Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 82
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Cell Death Discov ; 10(1): 403, 2024 Sep 13.
Article de Anglais | MEDLINE | ID: mdl-39271670

RÉSUMÉ

Staphylococcus aureus is an important human commensal which persistently colonizes up to 30% of the human population, predominantly within the nasal cavity. The commensal lifestyle of S. aureus is complex, and the mechanisms underpinning colonization are not fully understood. S. aureus can induce an immunosuppressive environment in the nasal tissue (NT) by driving IL-10 and IL-27 to facilitate nasal colonization, indicating that S. aureus has the capacity to modulate the local immune environment for its commensal habitation. Mounting evidence suggests commensal bacteria drive type 1 interferons (IFN-I) to establish an immunosuppressive environment and whilst S. aureus can induce IFN-I during infection, its role in colonization has not yet been examined. Here, we show that S. aureus preferentially induces IFN signaling in macrophages. This IFN-I in turn upregulates expression of proapoptotic genes within macrophages culminating in caspase-3 cleavage. Importantly, S. aureus was found to drive phagocytic cell apoptosis in the nasal tissue during nasal colonization in an IFN-I dependent manner with colonization significantly reduced under caspase-3 inhibition. Overall, loss of IFN-I signaling significantly diminished S. aureus nasal colonization implicating a pivotal role for IFN-I in controlling S. aureus persistence during colonization through its ability to induce phagocyte apoptosis. Together, this study reveals a novel strategy utilized by S. aureus to circumvent host immunity in the nasal mucosa to facilitate nasal colonization.

2.
JCI Insight ; 9(13)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38973612

RÉSUMÉ

Staphylococcus aureus is a major human pathogen. An effective anti-S. aureus vaccine remains elusive as the correlates of protection are ill-defined. Targeting specific T cell populations is an important strategy for improving anti-S. aureus vaccine efficacy. Potential bottlenecks that remain are S. aureus-induced immunosuppression and the impact this might have on vaccine-induced immunity. S. aureus induces IL-10, which impedes effector T cell responses, facilitating persistence during both colonization and infection. Thus, it was hypothesized that transient targeting of IL-10 might represent an innovative way to improve vaccine efficacy. In this study, IL-10 expression was elevated in the nares of persistent carriers of S. aureus, and this was associated with reduced systemic S. aureus-specific Th1 responses. This suggests that systemic responses are remodeled because of commensal exposure to S. aureus, which negatively implicates vaccine function. To provide proof of concept that targeting immunosuppressive responses during immunization may be a useful approach to improve vaccine efficacy, we immunized mice with T cell-activating vaccines in combination with IL-10-neutralizing antibodies. Blocking IL-10 during vaccination enhanced effector T cell responses and improved bacterial clearance during subsequent systemic and subcutaneous infection. Taken together, these results reveal a potentially novel strategy for improving anti-S. aureus vaccine efficacy.


Sujet(s)
Interleukine-10 , Infections à staphylocoques , Vaccins antistaphylococciques , Staphylococcus aureus , Interleukine-10/métabolisme , Interleukine-10/immunologie , Animaux , Infections à staphylocoques/prévention et contrôle , Infections à staphylocoques/immunologie , Vaccins antistaphylococciques/immunologie , Souris , Staphylococcus aureus/immunologie , Femelle , Souris de lignée C57BL , Lymphocytes auxiliaires Th1/immunologie , Immunisation/méthodes , Humains , Anticorps neutralisants/immunologie , 59641 , Vaccination/méthodes
3.
JCI Insight ; 9(9)2024 Apr 11.
Article de Anglais | MEDLINE | ID: mdl-38716729

RÉSUMÉ

Atopic dermatitis (AD) is an inflammatory skin condition with a childhood prevalence of up to 25%. Microbial dysbiosis is characteristic of AD, with Staphylococcus aureus the most frequent pathogen associated with disease flares and increasingly implicated in disease pathogenesis. Therapeutics to mitigate the effects of S. aureus have had limited efficacy and S. aureus-associated temporal disease flares are synonymous with AD. An alternative approach is an anti-S. aureus vaccine, tailored to AD. Experimental vaccines have highlighted the importance of T cells in conferring protective anti-S. aureus responses; however, correlates of T cell immunity against S. aureus in AD have not been identified. We identify a systemic and cutaneous immunological signature associated with S. aureus skin infection (ADS.aureus) in a pediatric AD cohort, using a combined Bayesian multinomial analysis. ADS.aureus was most highly associated with elevated cutaneous chemokines IP10 and TARC, which preferentially direct Th1 and Th2 cells to skin. Systemic CD4+ and CD8+ T cells, except for Th2 cells, were suppressed in ADS.aureus, particularly circulating Th1, memory IL-10+ T cells, and skin-homing memory Th17 cells. Systemic γδ T cell expansion in ADS.aureus was also observed. This study suggests that augmentation of protective T cell subsets is a potential therapeutic strategy in the management of S. aureus in AD.


