcus aureus. Here, we report that S. aureus secretes a family of proteins that potently inhibits the activity of neutrophil serine proteases (NSPs): neutrophil elastase (NE), proteinase 3, and cathepsin G. The NSPs, but not related serine proteases, are specifically blocked by the extracellular adherence protein (Eap) and the functionally orphan Eap homologs EapH1 and EapH2, with inhibitory-constant values in the low-nanomolar range. Eap proteins are together essential for NSP inhibition by S. aureus in vitro and promote staphylococcal infection in vivo. The crystal structure of the EapH1/NE complex showed that Eap molecules constitute a unique class of noncovalent protease inhibitors that occlude the catalytic cleft of NSPs. These findings increase our insights into the complex pathogenesis of S. aureus infections and create opportunities to design novel treatment strategies for inflammatory conditions related to excessive NSP activity.


Subject(s)
Neutrophils/metabolism , Serine Proteinase Inhibitors/metabolism , Staphylococcus aureus/metabolism , Animals , Bacterial Adhesion , Bacterial Proteins/metabolism , Biocatalysis , Extracellular Space/metabolism , Female , Humans , Leukocyte Elastase/antagonists & inhibitors , Leukocyte Elastase/metabolism , Mice, Inbred C57BL , Models, Molecular , Staphylococcal Infections/pathology
16.
J Innate Immun ; 6(6): 860-8, 2014.
Article in English | MEDLINE | ID: mdl-25012862

ABSTRACT

Neutrophil extracellular traps (NETs) have been described as a fundamental innate immune defence mechanism. They consist of a nuclear DNA backbone associated with different antimicrobial peptides (AMPs) which are able to engulf and kill pathogens. The AMP LL-37, a member of the cathelicidin family, is highly present in NETs. However, the function of LL-37 within NETs is still unknown because it loses its antimicrobial activity when bound to DNA in the NETs. Using immunofluorescence microscopy, we demonstrate that NETs treated with LL-37 are distinctly more resistant to S. aureus nuclease degradation than nontreated NETs. Biochemical assays utilising a random LL-37-fragment library indicated that the blocking effect of LL-37 on nuclease activity is based on the cationic character of the AMP, which facilitates the binding to neutrophil DNA, thus protecting it from degradation by the nuclease. In good correlation to these data, the cationic AMPs human beta defensin-3 and human neutrophil peptide-1 showed similar protection of neutrophil-derived DNA against nuclease degradation. In conclusion, this study demonstrates a novel role of AMPs in host immune defence: beside its direct antimicrobial activity against various pathogens, cationic AMPs can stabilise neutrophil-derived DNA or NETs against bacterial nuclease degradation.


Subject(s)
Bacterial Proteins/immunology , Cathelicidins/immunology , Extracellular Traps/immunology , Micrococcal Nuclease/immunology , Neutrophils/immunology , Staphylococcus aureus/immunology , Antimicrobial Cationic Peptides , Bacterial Proteins/metabolism , Cathelicidins/metabolism , Extracellular Traps/metabolism , Extracellular Traps/microbiology , Female , Humans , Male , Micrococcal Nuclease/metabolism , Neutrophils/metabolism , Staphylococcus aureus/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...