Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.517
Filtrar
1.
Vet Ital ; 60(1)2024 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-38989625

RESUMEN

Staphylococcus aureus, Escherichia coli and Mycoplasma bovis are the most commonly isolated mastitis pathogens. The aim of this study was to evaluate the efficacy of a new mixed vaccine against mastitis caused by  Staphylococcus aureus, Escherichia coli, and Mycoplasma bovis. For this purpose, a mixed inactivated vaccine was administered subcutaneously to 24 heifers as one dose (2 mL) on the 45th day before birth and the second dose 21 days later. In 9 heifers, 2 mL of PBS was administered as placebo instead of vaccine. Then, heifers were divided into 3 groups as 7 vaccinated and 3 unvaccinated animals. Staphylococcus aureus, Escherichia coli, and Mycoplasma bovis were administered to the groups through intramammary route. Three vaccinated heifers were considered the common control without bacteria in all groups. The parameters considered to assess the effect of vaccination were clinical findings, bacterial count in milk, somatic cell count, and antibody titers. Clinical signs were observed only in the unvaccinated placebo group. Bacteria count and somatic cell count in milk increased in vaccinated and unvaccinated heifers. However, this increase was less in vaccinated animals and gradually returned to the normal level. In the unvaccinated heifers, it was ever high. Serum antibody titers were measured before and after vaccination. Antibody titers were high in vaccinated heifers after vaccination and were negative in unvaccinated heifers. In conclusion, the mixed vaccine had beneficial effect against Staphylococcus aureus, Escherichia coli, and Mycoplasma bovis mastitis and stimulated the immune response of vaccinated heifers.


Asunto(s)
Escherichia coli , Mastitis Bovina , Infecciones por Mycoplasma , Mycoplasma bovis , Infecciones Estafilocócicas , Staphylococcus aureus , Vacunas de Productos Inactivados , Animales , Bovinos , Mycoplasma bovis/inmunología , Femenino , Mastitis Bovina/prevención & control , Mastitis Bovina/microbiología , Mastitis Bovina/inmunología , Staphylococcus aureus/inmunología , Infecciones por Mycoplasma/veterinaria , Infecciones por Mycoplasma/prevención & control , Vacunas de Productos Inactivados/inmunología , Infecciones Estafilocócicas/prevención & control , Infecciones Estafilocócicas/veterinaria , Vacunas Bacterianas/inmunología , Vacunas Bacterianas/administración & dosificación , Infecciones por Escherichia coli/prevención & control , Infecciones por Escherichia coli/veterinaria , Infecciones por Escherichia coli/inmunología
2.
Int J Mol Sci ; 25(13)2024 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-39000557

RESUMEN

The effects of intestinal microflora on extraintestinal immune response by intestinal cytokines and metabolites have been documented, but whether intestinal microbes stimulate serum antibody generation is unknown. Here, serum antibodies against 69 outer membrane proteins of Escherichia coli, a dominant bacterium in the human intestine, are detected in 141 healthy individuals of varying ages. Antibodies against E. coli outer membrane proteins are determined in all serum samples tested, and frequencies of antibodies to five outer membrane proteins (OmpA, OmpX, TsX, HlpA, and FepA) are close to 100%. Serum antibodies against E. coli outer membrane proteins are further validated by Western blot and bacterial pull-down. Moreover, the present study shows that OstA, HlpA, Tsx, NlpB, OmpC, YfcU, and OmpA provide specific immune protection against pathogenic E. coli, while HlpA and OmpA also exhibit cross-protection against Staphylococcus aureus infection. These finding indicate that intestinal E. coli activate extraintestinal antibody responses and provide anti-infective immunity.


Asunto(s)
Anticuerpos Antibacterianos , Proteínas de la Membrana Bacteriana Externa , Escherichia coli , Humanos , Escherichia coli/inmunología , Anticuerpos Antibacterianos/inmunología , Anticuerpos Antibacterianos/sangre , Proteínas de la Membrana Bacteriana Externa/inmunología , Adulto , Femenino , Staphylococcus aureus/inmunología , Masculino , Formación de Anticuerpos/inmunología , Persona de Mediana Edad , Proteínas de Escherichia coli/inmunología , Adulto Joven , Anciano , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Adolescente , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología
3.
Cell Mol Life Sci ; 81(1): 300, 2024 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-39001897