Sujet(s)
Eczéma atopique , Infections cutanées à staphylocoques , Staphylococcus aureus , Eczéma atopique/immunologie , Eczéma atopique/microbiologie , Humains , Staphylococcus aureus/immunologie , Enfant , Femelle , Infections cutanées à staphylocoques/immunologie , Infections cutanées à staphylocoques/microbiologie , Mâle , Enfant d'âge préscolaire , Peau/microbiologie , Peau/immunologie , Peau/anatomopathologie , Chimiokine CXCL10/immunologie , Chimiokine CXCL10/métabolisme , Lymphocytes auxiliaires Th1/immunologie , Lymphocytes auxiliaires Th2/immunologie , Cellules Th17/immunologie , Théorème de Bayes , Lymphocytes T CD8+/immunologie , Interleukine-10/métabolisme , Interleukine-10/immunologie , Lymphocytes intra-épithéliaux/immunologie , Antigènes de différenciation des lymphocytes T , Glycoprotéines membranaires
4.
J Infect Dis ; 230(3): e518-e523, 2024 Sep 23.
Article de Anglais | MEDLINE | ID: mdl-38290045

RÉSUMÉ

The objective was to determine if antigen-specific tissue-resident memory T (TRM) cells persist in respiratory tissues of adults immunized as children with whole-cell pertussis (wP) or acellular pertussis (aP) vaccines. Mononuclear cells from tonsil or nasal tissue cells were cultured with Bordetella pertussis antigens and TRM cells quantified by flow cytometry. Adults immunized with wP vaccines as children had significantly more interleukin 17A (IL-17A) and interferon-γ (IFN-γ)-producing TRM cells that respond to B. pertussis antigens in respiratory tissues when compared with aP-primed donors. Our findings demonstrate that wP vaccines induce CD4 TRM cells that can persist in respiratory tissues for decades.


Sujet(s)
Bordetella pertussis , Lymphocytes T CD4+ , Interféron gamma , Interleukine-17 , Vaccin anticoquelucheux , Coqueluche , Adulte , Enfant , Femelle , Humains , Mâle , Jeune adulte , Bordetella pertussis/immunologie , Lymphocytes T CD4+/immunologie , Mémoire immunologique , Interféron gamma/métabolisme , Interféron gamma/immunologie , Interleukine-17/métabolisme , Interleukine-17/immunologie , Cellules T mémoire/immunologie , Tonsille palatine/immunologie , Vaccin anticoquelucheux/immunologie , Vaccin anticoquelucheux/administration et posologie , Coqueluche/immunologie , Coqueluche/prévention et contrôle
5.
mBio ; 15(1): e0257123, 2024 Jan 16.
Article de Anglais | MEDLINE | ID: mdl-38108639

RÉSUMÉ

IMPORTANCE: Staphylococcus aureus is one of the leading causes of antimicrobial-resistant infections whose success as a pathogen is facilitated by its massive array of immune evasion tactics, including intracellular survival within critical immune cells such as neutrophils, the immune system's first line of defense. In this study, we describe a novel pathway by which intracellular S. aureus can suppress the antimicrobial capabilities of human neutrophils by using the anti-inflammatory adenosine receptor, adora2a (A2aR). We show that signaling through A2aR suppresses the pentose phosphate pathway, a metabolic pathway used to fuel the antimicrobial NADPH oxidase complex that generates reactive oxygen species (ROS). As such, neutrophils show enhanced ROS production and reduced intracellular S. aureus when treated with an A2aR inhibitor. Taken together, we identify A2aR as a potential therapeutic target for combatting intracellular S. aureus infection.


