Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 12: 667387, 2021.
Article in English | MEDLINE | ID: mdl-34220813

ABSTRACT

The success of Staphylococcus aureus as a human commensal and an opportunistic pathogen relies on its ability to adapt to several niches within the host. The innate immune response plays a key role in protecting the host against S. aureus infection; however, S. aureus adeptness at evading the innate immune system is indisputably evident. The "Trojan horse" theory has been postulated to describe a mechanism by which S. aureus takes advantage of phagocytes as a survival niche within the host to facilitate dissemination of S. aureus to secondary sites during systemic infection. Several studies have determined that S. aureus can parasitize both professional and non-professional phagocytes by manipulating the host autophagy pathway in order to create an intracellular survival niche. Neutrophils represent a critical cell type in S. aureus infection as demonstrated by the increased risk of infection among patients with congenital neutrophil disorders. However, S. aureus has been repeatedly shown to survive intracellularly within neutrophils with evidence now supporting a pathogenic role of host autophagy. By manipulating this pathway, S. aureus can also alter the apoptotic fate of the neutrophil and potentially skew other important signalling pathways for its own gain. Understanding these critical host-pathogen interactions could lead to the development of new host directed therapeutics for the treatment of S. aureus infection by removing its intracellular niche and restoring host bactericidal functions. This review discusses the current findings surrounding intracellular survival of S. aureus within neutrophils, the pathogenic role autophagy plays in this process and considers the therapeutic potential for targeting this immune evasion mechanism.


Subject(s)
Autophagy , Neutrophils/immunology , Neutrophils/microbiology , Staphylococcus aureus/immunology , Humans , Immune Evasion , Immunity, Innate
2.
Front Immunol ; 11: 565545, 2020.
Article in English | MEDLINE | ID: mdl-33262756

ABSTRACT

Polymorphonuclear neutrophils (PMN) are critical for first line innate immune defence against Staphylococcus aureus. Mature circulating PMN maintain a short half-life ending in constitutive apoptotic cell death. This makes them unlikely candidates as a bacterial intracellular niche. However, there is significant evidence to suggest that S. aureus can survive intracellularly within PMN and this contributes to persistence and dissemination during infection. The precise mechanism by which S. aureus parasitizes these cells remains to be established. Herein we propose a novel mechanism by which S. aureus subverts both autophagy and apoptosis in PMN in order to maintain an intracellular survival niche during infection. Intracellular survival of S. aureus within primary human PMN was associated with an accumulation of the autophagic flux markers LC3-II and p62, while inhibition of the autophagy pathway led to a significant reduction in intracellular survival of bacteria. This intracellular survival of S. aureus was coupled with a delay in neutrophil apoptosis as well as increased expression of several anti-apoptotic factors. Importantly, blocking autophagy in infected PMN partially restored levels of apoptosis to that of uninfected PMN, suggesting a connection between the autophagic and apoptotic pathways during intracellular survival. These results provide a novel mechanism for S. aureus intracellular survival and suggest that S. aureus may be subverting crosstalk between the autophagic and apoptosis pathways in order to maintain an intracellular niche within human PMN.


Subject(s)
Apoptosis , Autophagy , Neutrophils/microbiology , Staphylococcus aureus , Humans , Microscopy, Electron, Transmission , Neutrophils/ultrastructure , Staphylococcus aureus/ultrastructure
3.
J Biol Chem ; 294(45): 17031-17042, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31558608

ABSTRACT

Pathogen activation of innate immune pattern recognition receptors (PRRs) such as Toll-like receptors (TLRs) stimulates cellular signaling pathways. This often leads to outcomes that contribute to pathogen clearance. Alternatively, activation of specific PRR pathways can aid pathogen survival. The human pathogen Staphylococcus aureus is a case in point, employing strategies to escape innate immune recognition and killing by the host. As for other bacteria, PRR-stimulated type I interferon (IFN-I) induction has been proposed as one such immune escape pathway that may favor S. aureus Cell wall components of S. aureus elicit TLR2-dependent cellular responses, but the exact signaling pathways activated by S. aureus-TLR2 engagement and the consequences of their activation for the host and bacterium are not fully known. We previously showed that TLR2 activates both a cytoplasmic and an endosome-dependent signaling pathway, the latter leading to IFN-I production. Here, we demonstrate that S. aureus infection of human monocytes activates a TLR2-dependent endosomal signaling pathway, leading to IFN-I induction. We mapped the signaling components of this pathway and identified roles in IFN-I stimulation for the Toll-interleukin-1 receptor (TIR) adaptor Myd88 adaptor-like (Mal), TNF receptor-associated factor 6 (TRAF6), and IκB kinase (IKK)-related kinases, but not for TRIF-related adaptor molecule (TRAM) and TRAF3. Importantly, monocyte TLR2-dependent endosomal signaling enabled immune escape for S. aureus, because this pathway, but not IFN-I per se, contributed to intracellular bacterial survival. These results reveal a TLR2-dependent mechanism in human monocytes whereby S. aureus manipulates innate immune signaling for its survival in cells.