RESUMEN

BACKGROUND: Age-associated impairments in innate immunity are believed to be a causative factor responsible for severe pathogenesis of Staphylococcus aureus (S. aureus) infection in the bone tissue. However, the basis for age-associated decline in innate immune response upon S. aureus infection remains poorly understood. RESULTS: Our transcriptional data (GEO: GSE166522) from a mouse model of S. aureus osteomyelitis show up-regulated CXCL9 and CXCL10 (CXCL9/10), which is further confirmed in vitro and in vivo by the present study. Notably, monocytes are a main source for CXCL9/10 production in bone marrow upon S. aureus challenge, but this response declines in middle-aged mice. Interestingly, conditional medium of bone marrow monocytes from middle-aged mice has a strikingly decreased effect on bactericidal functions of neutrophils and macrophages compares with that from young mice. We further show that activation of CXCL9/10-CXCR3 axis between monocytes and macrophages/neutrophils promotes the bactericidal function of the cells, whereas blocking the axis impairs such function. Importantly, treatment with either exogenous CXCL9 or CXCL10 in a middle-aged mice model enhances, while pharmacological inhibition of CXCR3 in young mice model impairs, bacterial clearance and bone marrow structure. CONCLUSIONS: These findings demonstrate that bone marrow monocytes act as a critical promotor of innate immune response via the CXLCL9/10-CXCR3 axis upon S. aureus infection, and that the increased susceptibility to S. aureus infection in skeleton in an aged host may be largely attributable to the declined induction of CXCR9/10 in monocytes.


Asunto(s)
Quimiocina CXCL10 , Quimiocina CXCL9 , Modelos Animales de Enfermedad , Inmunidad Innata , Monocitos , Osteomielitis , Infecciones Estafilocócicas , Staphylococcus aureus , Animales , Osteomielitis/microbiología , Osteomielitis/inmunología , Osteomielitis/metabolismo , Osteomielitis/patología , Monocitos/inmunología , Monocitos/metabolismo , Quimiocina CXCL9/metabolismo , Quimiocina CXCL9/genética , Staphylococcus aureus/inmunología , Ratones , Quimiocina CXCL10/metabolismo , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología , Infecciones Estafilocócicas/metabolismo , Ratones Endogámicos C57BL , Receptores CXCR3/metabolismo , Receptores CXCR3/genética , Envejecimiento/inmunología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo
4.
Adv Exp Med Biol ; 1445: 151-156, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38967757

RESUMEN

Skin is the most prominent tissue and organ, as well as the first line of defence, of the body. Because it is situated on the body's surface, it is constantly exposed to microbial, chemical, and physical factors such as mechanical stimulation. Therefore, skin has evolved substantial immune defences, regenerative ability, and anti-injury capacity. Epidermal cells produce antibacterial peptides that play a role in immune defence under physiological conditions. Additionally, IgG or IgA in the skin also participates in local anti-infective immunity. However, based on the classical theory of immunology, Ig can only be produced by B cells which should be derived from local B cells. This year, thanks to the discovery of Ig derived from non B cells (non B-Ig), Ig has also been found to be expressed in epidermal cells and contributes to immune defence. Epidermal cell-derived IgG and IgA have been demonstrated to have potential antibody activity by binding to pathogens. However, these epidermal cell-derived Igs show different microbial binding characteristics. For instance, IgG binds to Staphylococcus aureus and IgA binds to Staphylococcus epidermidis. Epidermal cells producing IgG and IgA may serve as an effective defense mechanism alongside B cells, providing a novel insight into skin immunity.


Asunto(s)
Inmunoglobulina A , Piel , Humanos , Inmunoglobulina A/inmunología , Inmunoglobulina A/metabolismo , Piel/inmunología , Animales , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Linfocitos B/inmunología , Inmunoglobulinas/inmunología , Inmunoglobulinas/metabolismo , Staphylococcus aureus/inmunología , Staphylococcus epidermidis/inmunología , Epidermis/inmunología , Epidermis/metabolismo , Células Epidérmicas/inmunología , Células Epidérmicas/metabolismo
5.
Sci Rep ; 14(1): 15472, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38969796

RESUMEN

This study evaluated the determinants of mortality and the T cell immune response in patients with persistent Staphylococcus aureus bacteremia (SAB). This was a prospective cohort study and patients with confirmed SAB were enrolled from 2008 to 2020. We compared clinical, microbiological, and genotypic features between surviving and deceased patients with persistent SAB. The concentrations of cytokines and the proportions of IFN-γ secreting CD4+ T cells were measured serially during the bacteremia period. Of the 1760 patients, 242 had persistent bacteremia (PB), and 49 PB patients died within 30 days. In the multivariate analysis, the APACHE II score and female sex were independently associated with 30 days mortality. The level of IL-10 was significantly increased in the plasma of patients with a high Pitt bacteremia score and those who died within 12 weeks from the index day. The proportion of IFN-γ-secreting CD4+ T cells were the highest just before the positive-to-negative conversion of blood cultures in patients with a low Pitt bacteremia score and those who survived for 12 weeks. The level of IL-10 is correlated with clinical outcomes in PB patients. IFN-γ secreting CD4+ T cells might play a pivotal role in SAB PB.