Sujet(s)
Anti-infectieux , Infections à staphylocoques , Humains , Granulocytes neutrophiles , Staphylococcus aureus/métabolisme , Espèces réactives de l'oxygène/métabolisme , Voie des pentoses phosphates , Interactions hôte-pathogène , Anti-infectieux/métabolisme , Récepteurs purinergiques P1/métabolisme
6.
Redox Biol ; 67: 102905, 2023 11.
Article de Anglais | MEDLINE | ID: mdl-37820403

RÉSUMÉ

Inflammatory bowel diseases (IBD) are chronic intestinal disorders that result from an inappropriate inflammatory response to the microbiota in genetically susceptible individuals, often triggered by environmental stressors. Part of this response is the persistent inflammation and tissue injury associated with deficiency or excess of reactive oxygen species (ROS). The NADPH oxidase NOX1 is highly expressed in the intestinal epithelium, and inactivating NOX1 missense mutations are considered a risk factor for developing very early onset IBD. Albeit NOX1 has been linked to wound healing and host defence, many questions remain about its role in intestinal homeostasis and acute inflammatory conditions. Here, we used in vivo imaging in combination with inhibitor studies and germ-free conditions to conclusively identify NOX1 as essential superoxide generator for microbiota-dependent peroxynitrite production in homeostasis and during early endotoxemia. NOX1 loss-of-function variants cannot support peroxynitrite production, suggesting that the gut barrier is persistently weakened in these patients. One of the loss-of-function NOX1 variants, NOX1 p. Asn122His, features replacement of an asparagine residue located in a highly conserved HxxxHxxN motif. Modelling the NOX1-p22phox complex revealed near the distal heme an internal pocket restricted by His119 and Asn122 that is part of the oxygen reduction site. Functional studies in several human NADPH oxidases show that substitution of asparagine with amino acids with larger side chains is not tolerated, while smaller side chains can support catalytic activity. Thus, we identified a previously unrecognized structural feature required for the electron transfer mechanism in human NADPH oxidases.


Sujet(s)
Asparagine , Maladies inflammatoires intestinales , Humains , Acide peroxynitreux , NADPH oxidase/génétique , NADPH oxidase/métabolisme , Maladies inflammatoires intestinales/génétique , Espèces réactives de l'oxygène/métabolisme , NADPH Oxidase 1/génétique
8.
Elife ; 122023 07 04.
Article de Anglais | MEDLINE | ID: mdl-37401629

RÉSUMÉ

The bloodstream represents a hostile environment that bacteria must overcome to cause bacteraemia. To understand how the major human pathogen Staphylococcus aureus manages this we have utilised a functional genomics approach to identify a number of new loci that affect the ability of the bacteria to survive exposure to serum, the critical first step in the development of bacteraemia. The expression of one of these genes, tcaA, was found to be induced upon exposure to serum, and we show that it is involved in the elaboration of a critical virulence factor, the wall teichoic acids (WTA), within the cell envelope. The activity of the TcaA protein alters the sensitivity of the bacteria to cell wall attacking agents, including antimicrobial peptides, human defence fatty acids, and several antibiotics. This protein also affects the autolytic activity and lysostaphin sensitivity of the bacteria, suggesting that in addition to changing WTA abundance in the cell envelope, it also plays a role in peptidoglycan crosslinking. With TcaA rendering the bacteria more susceptible to serum killing, while simultaneously increasing the abundance of WTA in the cell envelope, it was unclear what effect this protein may have during infection. To explore this, we examined human data and performed murine experimental infections. Collectively, our data suggests that whilst mutations in tcaA are selected for during bacteraemia, this protein positively contributes to the virulence of S. aureus through its involvement in altering the cell wall architecture of the bacteria, a process that appears to play a key role in the development of bacteraemia.


Sujet(s)
Bactériémie , Infections à staphylocoques , Animaux , Humains , Souris , Infections à staphylocoques/microbiologie , Staphylococcus aureus/métabolisme , Paroi cellulaire/métabolisme , Antibactériens/pharmacologie , Acides teichoïques/métabolisme
9.
Nat Commun ; 14(1): 3513, 2023 06 14.
Article de Anglais | MEDLINE | ID: mdl-37316487