Subject(s)
Endosomes/metabolism , Interferon Type I/metabolism , Microbial Viability , Monocytes/microbiology , Signal Transduction , Staphylococcus aureus/physiology , Toll-Like Receptor 2/metabolism , Animals , Cell Line , Humans , I-kappa B Kinase/metabolism , Mice , Monocytes/cytology , TNF Receptor-Associated Factor 6/metabolism
4.
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
5.
J Immunol ; 198(6): 2352-2365, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28167629

ABSTRACT

IL-10 is a potent anti-inflammatory mediator that plays a crucial role in limiting host immunopathology during bacterial infections by controlling effector T cell activation. Staphylococcus aureus has previously been shown to manipulate the IL-10 response as a mechanism of immune evasion during chronic systemic and biofilm models of infection. In the present study, we demonstrate divergent roles for IL-10 depending on the site of infection. During acute systemic S. aureus infection, IL-10 plays an important protective role and is required to prevent bacterial dissemination and host morbidity by controlling effector T cells and the associated downstream hyperactivation of inflammatory phagocytes, which are capable of host tissue damage. CD19+CD11b+CD5+ B1a regulatory cells were shown to rapidly express IL-10 in a TLR2-dependent manner in response to S. aureus, and adoptive transfer of B1a cells was protective during acute systemic infection in IL-10-deficient hosts. In contrast, during localized s.c. infection, IL-10 production plays a detrimental role by facilitating bacterial persistence via the same mechanism of controlling proinflammatory T cell responses. Our findings demonstrate that induction of IL-10 has a major influence on disease outcome during acute S. aureus infection. Too much IL-10 at one end of the scale may suppress otherwise protective T cell responses, thus facilitating persistence of the bacteria, and at the other end, too little IL-10 may tend toward fatal host-mediated pathology through excessive activation of T cells and associated phagocyte-mediated damage.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Bacteremia/immunology , Interleukin-10/metabolism , Peritonitis/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , T-Lymphocytes/immunology , Acute Disease , Animals , B-Lymphocytes, Regulatory/virology , Bacteremia/complications , Biofilms , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritonitis/microbiology , Staphylococcal Infections/complications , T-Lymphocytes/microbiology , Toll-Like Receptor 2/metabolism
6.
mBio ; 7(6)2016 12 13.
Article in English | MEDLINE | ID: mdl-27965455

ABSTRACT

Nasal carriage of Staphylococcus aureus is a significant risk factor for secondary staphylococcal pneumonia in influenza A virus (IAV)-infected hosts. However, little research has been undertaken to define the environmental and physiological changes that cause S. aureus to shift from commensal to pathogenic organism in this setting. The ability of virus-driven danger signals to cause S. aureus to transition from commensalism to pulmonary infection was explored in a recent study by Reddinger et al. R. M. Reddinger, N. R. Luke-Marshall, A. P. Hakansson, and A. A. Campagnari, mBio 7(6):e01235-16, 2016, http://dx.doi.org/10.1128/mBio.01235-16 The authors report that physiological host changes, including febrile temperature and a combination of host stress response signals, caused S. aureus biofilms to disperse from the nasal environment and cause active pulmonary infection. This commentary discusses the new finding in light of the current understanding of the mechanisms behind staphylococcal coinfection with IAV. In addition, it considers the mechanisms behind staphylococcal dispersal in this model. Overall, the study indicates that interkingdom signaling may occur following IAV infection and this likely contributes to sensitizing the IAV-infected host to secondary staphylococcal pneumonia.


Subject(s)
Influenza A virus , Staphylococcus aureus , Coinfection , Humans , Orthomyxoviridae Infections , Pneumonia, Staphylococcal , Staphylococcal Infections
7.
Trends Microbiol ; 24(11): 872-886, 2016 11.
Article in English | MEDLINE | ID: mdl-27474529

ABSTRACT

Staphylococcus aureus persistently colonizes the anterior nares of approximately one fifth of the population and nasal carriage is a significant risk factor for infection. Recent advances have significantly refined our understanding of S. aureus-host communication during nasal colonization. Novel bacterial adherence mechanisms in the nasal epithelium have been identified, and novel roles for both the innate and the adaptive immune response in controlling S. aureus nasal colonization have been defined, through the use of both human and rodent models. It is clear that S. aureus maintains a unique, complex relationship with the host immune system and that S. aureus nasal colonization is overall a multifactorial process which is as yet incompletely understood.