Asunto(s)
Bacteriemia , Linfocitos T CD4-Positivos , Infecciones Estafilocócicas , Staphylococcus aureus , Humanos , Masculino , Femenino , Bacteriemia/mortalidad , Bacteriemia/microbiología , Bacteriemia/inmunología , Linfocitos T CD4-Positivos/inmunología , Infecciones Estafilocócicas/mortalidad , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/inmunología , Persona de Mediana Edad , Factores de Riesgo , Anciano , Estudios Prospectivos , Interferón gamma/sangre , Interferón gamma/metabolismo , Interleucina-10/sangre , Adulto , Citocinas/sangre , Citocinas/metabolismo
6.
JCI Insight ; 9(13)2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38973612

RESUMEN

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.


Asunto(s)
Interleucina-10 , Infecciones Estafilocócicas , Vacunas Estafilocócicas , Staphylococcus aureus , Interleucina-10/metabolismo , Interleucina-10/inmunología , Animales , Infecciones Estafilocócicas/prevención & control , Infecciones Estafilocócicas/inmunología , Vacunas Estafilocócicas/inmunología , Ratones , Staphylococcus aureus/inmunología , Femenino , Ratones Endogámicos C57BL , Células TH1/inmunología , Inmunización/métodos , Humanos , Anticuerpos Neutralizantes/inmunología , Eficacia de las Vacunas , Vacunación/métodos
7.
Front Immunol ; 15: 1417220, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38868766

RESUMEN

Staphylococcus aureus bacteremia causes significant morbidity and mortality. Treatment of staphylococcal infections is hindered by widespread antibiotic resistance, and attempts to develop an S. aureus vaccine have failed. Improved S. aureus treatment and infection prevention options require a deeper understanding of the correlates of protective immunity. CD4+ T cells have been identified as key orchestrators in the defense against S. aureus, but uncertainties persist regarding the subset, polarity, and breadth of the memory CD4+ T-cell pool required for protection. Here, using a mouse model of systemic S. aureus infection, we discovered that the breadth of bacterium-specific memory CD4+ T-cell pool is a critical factor for protective immunity against invasive S. aureus infections. Seeding mice with a monoclonal bacterium-specific circulating memory CD4+ T-cell population failed to protect against systemic S. aureus infection; however, the introduction of a polyclonal and polyfunctional memory CD4+ T-cell pool significantly reduced the bacterial burden. Our findings support the development of a multi-epitope T-cell-based S. aureus vaccine, as a strategy to mitigate the severity of S. aureus bacteremia.


Asunto(s)
Bacteriemia , Linfocitos T CD4-Positivos , Infecciones Estafilocócicas , Staphylococcus aureus , Animales , Bacteriemia/inmunología , Bacteriemia/microbiología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/inmunología , Ratones , Linfocitos T CD4-Positivos/inmunología , Células T de Memoria/inmunología , Memoria Inmunológica , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Femenino , Vacunas Estafilocócicas/inmunología , Índice de Severidad de la Enfermedad
8.
Hum Vaccin Immunother ; 20(1): 2360338, 2024 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-38857905