RÉSUMÉ

Excessive inflammation-associated coagulation is a feature of infectious diseases, occurring in such conditions as bacterial sepsis and COVID-19. It can lead to disseminated intravascular coagulation, one of the leading causes of mortality worldwide. Recently, type I interferon (IFN) signaling has been shown to be required for tissue factor (TF; gene name F3) release from macrophages, a critical initiator of coagulation, providing an important mechanistic link between innate immunity and coagulation. The mechanism of release involves type I IFN-induced caspase-11 which promotes macrophage pyroptosis. Here we find that F3 is a type I IFN-stimulated gene. Furthermore, F3 induction by lipopolysaccharide (LPS) is inhibited by the anti-inflammatory agents dimethyl fumarate (DMF) and 4-octyl itaconate (4-OI). Mechanistically, inhibition of F3 by DMF and 4-OI involves suppression of Ifnb1 expression. Additionally, they block type I IFN- and caspase-11-mediated macrophage pyroptosis, and subsequent TF release. Thereby, DMF and 4-OI inhibit TF-dependent thrombin generation. In vivo, DMF and 4-OI suppress TF-dependent thrombin generation, pulmonary thromboinflammation, and lethality induced by LPS, E. coli, and S. aureus, with 4-OI additionally attenuating inflammation-associated coagulation in a model of SARS-CoV-2 infection. Our results identify the clinically approved drug DMF and the pre-clinical tool compound 4-OI as anticoagulants that inhibit TF-mediated coagulopathy via inhibition of the macrophage type I IFN-TF axis.


Sujet(s)
COVID-19 , Interféron de type I , Thrombose , Humains , Anticoagulants , Thromboplastine , Fumarate de diméthyle/pharmacologie , Fumarate de diméthyle/usage thérapeutique , Escherichia coli , Inflammation , Lipopolysaccharides , Staphylococcus aureus , Thrombine , SARS-CoV-2 , Macrophages , Caspases
10.
Mucosal Immunol ; 16(4): 399-407, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-37088262

RÉSUMÉ

The respiratory tract is home to a diverse microbial community whose influence on local and systemic immune responses is only beginning to be appreciated. The airways have been linked with the trafficking of myelin-specific T-cells in the preclinical stages of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Th17 cells are important pathogenic effectors in MS and EAE but are innocuous immediately following differentiation. Upregulation of the cytokine GM-CSF appears to be a critical step in their acquisition of pathogenic potential, but little is known about the mechanisms that mediate this process. Here, primed myelin-specific Th17 cells were transferred to congenic recipient mice prior to exposure to various human respiratory tract-associated bacteria and T-cell trafficking, phenotype and the severity of resulting EAE were monitored. Disease was exacerbated in mice exposed to the Proteobacteria Moraxella catarrhalis and Klebsiella pneumoniae, but not the Firmicute Veillonella parvula, and this was associated with significantly increased GM-CSF+ and GM-CSF+IFNγ+ ex-Th17-like donor CD4 T cells in the lungs and central nervous system (CNS) of these mice. These findings support the concept that respiratory bacteria may contribute to the pathophysiology of CNS autoimmunity by modulating pathogenicity in crucial T-cell subsets that orchestrate neuroinflammation.


Sujet(s)
Encéphalomyélite auto-immune expérimentale , Sclérose en plaques , Souris , Humains , Animaux , Facteur de stimulation des colonies de granulocytes et de macrophages , Moraxella catarrhalis , Klebsiella pneumoniae , Gaine de myéline/anatomopathologie , Cellules Th17 , Virulence , Encéphalomyélite auto-immune expérimentale/anatomopathologie , Sclérose en plaques/anatomopathologie , Appareil respiratoire , Souris de lignée C57BL , Lymphocytes auxiliaires Th1
11.
Cell Rep ; 42(4): 112341, 2023 04 25.
Article de Anglais | MEDLINE | ID: mdl-37018072

RÉSUMÉ

PYHIN proteins AIM2 and IFI204 sense pathogen DNA, while other PYHINs have been shown to regulate host gene expression through as-yet unclear mechanisms. We characterize mouse PYHIN IFI207, which we find is not involved in DNA sensing but rather is required for cytokine promoter induction in macrophages. IFI207 co-localizes with both active RNA polymerase II (RNA Pol II) and IRF7 in the nucleus and enhances IRF7-dependent gene promoter induction. Generation of Ifi207-/- mice shows no role for IFI207 in autoimmunity. Rather, IFI207 is required for the establishment of a Klebsiella pneumoniae lung infection and for Klebsiella macrophage phagocytosis. These insights into IFI207 function illustrate that PYHINs can have distinct roles in innate immunity independent of DNA sensing and highlight the need to better characterize the whole mouse locus, one gene at a time.