Subject(s)
Host-Pathogen Interactions/immunology , Nasal Cavity/microbiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/growth & development , Staphylococcus aureus/immunology , Adaptive Immunity , Animals , Bacterial Adhesion , Carrier State/diagnosis , Carrier State/immunology , Carrier State/microbiology , Disease Models, Animal , Epithelial Cells/microbiology , Host-Pathogen Interactions/physiology , Humans , Immunity, Innate , Microbial Interactions/immunology , Microbiota , Nasal Cavity/immunology , Nasal Mucosa/immunology , Nasal Mucosa/microbiology , Nose/immunology , Nose/microbiology , Risk Factors , Rodentia , Staphylococcal Infections/immunology , Staphylococcal Infections/transmission , Staphylococcus aureus/pathogenicity , Staphylococcus aureus/physiology
8.
Infect Immun ; 81(12): 4478-89, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24082072

ABSTRACT

Recent work has identified T cells and the cytokines they produce as important correlates of immune protection during Staphylococcus aureus infections through the ability of these T cells to regulate local neutrophil responses. However, the specific T-cell subsets that are involved in coordinating protection at distinct sites of infection remains to be established. In this study, we identify for the first time an important role for γδT cells in controlling S. aureus surgical site infection (SSI). γδT cells are recruited to the wound site following S. aureus challenge, where they represent the primary source of interleukin 17 (IL-17), with a small contribution from other non-γδT cells. The IL-17 response is entirely dependent upon IL-1 receptor signaling. Using IL-17 receptor-deficient mice, we demonstrate that IL-17 is required to control bacterial clearance during S. aureus SSI. However, we demonstrate a strain-dependent requirement for γδT cells in this process due to the differential abilities of individual strains to activate IL-1ß production. IL-1ß processing relies upon activation of the Nlrp3 inflammasome complex, and we demonstrate that Nlrp3-deficient and IL-1 receptor-deficient mice have an impaired ability to control S. aureus SSI due to reduced production of IL-17 by γδT cells at the site of infection. Given that IL-17 has been identified as an important correlate of immune protection during S. aureus infection, it is vital that the unique cellular sources of this cytokine and mechanisms inducing its activation are identified at distinct sites of infection. Our study demonstrates that while IL-17 may be critically important for mediating immune protection during S. aureus SSI, the relative contribution of γδT cells to these protective effects may be strain dependent.


Subject(s)
Carrier Proteins/metabolism , Interleukin-17/immunology , Staphylococcal Infections/immunology , Surgical Wound Infection/immunology , T-Lymphocytes/immunology , Animals , Carrier Proteins/genetics , Interleukin-17/biosynthesis , Interleukin-17/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Neutrophils/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/genetics , Signal Transduction/immunology , Staphylococcus aureus/immunology , T-Lymphocytes/metabolism
9.
PLoS Pathog ; 8(12): e1003092, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23300445

ABSTRACT

Staphylococcus aureus asymptomatically colonises the anterior nares, but the host and bacterial factors that facilitate colonisation remain incompletely understood. The S. aureus surface protein ClfB has been shown to mediate adherence to squamous epithelial cells in vitro and to promote nasal colonisation in both mice and humans. Here, we demonstrate that the squamous epithelial cell envelope protein loricrin represents the major target ligand for ClfB during S. aureus nasal colonisation. In vitro adherence assays indicated that bacteria expressing ClfB bound loricrin most likely by the "dock, lock and latch" mechanism. Using surface plasmon resonance we showed that ClfB bound cytokeratin 10 (K10), a structural protein of squamous epithelial cells, and loricrin with similar affinities that were in the low µM range. Loricrin is composed of three separate regions comprising GS-rich omega loops. Each loop was expressed separately and found to bind ClfB, However region 2 bound with highest affinity. To investigate if the specific interaction between ClfB and loricrin was sufficient to facilitate S. aureus nasal colonisation, we compared the ability of ClfB⁺S. aureus to colonise the nares of wild-type and loricrin-deficient (Lor⁻/⁻) mice. In the absence of loricrin, S. aureus nasal colonisation was significantly impaired. Furthermore a ClfB⁻ mutant colonised wild-type mice less efficiently than the parental ClfB⁺ strain whereas a similar lower level of colonisation was observed with both the parental strain and the ClfB⁻ mutant in the Lor⁻/⁻ mice. The ability of ClfB to support nasal colonisation by binding loricrin in vivo was confirmed by the ability of Lactococcus lactis expressing ClfB to be retained in the nares of WT mice but not in the Lor⁻/⁻ mice. By combining in vitro biochemical analysis with animal model studies we have identified the squamous epithelial cell envelope protein loricrin as the target ligand for ClfB during nasal colonisation by S. aureus.


Subject(s)
Adhesins, Bacterial/metabolism , Coagulase/metabolism , Membrane Proteins/metabolism , Nose/microbiology , Staphylococcus aureus/metabolism , Adhesins, Bacterial/genetics , Animals , Bacterial Adhesion/physiology , Cell Line , Epithelial Cells/microbiology , Female , Humans , Keratin-10/metabolism , Lactococcus lactis/metabolism , Membrane Proteins/genetics , Mice , Mice, Knockout , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL
...