RESUMEN

Staphylococcal Enterotoxin B (SEB), produced by Staphylococcus aureus (S. aureus), is a powerful superantigen that induces severe immune disruption and toxic shock syndrome (TSS) upon binding to MHC-II and TCR. Despite its significant impact on the pathogenesis of S. aureus, there are currently no specific therapeutic interventions available to counteract the mechanism of action exerted by this toxin. In this study, we have identified a human monoclonal antibody, named Hm0487, that specifically targets SEB by single-cell sequencing using PBMCs isolated from volunteers enrolled in a phase I clinical trial of the five-antigen S. aureus vaccine. X-ray crystallography studies revealed that Hm0487 exhibits high affinity for a linear B cell epitope in SEB (SEB138-147), which is located distantly from the site involved in the formation of the MHC-SEB-TCR ternary complex. Furthermore, in vitro studies demonstrated that Hm0487 significantly impacts the interaction of SEB with both receptors and the binding to immune cells, probably due to an allosteric effect on SEB rather than competing with receptors for binding sites. Moreover, both in vitro and in vivo studies validated that Hm0487 displayed efficient neutralizing efficacy in models of lethal shock and sepsis induced by either SEB or bacterial challenge. Our findings unveil an alternative mechanism for neutralizing the pathogenesis of SEB by Hm0487, and this antibody provides a novel strategy for mitigating both SEB-induced toxicity and S. aureus infection.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Neutralizantes , Enterotoxinas , Enterotoxinas/inmunología , Enterotoxinas/antagonistas & inhibidores , Humanos , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Animales , Cristalografía por Rayos X , Staphylococcus aureus/inmunología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/prevención & control , Epítopos de Linfocito B/inmunología , Ratones , Choque Séptico/inmunología , Choque Séptico/prevención & control , Femenino , Leucocitos Mononucleares/inmunología , Vacunas Estafilocócicas/inmunología , Anticuerpos Antibacterianos/inmunología , Superantígenos/inmunología
9.
Front Immunol ; 15: 1379042, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38903508

RESUMEN

Human milk oligosaccharides (HMOs) are present in high numbers in milk of lactating women. They are beneficial to gut health and the habitant microbiota, but less is known about their effect on cells from the immune system. In this study, we investigated the direct effect of three structurally different HMOs on human derived macrophages before challenge with Staphylococcus aureus (S. aureus). The study demonstrates that individual HMO structures potently affect the activation, differentiation and development of monocyte-derived macrophages in response to S. aureus. 6´-Sialyllactose (6'SL) had the most pronounced effect on the immune response against S. aureus, as illustrated by altered expression of macrophage surface markers, pointing towards an activated M1-like macrophage-phenotype. Similarly, 6'SL increased production of the pro-inflammatory cytokines TNF-α, IL-6, IL-8, IFN-γ and IL-1ß, when exposing cells to 6'SL in combination with S. aureus compared with S. aureus alone. Interestingly, macrophages treated with 6'SL exhibited an altered proliferation profile and increased the production of the classic M1 transcription factor NF-κB. The HMOs also enhanced macrophage phagocytosis and uptake of S. aureus. Importantly, the different HMOs did not notably affect macrophage activation and differentiation without S. aureus exposure. Together, these findings show that HMOs can potently augment the immune response against S. aureus, without causing inflammatory activation in the absence of S. aureus, suggesting that HMOs assist the immune system in targeting important pathogens during early infancy.


Asunto(s)
Citocinas , Activación de Macrófagos , Macrófagos , Leche Humana , Oligosacáridos , Fagocitosis , Staphylococcus aureus , Humanos , Leche Humana/inmunología , Staphylococcus aureus/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Oligosacáridos/farmacología , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/inmunología , Citocinas/metabolismo , Fagocitosis/efectos de los fármacos , Femenino , Diferenciación Celular/efectos de los fármacos , Infecciones Estafilocócicas/inmunología , Células Cultivadas
10.
PLoS Pathog ; 20(6): e1012306, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38843309

RESUMEN

Staphylococcus aureus is a human-adapted pathogen that replicates by asymptomatically colonizing its host. S. aureus is also the causative agent of purulent skin and soft tissue infections as well as bloodstream infections that result in the metastatic seeding of abscess lesions in all organ tissues. Prolonged colonization, infection, disease relapse, and recurrence point to the versatile capacity of S. aureus to bypass innate and adaptive immune defenses as well as the notion that some hosts fail to generate protective immune responses. Here, we find a genetic trait that provides protection against this pathogen. Mice lacking functional H2-O, the equivalent of human HLA-DO, inoculated with a mouse-adapted strain of S. aureus, efficiently decolonize the pathogen. Further, these decolonized animals resist subsequent bloodstream challenge with methicillin-resistant S. aureus. A genetic approach demonstrates that T-cell dependent B cell responses are required to control S. aureus colonization and infection in H2-O-deficient mice. Reduced bacterial burdens in these animals correlate with increased titers and enhanced phagocytic activity of S. aureus-specific antibodies. H2-O negatively regulates the loading of high affinity peptides on major histocompatibility class II (MHC-II) molecules. Thus, we hypothesize that immune responses against S. aureus are derepressed in mice lacking H2-O because more high affinity peptides are presented by MHC-II. We speculate that loss-of-function HLA-DO alleles may similarly control S. aureus replication in humans.