Sujet(s)
Cytokines , Klebsiella pneumoniae , Souris , Animaux , Klebsiella pneumoniae/génétique , Protéines nucléaires/métabolisme , Immunité innée , ADN
12.
bioRxiv ; 2023 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-36865143

RÉSUMÉ

The bloodstream represents a hostile environment that bacteria must overcome to cause bacteraemia. To understand how the major human pathogen Staphylococcus aureus manages this we have utilised a functional genomics approach to identify a number of new loci that affect the ability of the bacteria to survive exposure to serum, the critical first step in the development of bacteraemia. The expression of one of these genes, tcaA, was found to be induced upon exposure to serum, and we show that it is involved in the elaboration of a critical virulence factor, the wall teichoic acids (WTA), within the cell envelope. The activity of the TcaA protein alters the sensitivity of the bacteria to cell wall attacking agents, including antimicrobial peptides, human defence fatty acids, and several antibiotics. This protein also affects the autolytic activity and lysostaphin sensitivity of the bacteria, suggesting that in addition to changing WTA abundance in the cell envelope, it also plays a role in peptidoglycan crosslinking. With TcaA rendering the bacteria more susceptible to serum killing, while simultaneously increasing the abundance of WTA in the cell envelope, it was unclear what effect this protein may have during infection. To explore this, we examined human data and performed murine experimental infections. Collectively, our data suggests that whilst mutations in tcaA are selected for during bacteraemia, this protein positively contributes to the virulence of S. aureus through its involvement in altering the cell wall architecture of the bacteria, a process that appears to play a key role in the development of bacteraemia.

13.
Eur J Immunol ; 53(5): e2250247, 2023 05.
Article de Anglais | MEDLINE | ID: mdl-36681765

RÉSUMÉ

Tissue-resident memory CD4 T (TRM ) cells induced by infection with Bordetella pertussis persist in respiratory tissues and confer long-term protective immunity against reinfection. However, it is not clear how they are maintained in respiratory tissues. Here, we demonstrate that B. pertussis-specific CD4 TRM cells produce IL-17A in response to in vitro stimulation with LPS or heat-killed Klebsiella pneumoniae (HKKP) in the presence of dendritic cells. Furthermore, IL-17A-secreting CD4 TRM cells expand in the lung and nasal tissue of B. pertussis convalescent mice following in vivo administration of LPS or HKKP. Bystander activation of CD4 TRM cells was suppressed by anti-IL-12p40 but not by anti-MHCII antibodies. Furthermore, purified respiratory tissue-resident, but not circulating, CD4 T cells from convalescent mice produced IL-17A following direct stimulation with IL-23 and IL-1ß or IL-18. Intranasal immunization of mice with a whole-cell pertussis vaccine induced respiratory CD4 TRM cells that were reactivated following stimulation with K. pneumoniae. Furthermore, the nasal pertussis vaccine conferred protective immunity against B. pertussis but also attenuated infection with K. pneumoniae. Our findings demonstrate that CD4 TRM cells induced by respiratory infection or vaccination can undergo bystander activation and confer heterologous immunity to an unrelated respiratory pathogen.


Sujet(s)
Bordetella pertussis , Coqueluche , Animaux , Souris , Bordetella pertussis/physiologie , Coqueluche/prévention et contrôle , Lymphocytes T CD4+ , Interleukine-17 , Klebsiella pneumoniae , Immunité hétérologue , Lipopolysaccharides , Mémoire immunologique , Vaccin anticoquelucheux
14.
Clin Rev Allergy Immunol ; 64(2): 161-178, 2023 Apr.
Article de Anglais | MEDLINE | ID: mdl-35275333

RÉSUMÉ

The respiratory tract is home to a diverse microbial community whose influence on local and systemic immune responses is only beginning to be appreciated. Increasing reports have linked changes in this microbiome to a range of pulmonary and extrapulmonary disorders, including asthma, chronic obstructive pulmonary disease and rheumatoid arthritis. Central to many of these findings is the role of IL-17-type immunity as an important driver of inflammation. Despite the crucial role played by IL-17-mediated immune responses in protection against infection, overt Th17 cell responses have been implicated in the pathogenesis of several chronic inflammatory diseases. However, our knowledge of the influence of bacteria that commonly colonise the respiratory tract on IL-17-driven inflammatory responses remains sparse. In this article, we review the current knowledge on the role of specific members of the airway microbiota in the modulation of IL-17-type immunity and discuss how this line of research may support the testing of susceptible individuals and targeting of inflammation at its earliest stages in the hope of preventing the development of chronic disease.