Asunto(s)
Infecciones Estafilocócicas , Staphylococcus aureus , Animales , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Ratones , Staphylococcus aureus/inmunología , Ratones Noqueados , Ratones Endogámicos C57BL , Antígenos de Histocompatibilidad Clase II/inmunología , Staphylococcus aureus Resistente a Meticilina/inmunología , Humanos
11.
PLoS One ; 19(5): e0298502, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38814922

RESUMEN

The skin microbiome maintains healthy human skin, and disruption of the microbiome balance leads to inflammatory skin diseases such as folliculitis and atopic dermatitis. Staphylococcus aureus and Cutibacterium acnes are pathogenic bacteria that simultaneously inhabit the skin and cause inflammatory diseases of the skin through the activation of innate immune responses. Silkworms are useful invertebrate animal models for evaluating innate immune responses. In silkworms, phenoloxidase generates melanin as an indicator of innate immune activation upon the recognition of bacterial or fungal components. We hypothesized that S. aureus and C. acnes interact to increase the innate immunity-activating properties of S. aureus. In the present study, we showed that acidification is involved in the activation of silkworm hemolymph melanization by S. aureus. Autoclaved-killed S. aureus (S. aureus [AC]) alone does not greatly activate silkworm hemolymph melanization. On the other hand, applying S. aureus [AC] treated with C. acnes culture supernatant increased the silkworm hemolymph melanization. Adding C. acnes culture supernatant to the medium decreased the pH. S. aureus [AC] treated with propionic acid, acetic acid, or lactic acid induced higher silkworm hemolymph melanization activity than untreated S. aureus [AC]. S. aureus [AC] treated with hydrochloric acid also induced silkworm hemolymph melanization. The silkworm hemolymph melanization activity of S. aureus [AC] treated with hydrochloric acid was inhibited by protease treatment of S. aureus [AC]. These results suggest that acid treatment of S. aureus induces innate immune activation in silkworms and that S. aureus proteins are involved in the induction of innate immunity in silkworms.


Asunto(s)
Bombyx , Hemolinfa , Melaninas , Staphylococcus aureus , Animales , Hemolinfa/metabolismo , Hemolinfa/microbiología , Hemolinfa/inmunología , Bombyx/microbiología , Bombyx/inmunología , Staphylococcus aureus/inmunología , Melaninas/metabolismo , Inmunidad Innata , Concentración de Iones de Hidrógeno , Monofenol Monooxigenasa/metabolismo
13.
J Allergy Clin Immunol ; 154(1): 31-41, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38761999

RESUMEN

Inflammatory skin diseases such as atopic eczema (atopic dermatitis [AD]) affect children and adults globally. In AD, the skin barrier is impaired on multiple levels. Underlying factors include genetic, chemical, immunologic, and microbial components. Increased skin pH in AD is part of the altered microbial microenvironment that promotes overgrowth of the skin microbiome with Staphylococcus aureus. The secretion of virulence factors, such as toxins and proteases, by S aureus further aggravates the skin barrier deficiency and additionally disrupts the balance of an already skewed immune response. Skin commensal bacteria, however, can inhibit the growth and pathogenicity of S aureus through quorum sensing. Therefore, restoring a healthy skin microbiome could contribute to remission induction in AD. This review discusses direct and indirect approaches to targeting the skin microbiome through modulation of the skin pH; UV treatment; and use of prebiotics, probiotics, and postbiotics. Furthermore, exploratory techniques such as skin microbiome transplantation, ozone therapy, and phage therapy are discussed. Finally, we summarize the latest findings on disease and microbiome modification through targeted immunomodulatory systemic treatments and biologics. We believe that targeting the skin microbiome should be considered a crucial component of successful AD treatment in the future.


Asunto(s)
Dermatitis Atópica , Microbiota , Piel , Dermatitis Atópica/inmunología , Dermatitis Atópica/microbiología , Dermatitis Atópica/terapia , Humanos , Microbiota/inmunología , Piel/microbiología , Piel/inmunología , Animales , Probióticos/uso terapéutico , Staphylococcus aureus/inmunología , Prebióticos/administración & dosificación
14.
Cytokine ; 180: 156634, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38810500