Sujet(s)
Inflammation , Interleukine-17 , Poumon , Microbiote , Humains , Maladie chronique , Inflammation/immunologie , Interleukine-17/immunologie , Poumon/immunologie , Poumon/microbiologie , Microbiote/immunologie , Appareil respiratoire/immunologie , Appareil respiratoire/microbiologie
15.
PLoS Pathog ; 18(7): e1010647, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-35776778

RÉSUMÉ

Staphylococcus aureus persistently colonises the anterior nares of a significant proportion of the healthy population, however the local immune response elicited during S. aureus nasal colonisation remains ill-defined. Local activation of IL-17/IL-22 producing T cells are critical for controlling bacterial clearance from the nasal cavity. However, recurrent and long-term colonisation is commonplace indicating efficient clearance does not invariably occur. Here we identify a central role for the regulatory cytokine IL-10 in facilitating bacterial persistence during S. aureus nasal colonisation in a murine model. IL-10 is produced rapidly within the nasal cavity following S. aureus colonisation, primarily by myeloid cells. Colonised IL-10-/- mice demonstrate enhanced IL-17+ and IL-22+ T cell responses and more rapidly clear bacteria from the nasal tissues as compared with wild-type mice. S. aureus also induces the regulatory cytokine IL-27 within the nasal tissue, which acts upstream of IL-10 promoting its production. IL-27 blockade reduces IL-10 production within the nasal cavity and improves bacterial clearance. TLR2 signalling was confirmed to be central to controlling the IL-10 response. Our findings conclude that during nasal colonisation S. aureus creates an immunosuppressive microenvironment through the local induction of IL-27 and IL-10, to dampen protective T cell responses and facilitate its persistence.


Sujet(s)
Interleukine-27 , Infections à staphylocoques , Animaux , Cytokines , Immunosuppression thérapeutique , Interleukine-10 , Interleukine-17 , Souris , Infections à staphylocoques/microbiologie , Staphylococcus aureus
16.
Heliyon ; 8(4): e09230, 2022 Apr.
Article de Anglais | MEDLINE | ID: mdl-35386227

RÉSUMÉ

SARS-CoV-2 infection causes a wide spectrum of disease severity. Identifying the immunological characteristics of severe disease and the risk factors for their development are important in the management of COVID-19. This study aimed to identify and rank clinical and immunological features associated with progression to severe COVID-19 in order to investigate an immunological signature of severe disease. One hundred and eight patients with positive SARS-CoV-2 PCR were recruited. Routine clinical and laboratory markers were measured, as well as myeloid and lymphoid whole-blood immunophenotyping and measurement of the pro-inflammatory cytokines IL-6 and soluble CD25. All analysis was carried out in a routine hospital diagnostic laboratory. Univariate analysis demonstrated that severe disease was most strongly associated with elevated CRP and IL-6, loss of DLA-DR expression on monocytes and CD10 expression on neutrophils. Unbiased machine learning demonstrated that these four features were strongly associated with severe disease, with an average prediction score for severe disease of 0.925. These results demonstrate that these four markers could be used to identify patients developing severe COVID-19 and allow timely delivery of therapeutics.

17.
Microb Genom ; 8(4)2022 04.
Article de Anglais | MEDLINE | ID: mdl-35416147

RÉSUMÉ

Streptococcus pneumoniae is a major human pathogen that can cause severe invasive diseases such as pneumonia, septicaemia and meningitis. Young children are at a particularly high risk, with an estimated 3-4 million cases of severe disease and between 300 000 and 500 000 deaths attributable to pneumococcal disease each year. The haemolytic toxin pneumolysin (Ply) is a primary virulence factor for this bacterium, yet despite its key role in pathogenesis, immune evasion and transmission, the regulation of Ply production is not well defined. Using a genome-wide association approach, we identified a large number of potential affectors of Ply activity, including a gene acquired horizontally on the antibiotic resistance-conferring Integrative and Conjugative Element (ICE) ICESp23FST81. This gene encodes a novel modular protein, ZomB, which has an N-terminal UvrD-like helicase domain followed by two Cas4-like domains with potent ATP-dependent nuclease activity. We found the regulatory effect of ZomB to be specific for the ply operon, potentially mediated by its high affinity for the BOX repeats encoded therein. Using a murine model of pneumococcal colonization, we further demonstrate that a ZomB mutant strain colonizes both the upper respiratory tract and lungs at higher levels when compared to the wild-type strain. While the antibiotic resistance-conferring aspects of ICESp23FST81 are often credited with contributing to the success of the S. pneumoniae lineages that acquire it, its ability to control the expression of a major virulence factor implicated in bacterial transmission is also likely to have played an important role.