RESUMEN

BACKGROUND: Serine protease-like (Spl) proteins produced by Staphylococcus (S.) aureus have been associated with allergic inflammation. However, effects of Spls on the epidermal immune response have not been investigated. OBJECTIVES: To assess the epidermal immune response to SplA, SplD and SplE dependent on differentiation of keratinocytes and a Th2 or Th17 cytokine milieu. METHODS: Human keratinocytes of healthy controls and a STAT3-hyper-IgE syndrome (STAT3-HIES) patient were cultured in different calcium concentrations in the presence of Spls and Th2 or Th17 cytokines. Keratinocyte-specific IL-8 production and concomitant migration of neutrophils were assessed. RESULTS: SplE and more significantly SplA, induced IL-8 in keratinocytes. Suprabasal-like keratinocytes showed a higher Spl-mediated IL-8 production and neutrophil migration compared to basal-like keratinocytes. Th17 cytokines amplified Spl-mediated IL-8 production, which correlated with neutrophil recruitment. Neutrophil recruitment by keratinocytes of the STAT3-HIES patient was similar to healthy control cells. CONCLUSION: S. aureus-specific Spl proteases synergized with IL-17A on human keratinocytes with respect to IL-8 release and neutrophil migration, highlighting the importance of keratinocytes and Th17 immunity in barrier function.


Asunto(s)
Interleucina-17 , Interleucina-8 , Queratinocitos , Neutrófilos , Staphylococcus aureus , Humanos , Queratinocitos/metabolismo , Queratinocitos/inmunología , Queratinocitos/efectos de los fármacos , Interleucina-17/metabolismo , Interleucina-8/metabolismo , Staphylococcus aureus/inmunología , Neutrófilos/metabolismo , Neutrófilos/inmunología , Células Th17/inmunología , Células Th17/metabolismo , Proteínas Bacterianas/metabolismo , Factor de Transcripción STAT3/metabolismo , Movimiento Celular/efectos de los fármacos , Serina Proteasas/metabolismo , Células Cultivadas
15.
J Dermatol Sci ; 114(3): 104-114, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38806322

RESUMEN

BACKGROUND: Although several mouse models of exogenous-agent-induced atopic dermatitis (AD) are currently available, the lack of certainty regarding their similarity with human AD has limited their scientific value. Thus, comprehensive evaluation of the characteristics of mouse models and their similarity with human AD is essential. OBJECTIVE: To compare six different exogenous-agent-induced AD mouse models and find out the optimum models for study. METHODS: Female BALB/c mice underwent induction of AD-like dermatitis by MC903 alone or in combination with ovalbumin (OVA), dinitrofluorobenzene (DNFB) alone or in combination with OVA, OVA alone, or Staphylococcus aureus. Gross phenotype, total immunoglobulin E (IgE) level, histopathological manifestations, and skin lesion transcriptome were analyzed, and metagenomic sequencing of the gut microbiome was performed. RESULTS: The DNFB plus OVA model showed the highest disease severity, while the OVA model showed the lowest severity. The MC903 and MC903 plus OVA models showed high expression of T-helper (Th)2- and Th17-related genes; the DNFB and DNFB plus OVA models showed upregulation of Th1-, Th2-, and Th17-related genes; while the S. aureus inoculation model showed more enhanced Th1 and Th17 immune responses. In contrast to the other models, the OVA-induced model showed the lowest expression levels of inflammation-related genes, while the MC903 model shared the largest overlap with human AD profiles. The intestinal microbiota of all groups showed significant differences after modeling. CONCLUSION: Each AD mouse model exhibited different characteristics. The MC903 model was the best to recapitulate most features of human AD among these exogenous-agent-induced AD models.


Asunto(s)
Dermatitis Atópica , Dinitrofluorobenceno , Modelos Animales de Enfermedad , Ratones Endogámicos BALB C , Ovalbúmina , Fenotipo , Staphylococcus aureus , Transcriptoma , Animales , Dermatitis Atópica/inmunología , Dermatitis Atópica/microbiología , Dermatitis Atópica/inducido químicamente , Femenino , Ratones , Ovalbúmina/inmunología , Staphylococcus aureus/inmunología , Humanos , Piel/inmunología , Piel/patología , Piel/microbiología , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Microbioma Gastrointestinal/inmunología , Índice de Severidad de la Enfermedad , Perfilación de la Expresión Génica , Calcitriol/análogos & derivados
16.
Proc Natl Acad Sci U S A ; 121(22): e2402764121, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38771879

RESUMEN

Staphylococcus aureus (S. aureus) can evade antibiotics and host immune defenses by persisting within infected cells. Here, we demonstrate that in infected host cells, S. aureus type VII secretion system (T7SS) extracellular protein B (EsxB) interacts with the stimulator of interferon genes (STING) protein and suppresses the inflammatory defense mechanism of macrophages during early infection. The binding of EsxB with STING disrupts the K48-linked ubiquitination of EsxB at lysine 33, thereby preventing EsxB degradation. Furthermore, EsxB-STING binding appears to interrupt the interaction of 2 vital regulatory proteins with STING: aspartate-histidine-histidine-cysteine domain-containing protein 3 (DHHC3) and TNF receptor-associated factor 6. This persistent dual suppression of STING interactions deregulates intracellular proinflammatory pathways in macrophages, inhibiting STING's palmitoylation at cysteine 91 and its K63-linked ubiquitination at lysine 83. These findings uncover an immune-evasion mechanism by S. aureus T7SS during intracellular macrophage infection, which has implications for developing effective immunomodulators to combat S. aureus infections.


Asunto(s)
Proteínas Bacterianas , Macrófagos , Proteínas de la Membrana , Infecciones Estafilocócicas , Staphylococcus aureus , Sistemas de Secreción Tipo VII , Ubiquitinación , Staphylococcus aureus/inmunología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/inmunología , Humanos , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Animales , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/metabolismo , Sistemas de Secreción Tipo VII/metabolismo , Sistemas de Secreción Tipo VII/inmunología , Sistemas de Secreción Tipo VII/genética , Ratones , Evasión Inmune , Interacciones Huésped-Patógeno/inmunología
17.
mBio ; 15(6): e0086224, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38767353

RESUMEN

Mammalian target of rapamycin (mTOR) is a key regulator of metabolism in the mammalian cell. Here, we show the essential role for mTOR signaling in the immune response to bacterial infection. Inhibition of mTOR during infection with Staphylococcus aureus revealed that mTOR signaling is required for bactericidal free radical production by phagocytes. Mechanistically, mTOR supported glucose transporter GLUT1 expression, potentially through hypoxia-inducible factor 1α, upon phagocyte activation. Cytokine and chemokine signaling, inducible nitric oxide synthase, and p65 nuclear translocation were present at similar levels during mTOR suppression, suggesting an NF-κB-independent role for mTOR signaling in the immune response during bacterial infection. We propose that mTOR signaling primarily mediates the metabolic requirements necessary for phagocyte bactericidal free radical production. This study has important implications for the metabolic requirements of innate immune cells during bacterial infection as well as the clinical use of mTOR inhibitors.IMPORTANCESirolimus, everolimus, temsirolimus, and similar are a class of pharmaceutics commonly used in the clinical treatment of cancer and the anti-rejection of transplanted organs. Each of these agents suppresses the activity of the mammalian target of rapamycin (mTOR), a master regulator of metabolism in human cells. Activation of mTOR is also involved in the immune response to bacterial infection, and treatments that inhibit mTOR are associated with increased susceptibility to bacterial infections in the skin and soft tissue. Infections caused by Staphylococcus aureus are among the most common and severe. Our study shows that this susceptibility to S. aureus infection during mTOR suppression is due to an impaired function of phagocytic immune cells responsible for controlling bacterial infections. Specifically, we observed that mTOR activity is required for phagocytes to produce antimicrobial free radicals. These results have important implications for immune responses during clinical treatments and in disease states where mTOR is suppressed.


Asunto(s)
Transportador de Glucosa de Tipo 1 , Fagocitos , Transducción de Señal , Infecciones Estafilocócicas , Staphylococcus aureus , Serina-Treonina Quinasas TOR , Staphylococcus aureus/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Serina-Treonina Quinasas TOR/genética , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Fagocitos/inmunología , Fagocitos/metabolismo , Fagocitos/microbiología , Humanos , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 1/genética , Animales , Radicales Libres/metabolismo , Ratones , Ratones Endogámicos C57BL
18.
Vet Immunol Immunopathol ; 272: 110770, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38735115

RESUMEN

Interferon lambda (IFN-λ) is an important type III interferon triggered mainly by viral infection. IFN-λ binds to their heterodimeric receptors and signals through JAK-STAT pathways similar to type I IFN. In this study, we deduced the buffalo IFN-λ sequences through the polymerase chain reaction, and then studied IFN-λ's expression patterns in different tissues, and post induction with poly I:C and live MRSA using RT-qPCR. The full-length sequences of buffalo IFN-λ3, IFN-λ receptors, and a transcript variant of IFN-λ4 were determined. IFN-λ1 is identified as a pseudogene. Virus response elements and a recombination hotspot factor was observed in the regulatory region of IFN-λ. The IFN-λ3 expressed highest in lungs and monocytes but IFN-λ4 did not. The expression of Interferon Lambda Receptor 1 was tissue specific, while Interleukin 10 Receptor subunit beta was ubiquitous. Following poly I:C induction, IFN-λ3 expression was primarily observed in epithelial cells as opposed to fibroblasts, displaying cell type-dependent expression. The cytosolic RNA sensors were expressed highest in endometrial epithelial cells, whereas the endosomal receptor was higher in fibroblasts. 2',5'-oligoadenylate synthetase expressed higher in fibroblasts, myxoma resistance protein 1 and IFN-stimulated gene 56 in epithelial cells, displaying cell-specific antiviral response of the interferon stimulated genes (ISGs). The endometrial epithelial cells expressed IFN-λ3 after live S. aureus infection indicating its importance in bacterial infection. The induction of IFN-λ3 was S. aureus isolate specific at the same multiplicity of infection (MOI). This study elucidates the IFN-λ sequences, diverse expression patterns revealing tissue specificity, and specificity in response to poly I:C and bacterial stimuli, emphasising its crucial role in innate immune response modulation.


Asunto(s)
Búfalos , Interferones , Animales , Búfalos/inmunología , Búfalos/genética , Interferones/genética , Interferones/inmunología , Poli I-C/farmacología , Perfilación de la Expresión Génica/veterinaria , Filogenia , Interferón lambda , Secuencia de Aminoácidos , Receptores de Interferón/genética , Receptores de Interferón/inmunología , Femenino , 2',5'-Oligoadenilato Sintetasa/genética , 2',5'-Oligoadenilato Sintetasa/metabolismo , Staphylococcus aureus/inmunología
19.
JCI Insight ; 9(9)2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38716729

RESUMEN

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.


Asunto(s)
Dermatitis Atópica , Infecciones Cutáneas Estafilocócicas , Staphylococcus aureus , Dermatitis Atópica/inmunología , Dermatitis Atópica/microbiología , Humanos , Staphylococcus aureus/inmunología , Niño , Femenino , Infecciones Cutáneas Estafilocócicas/inmunología , Infecciones Cutáneas Estafilocócicas/microbiología , Masculino , Preescolar , Piel/microbiología , Piel/inmunología , Piel/patología , Quimiocina CXCL10/inmunología , Quimiocina CXCL10/metabolismo , Células TH1/inmunología , Células Th2/inmunología , Células Th17/inmunología , Teorema de Bayes , Linfocitos T CD8-positivos/inmunología , Interleucina-10/metabolismo , Interleucina-10/inmunología , Linfocitos Intraepiteliales/inmunología , Antígenos de Diferenciación de Linfocitos T , Glicoproteínas de Membrana
20.
Vaccine ; 42(15): 3445-3454, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38631956

RESUMEN

Major histocompatibility complex class II (MHC-II) molecules are involved in immune responses against pathogens and vaccine candidates' immunogenicity. Immunopeptidomics for identifying cancer and infection-related antigens and epitopes have benefited from advances in immunopurification methods and mass spectrometry analysis. The mouse anti-MHC-II-DR monoclonal antibody L243 (mAb-L243) has been effective in recognising MHC-II-DR in both human and non-human primates. It has also been shown to cross-react with other animal species, although it has not been tested in livestock. This study used mAb-L243 to identify Staphylococcus aureus and Salmonella enterica serovar Typhimurium peptides binding to cattle and swine macrophage MHC-II-DR molecules using flow cytometry, mass spectrometry and two immunopurification techniques. Antibody cross-reactivity led to identifying expressed MHC-II-DR molecules, together with 10 Staphylococcus aureus peptides in cattle and 13 S. enterica serovar Typhimurium peptides in swine. Such data demonstrates that MHC-II-DR expression and immunocapture approaches using L243 mAb represents a viable strategy for flow cytometry and immunopeptidomics analysis of bovine and swine antigen-presenting cells.


Asunto(s)
Anticuerpos Monoclonales , Macrófagos , Salmonella typhimurium , Staphylococcus aureus , Animales , Bovinos , Porcinos/inmunología , Staphylococcus aureus/inmunología , Anticuerpos Monoclonales/inmunología , Macrófagos/inmunología , Salmonella typhimurium/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Reacciones Cruzadas/inmunología , Citometría de Flujo , Espectrometría de Masas , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...