Sujet(s)
Étude d'association pangénomique , Streptococcus pneumoniae , Animaux , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Séquences répétées dispersées/génétique , Souris , Streptococcus pneumoniae/génétique , Streptolysines , Facteurs de virulence/génétique , Facteurs de virulence/métabolisme
18.
Microorganisms ; 10(1)2022 Jan 12.
Article de Anglais | MEDLINE | ID: mdl-35056597

RÉSUMÉ

Mucosal-Associated Invariant T (MAIT) cells have been shown to play protective roles during infection with diverse pathogens through their propensity for rapid innate-like cytokine production and cytotoxicity. Among the potential applications for MAIT cells is to defend against Staphylococcus aureus, a pathogen of serious clinical significance. However, it is unknown how MAIT cell responses to S. aureus are elicited, nor has it been investigated whether MAIT cell cytotoxicity is mobilized against intracellular S. aureus. In this study, we investigate the capacity of human MAIT cells to respond directly to S. aureus. MAIT cells co-cultured with dendritic cells (DCs) infected with S. aureus rapidly upregulate CD69, express IFNγ and Granzyme B and degranulate. DC secretion of IL-12, but not IL-18, was implicated in this immune response, while TCR binding of MR1 is required to commence cytokine production. MAIT cell cytotoxicity resulted in apoptosis of S. aureus-infected cells, and reduced intracellular persistence of S. aureus. These findings implicate these unconventional T cells in important, rapid anti-S. aureus responses that may be of great relevance to the ongoing development of novel anti-S. aureus treatments.

19.
Front Immunol ; 12: 667387, 2021.
Article de Anglais | MEDLINE | ID: mdl-34220813

RÉSUMÉ

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.


Sujet(s)
Autophagie , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/microbiologie , Staphylococcus aureus/immunologie , Humains , Échappement immunitaire , Immunité innée
20.
Front Immunol ; 12: 705360, 2021.
Article de Anglais | MEDLINE | ID: mdl-34305945

RÉSUMÉ

Staphylococcus aureus is one of the most important human pathogens worldwide. Its high antibiotic resistance profile reinforces the need for new interventions like vaccines in addition to new antibiotics. Vaccine development efforts against S. aureus have failed so far however, the findings from these human clinical and non-clinical studies provide potential insight for such failures. Currently, research is focusing on identifying novel vaccine formulations able to elicit potent humoral and cellular immune responses. Translational science studies are attempting to discover correlates of protection using animal models as well as in vitro and ex vivo models assessing efficacy of vaccine candidates. Several new vaccine candidates are being tested in human clinical trials in a variety of target populations. In addition to vaccines, bacteriophages, monoclonal antibodies, centyrins and new classes of antibiotics are being developed. Some of these have been tested in humans with encouraging results. The complexity of the diseases and the range of the target populations affected by this pathogen will require a multipronged approach using different interventions, which will be discussed in this review.


Sujet(s)
Infections à staphylocoques/prévention et contrôle , Vaccins antistaphylococciques , Staphylococcus aureus/immunologie , Développement de vaccin , Adjuvants immunologiques , Animaux , Antigènes bactériens/immunologie , Essais cliniques comme sujet , Vésicules extracellulaires/immunologie , Glycoconjugués/immunologie , Bactéries à Gram négatif/immunologie , Interactions hôte-pathogène , Humains , Immunité cellulaire , Immunité humorale , Immunogénicité des vaccins , Techniques in vitro , Souris , Modèles animaux , Vaccins à base d'acide nucléique/immunologie , Périplasme/immunologie , Protéines recombinantes/immunologie , Vaccins antistaphylococciques/immunologie , Vaccins antistaphylococciques/usage thérapeutique , Science biomédicale translationnelle , Vaccins atténués/immunologie , Vaccins synthétiques/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE