Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 184
Filtrar
1.
Am J Physiol Lung Cell Mol Physiol ; 324(4): L445-L455, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36749572

RESUMEN

Acute respiratory distress syndrome (ARDS) remains a significant cause of morbidity and mortality in critically ill patients. Oxidative stress and inflammation play a crucial role in the pathogenesis of ARDS. Extracellular superoxide dismutase (EC-SOD) is abundant in the lung and is an important enzymatic defense against superoxide. Human single-nucleotide polymorphism in matrix binding region of EC-SOD leads to the substitution of arginine to glycine at position 213 (R213G) and results in release of EC-SOD into alveolar fluid, without affecting enzyme activity. We hypothesized that R213G EC-SOD variant protects against lung injury and inflammation via the blockade of neutrophil recruitment in infectious model of methicillin-resistant S. aureus (MRSA) pneumonia. After inoculation with MRSA, wild-type (WT) mice had impaired integrity of alveolar-capillary barrier and increased levels of IL-1ß, IL-6, and TNF-α in the broncho-alveolar lavage fluid (BALF), while infected mice expressing R213G EC-SOD variant maintained the integrity of alveolar-capillary interface and had attenuated levels of proinflammatory cytokines. MRSA-infected mice expressing R213G EC-SOD variant also had attenuated neutrophil numbers in BALF and decreased expression of neutrophil chemoattractant CXCL1 by the alveolar epithelial ATII cells, compared with the infected WT group. The decreased neutrophil numbers in R213G mice were not due to increased rate of apoptosis. Mice expressing R213G variant had a differential effect on neutrophil functionality-the generation of neutrophil extracellular traps (NETs) but not myeloperoxidase (MPO) levels were attenuated in comparison with WT controls. Despite having the same bacterial load in the lung as WT controls, mice expressing R213G EC-SOD variant were protected from extrapulmonary dissemination of bacteria.


Asunto(s)
Lesión Pulmonar Aguda , Staphylococcus aureus Resistente a Meticilina , Neumonía Estafilocócica , Síndrome de Dificultad Respiratoria , Humanos , Ratones , Animales , Staphylococcus aureus/metabolismo , Neumonía Estafilocócica/metabolismo , Neumonía Estafilocócica/patología , Inflamación/patología , Pulmón/metabolismo , Lesión Pulmonar Aguda/patología , Síndrome de Dificultad Respiratoria/patología , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo
2.
J Infect Dis ; 226(7): 1286-1294, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-35899844

RESUMEN

Respiratory coinfection of influenza with Staphylococcus aureus often causes severe disease; methicillin-resistant S. aureus (MRSA) coinfection is frequently fatal. Understanding disease pathogenesis may inform therapies. We aimed to identify host and pathogen transcriptomic (messenger RNA) signatures from the respiratory compartment of pediatric patients critically ill with influenza-S. aureus coinfection (ISAC), signatures that predict worse outcomes. Messenger RNA extracted from endotracheal aspirate samples was evaluated for S. aureus and host transcriptomic biosignatures. Influenza-MRSA outcomes were worse, but of 190 S. aureus virulence-associated genes, 6 were differentially expressed between MRSA-coinfected versus methicillin-susceptible S. aureus-coinfected patients, and none discriminated outcome. Host gene expression in patients with ISAC was compared with that in patients with influenza infection alone. Patients with poor clinical outcomes (death or prolonged multiorgan dysfunction) had relatively reduced expression of interferons and down-regulation of interferon γ-induced immune cell chemoattractants CXCL10 and CXCL11. In ISAC, airway host but not pathogen gene expression profiles predicted worse clinical outcomes.


Asunto(s)
Coinfección , Gripe Humana , Staphylococcus aureus Resistente a Meticilina , Neumonía Estafilocócica , Infecciones Estafilocócicas , Factores Quimiotácticos , Niño , Coinfección/patología , Humanos , Gripe Humana/complicaciones , Gripe Humana/genética , Interferón gamma , Meticilina , Staphylococcus aureus Resistente a Meticilina/genética , Neumonía Estafilocócica/genética , Neumonía Estafilocócica/patología , ARN Mensajero , Infecciones Estafilocócicas/complicaciones , Infecciones Estafilocócicas/genética , Staphylococcus aureus/genética , Transcriptoma
3.
JCI Insight ; 7(4)2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-34990413

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal disease characterized by collagen deposition within the lung interstitium. Bacterial infection is associated with increased morbidity and more rapid mortality in IPF patient populations, and pathogens such as methicillin-resistant Staphylococcus aureus (MRSA) are commonly isolated from the lungs of hospitalized patients with IPF. Despite this, the effects of fibrotic lung injury on critical immune responses to infection remain unknown. In the present study, we show that, like humans with IPF, fibrotic mice infected with MRSA exhibit increased morbidity and mortality compared with uninfected fibrotic mice. We determine that fibrosis conferred a defect in MRSA clearance compared with nonfibrotic mice, resulting from blunted innate immune responses. We show that fibrosis inhibited neutrophil intracellular killing of MRSA through impaired neutrophil elastase release and oxidative radical production. Additionally, we demonstrate that lung macrophages from fibrotic mice have impaired phagocytosis of MRSA. Our study describes potentially novel impairments of antimicrobial responses upon pulmonary fibrosis development, and our findings suggest a possible mechanism for why patients with IPF are at greater risk of morbidity and mortality related to infection.


Asunto(s)
Fibrosis Pulmonar Idiopática/inmunología , Inmunidad Innata/inmunología , Macrófagos Alveolares/metabolismo , Staphylococcus aureus Resistente a Meticilina/aislamiento & purificación , Neutrófilos/patología , Neumonía Estafilocócica/patología , Animales , Modelos Animales de Enfermedad , Fibrosis Pulmonar Idiopática/complicaciones , Macrófagos Alveolares/patología , Ratones , Ratones Endogámicos C57BL , Neutrófilos/metabolismo , Fagocitosis , Neumonía Estafilocócica/etiología , Neumonía Estafilocócica/inmunología
4.
Int J Mol Sci ; 22(18)2021 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-34575918

RESUMEN

Dehydrocostus lactone (DHL), a natural sesquiterpene lactone isolated from the traditional Chinese herbs Saussurea lappa and Inula helenium L., has important anti-inflammatory properties used for treating colitis, fibrosis, and Gram-negative bacteria-induced acute lung injury (ALI). However, the effects of DHL on Gram-positive bacteria-induced macrophage activation and ALI remains unclear. In this study, we found that DHL inhibited the phosphorylation of p38 MAPK, the degradation of IκBα, and the activation and nuclear translocation of NF-κB p65, but enhanced the phosphorylation of AMP-activated protein kinase (AMPK) and the expression of Nrf2 and HO-1 in lipoteichoic acid (LTA)-stimulated RAW264.7 cells and primary bone-marrow-derived macrophages (BMDMs). Given the critical role of the p38 MAPK/NF-κB and AMPK/Nrf2 signaling pathways in the balance of M1/M2 macrophage polarization and inflammation, we speculated that DHL would also have an effect on macrophage polarization. Further studies verified that DHL promoted M2 macrophage polarization and reduced M1 polarization, then resulted in a decreased inflammatory response. An in vivo study also revealed that DHL exhibited anti-inflammatory effects and ameliorated methicillin-resistant Staphylococcus aureus (MRSA)-induced ALI. In addition, DHL treatment significantly inhibited the p38 MAPK/NF-κB pathway and activated AMPK/Nrf2 signaling, leading to accelerated switching of macrophages from M1 to M2 in the MRSA-induced murine ALI model. Collectively, these data demonstrated that DHL can promote macrophage polarization to an anti-inflammatory M2 phenotype via interfering in p38 MAPK/NF-κB signaling, as well as activating the AMPK/Nrf2 pathway in vitro and in vivo. Our results suggested that DHL might be a novel candidate for treating inflammatory diseases caused by Gram-positive bacteria.


Asunto(s)
Antiinflamatorios/farmacología , Lactonas/farmacología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Neumonía Estafilocócica/etiología , Sesquiterpenos/farmacología , Enfermedad Aguda , Animales , Plasticidad de la Célula/efectos de los fármacos , Plasticidad de la Célula/inmunología , Modelos Animales de Enfermedad , Activación de Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Modelos Biológicos , FN-kappa B/metabolismo , Fosforilación , Neumonía Estafilocócica/tratamiento farmacológico , Neumonía Estafilocócica/metabolismo , Neumonía Estafilocócica/patología , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos
5.
Int Immunopharmacol ; 90: 107221, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33293260

RESUMEN

Salvinorin A (SA), a neoclerodane diterpene, is isolated from the dried leaves ofSalvia divinorum. SA has traditionally been used treatments for chronic pain diseases. Recent research has demonstrated that SA possesses the anti-inflammatory property. The present study aim to explore the effects and potentialmechanisms ofSA in protection against Methicillin Resistant Staphylococcus aureus (MRSA)-induced acute lung injury (ALI). Here, we firstly found that verylowdosesof SA (50 µg/kg) could markedly decrease the infiltration of pulmonary neutrophils, mRNA expression of pro-inflammatory cytokines (TNF-α, IL-1ß and IL-6) and then attenuated ALI cause by MRSA infection in mice. In vitro findings revealed that SA attenuated lipoteichoicacid-induced apoptosis, inflammation and oxidative stress in RAW264.7 cells. Mechanism research revealed that SA increased both mRNA levels and protein levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and up-regulated mRNA expression of its downstream genes (HO-1, Gclm, Trx-1, SOD1 and SOD2). Additionally, Nrf2 knockout mice abolished the inhibitory effect of SA on neutrophil accumulation and oxidative stress in MRSA-induced ALI. In conclusion, SA attenuates MRSA-induced ALI via Nrf2 signaling pathways.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Antiinflamatorios/farmacología , Diterpenos de Tipo Clerodano/farmacología , Pulmón/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Factor 2 Relacionado con NF-E2/metabolismo , Neumonía Estafilocócica/prevención & control , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/microbiología , Lesión Pulmonar Aguda/patología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Pulmón/metabolismo , Pulmón/microbiología , Pulmón/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/microbiología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 2 Relacionado con NF-E2/genética , Infiltración Neutrófila/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Neumonía Estafilocócica/metabolismo , Neumonía Estafilocócica/microbiología , Neumonía Estafilocócica/patología , Células RAW 264.7 , Transducción de Señal
6.
J Infect Dis ; 222(8): 1400-1404, 2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32386322

RESUMEN

Staphylococcus aureus is a leading cause of pneumonia. We show here that the ClpXP protease involved in protein turnover is important for pathogenesis in a murine model of acute pneumonia. Staphylococcus aureus lacking this protease is attenuated in vivo, being rapidly cleared from the airway and leading to decreased immune cell influx and inflammation. Characterization of defined mutations in vitro identified defects in intracellular survival and protection against neutrophil killing. Our results further expand on what is known about ClpXP in the pathogenesis of S. aureus to include the respiratory tract.


Asunto(s)
Proteínas Bacterianas/metabolismo , Endopeptidasa Clp/metabolismo , Neumonía Estafilocócica/microbiología , Staphylococcus aureus/patogenicidad , Animales , Proteínas Bacterianas/genética , Modelos Animales de Enfermedad , Endopeptidasa Clp/genética , Femenino , Interacciones Huésped-Patógeno , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Viabilidad Microbiana , Mutación , Neutrófilos/inmunología , Neumonía Estafilocócica/inmunología , Neumonía Estafilocócica/patología , Staphylococcus aureus/enzimología , Staphylococcus aureus/genética
7.
J Cell Physiol ; 235(1): 563-572, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31318050

RESUMEN

Sepsis is a severe clinical disease, which is resulted from the excessive host inflammation response to the infection. Growing evidence indicates that Staphylococcus aureus pneumonia is a significant cause of sepsis, which can lead to intestinal injury, inflammation, and apoptosis. Studies have shown that miR-182-5p can serve as a tumor oncogene or a tumor suppressive microRNA in various cancers, however, its biological role in sepsis is still uninvestigated. Here, we reported that miR-182-5p was obviously increased in S. aureus pneumonia mice models. Loss of miR-182-5p inhibited intestinal damage and intestinal apoptosis as indicated by the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. In addition, we observed the lack of miR-182-5p altered the local inflammatory response to pneumonia in the intestine. Elevated tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) levels were observed in intestinal tissue of pneumonia groups compared with the shams. Furthermore, miR-182-5p knockout (KO) pneumonia group demonstrated decreased levels of intestinal TNF-α and IL-6. Primary murine intestinal epithelial cells were isolated and cultured in our investigation. We exhibited downregulation of miR-182-5p repressed intestinal epithelial cells apoptosis and rescued the cell viability. Meanwhile, miR-182-5p caused elevated cell apoptosis and reduced cell proliferation. Moreover, the surfactant protein D (SP-D) binds with the bacterial pathogens and remove the pathogens and apoptotic bodies, which exhibits important roles in modulating immune responses. It was displayed in our study that SP-D was greatly decreased in pneumonia mice models. SP-D was predicted as a downstream target of miR-182-5p. These data concluded that miR-182-5p promoted intestinal injury in S. aureus pneumonia-induced sepsis via targeting SP-D.


Asunto(s)
Mucosa Intestinal/patología , MicroARNs/genética , Neumonía Estafilocócica/patología , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Animales , Apoptosis/genética , Supervivencia Celular/genética , Células Cultivadas , Modelos Animales de Enfermedad , Células Epiteliales/patología , Técnicas de Inactivación de Genes , Inflamación/patología , Interleucina-6/metabolismo , Mucosa Intestinal/microbiología , Ratones , Ratones Endogámicos C57BL , Oncogenes/genética , Neumonía Estafilocócica/genética , Sepsis/patología , Transducción de Señal , Staphylococcus aureus/patogenicidad , Factor de Necrosis Tumoral alfa/metabolismo
8.
Int Immunopharmacol ; 78: 106009, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31771815

RESUMEN

BACKGROUND: The lung infections by Staphylococcus aureus are strongly associated with its ability to produce enterotoxins. However, little is known about the mechanisms underlying trafficking of bone marrow (BM) neutrophils during airway inflammation induced by Staphylococcal enterotoxin B (SEB). We therefore aimed to investigate the effects of mouse airways SEB exposure on BM neutrophil counts and its adhesive properties as well as on the release of cytokines/chemokines that orchestrate BM neutrophils trafficking to lung tissue. METHODS: Male BALB/c mice were intranasally exposed to SEB (1 µg), and at 4, 16 and 24 h thereafter, BM, circulating blood, bronchoalveolar lavage (BAL) fluid and lung tissue were collected. BM neutrophils adhesion, MAC-1 and LFA1-α expressions (by flow cytometry) as well as measurement of cytokine and/or chemokines levels were assayed after SEB-airway exposure. RESULTS: Prior exposure to SEB promoted a marked influx of neutrophils to BAL and lung tissue, which was accompanied by increased counts of BM immature neutrophils and blood neutrophilia. BM neutrophil expressions of LFA1-α and MAC-1 were unchanged by SEB exposure whereas a significant enhancement of adhesion properties to VCAM-1 was observed. The early phase of airway SEB exposure was accompanied by high levels of GM-CSF, G-CSF, IFN-γ, TNF-α and KC/CXCL1, while the latter phase by the equilibrated actions of SDF1-α and MIP-2. CONCLUSION: Mouse airways exposure to SEB induces BM cytokines/chemokines release and their integrated actions enhance the adhesion of BM neutrophils leading to acute lung injury.


Asunto(s)
Células de la Médula Ósea/inmunología , Citocinas/metabolismo , Neutrófilos/inmunología , Neumonía Estafilocócica/inmunología , Staphylococcus aureus/inmunología , Administración Intranasal , Animales , Médula Ósea/inmunología , Médula Ósea/patología , Células de la Médula Ósea/metabolismo , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Citocinas/inmunología , Enterotoxinas/administración & dosificación , Enterotoxinas/inmunología , Humanos , Recuento de Leucocitos , Pulmón/citología , Pulmón/inmunología , Pulmón/patología , Masculino , Ratones , Infiltración Neutrófila/inmunología , Neutrófilos/metabolismo , Neumonía Estafilocócica/microbiología , Neumonía Estafilocócica/patología , Staphylococcus aureus/metabolismo
9.
Cell Prolif ; 53(1): e12721, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31782850

RESUMEN

OBJECTIVES: Secondary bacterial pneumonia is common following influenza infection. However, it remains unclear about the underlying molecular mechanisms. MATERIALS AND METHODS: We established a mouse model of post-influenza S aureus pneumonia using conditional Shp2 knockout mice (LysMCre/+ :Shp2flox/flox ). The survival, bacterial clearance, pulmonary histology, phenotype of macrophages, and expression of type I interferons and chemokines were assessed between SHP2 deletion and control mice (Shp2flox/flox ). We infused additional KC and MIP-2 to examine the reconstitution of antibacterial immune response in LysMCre/+ :Shp2flox/flox mice. The effect of SHP2 on signal molecules including MAPKs (JNK, p38 and Erk1/2), NF-κB p65 and IRF3 was further detected. RESULTS: LysMCre/+ :Shp2flox/flox mice displayed impaired antibacterial immunity and high mortality compared with control mice in post-influenza S aureus pneumonia. The attenuated antibacterial ability was associated with the induction of type I interferon and suppression of chemo-attractants KC and MIP-2, which reduced the infiltration of neutrophils into the lung upon secondary bacterial invasion. In additional, Shp2 knockout mice displayed enhanced polarization to alternatively activated macrophages (M2 phenotype). Further in vitro analyses consistently demonstrated that SHP2-deficient macrophages were skewed towards an M2 phenotype and had a decreased antibacterial capacity. Moreover, SHP2 modulated the inflammatory response to secondary bacterial infection via interfering with NF-κB and IRF3 signalling in macrophages. CONCLUSIONS: Our findings reveal that the SHP2 expression enhances the host immune response and prompts bacterial clearance in post-influenza S aureus pneumonia.


Asunto(s)
Virus de la Influenza A/inmunología , Macrófagos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Neumonía Estafilocócica/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/deficiencia , Staphylococcus aureus/inmunología , Animales , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Macrófagos/patología , Ratones , Ratones Transgénicos , FN-kappa B/genética , FN-kappa B/inmunología , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/patología , Neumonía Estafilocócica/etiología , Neumonía Estafilocócica/genética , Neumonía Estafilocócica/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/inmunología
10.
Pak J Pharm Sci ; 32(4): 1621-1630, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31608882

RESUMEN

Staphylococcus aureus and Pseudomonas aeruginosa are largely the cause of morbidity and mortality in both hospital and community settings. These pathogens remain the important cause of pulmonary infections in patients with cystic fibrosis with a worldwide prevalence. Although, antibiotics are efficient measures of treating bacterial lung infections, the occurrence of antibiotic resistant bacteria has been encouraging the researchers to explore novel therapeutic approaches. It has been discovered that certain lactic acid bacteria possess protective effects against bacterial and viral respiratory infections. The aim of present study was to investigate the capability of orally administered L. acidophilus to ameliorate S. aureus and P. aeruginosa pulmonary infections. Animals were exposed to aerosol of pathogenic suspension. After 24 hours of infection, L. acidophilus treatment was administered orally for 7 consecutive days. Evaluation of tissue bacteriology, histopathology and serum cytokinomics were performed. In parallel, human alveolar A549 cells were utilized to determine possible role of probiotic on pulmonary infections. Oral administration of L. acidophilus significantly (P<0.05) alleviate lung bacterial load and severity of infection as depicted by our histopathological studies. Results obtained from cytokinomics revealed that pro-inflammatory cytokines induced due to lung infection were suppressed in oral probiotic treatment groups. In addition, treatment with L. acidophilus induced murine lung anti inflammatory, IL-10 cytokine level. Current work suggests that orally administered L. acidophilus in mice is able to attenuate S. aureus and P. aeruginosa induced lung cytotoxicity by modulation of host immune response.


Asunto(s)
Lactobacillus acidophilus , Neumonía Estafilocócica/terapia , Probióticos/farmacología , Infecciones por Pseudomonas/terapia , Células A549 , Administración Oral , Animales , Citocinas/sangre , Citocinas/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/metabolismo , Neumonía Estafilocócica/patología , Probióticos/administración & dosificación , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa
11.
Artículo en Inglés | MEDLINE | ID: mdl-31380296

RESUMEN

α-Hemolysin (Hla) is a significant virulence factor in Staphylococcus aureus (S. aureus)-caused infectious diseases such as pneumonia. Thus, to prevent the production of Hla when treating S. aureus infection, it is necessary to choose an antibiotic with good antibacterial activity and effect. In our study, we observed that Fosfomycin (FOM) at a sub-inhibitory concentration inhibited expression of Hla. Molecular dynamics demonstrated that FOM bound to the binding sites LYS 154 and ASP 108 of Hla, potentially inhibiting Hla. Furthermore, we verified that staphylococcal membrane-derived vesicles (SMVs) contain Hla and that FOM treatment significantly reduced the production of SMVs and Hla. Based on our pharmacological inhibition analysis, ERK and p38 activated NLRP3 inflammasomes. Moreover, FOM inhibited expression of MAPKs and NLRP3 inflammasome-related proteins in S. aureus as well as SMV-infected human macrophages (MΦ) and alveolar epithelial cells. In vivo, SMVs isolated from S. aureus DU1090 (an isogenic Hla deletion mutant) or the strain itself caused weaker inflammation than that of its parent strain 8325-4. FOM also significantly reduced the phosphorylation levels of ERK and P38 and expression of NLRP3 inflammasome-related proteins. In addition, FOM decreased MPO activity, pulmonary vascular permeability and edema formation in the lungs of mice with S. aureus-caused pneumonia. Taken together, these data indicate that FOM exerts protective effects against S. aureus infection in vitro and in vivo by inhibiting Hla in SMVs and blocking ERK/P38-mediated NLRP3 inflammasome activation by Hla.


Asunto(s)
Antibacterianos/farmacología , Toxinas Bacterianas/antagonistas & inhibidores , Fosfomicina/farmacología , Proteínas Hemolisinas/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Neumonía Estafilocócica/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos , Factores de Virulencia/antagonistas & inhibidores , Animales , Antibacterianos/química , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Sitios de Unión , Línea Celular , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Vesículas Extracelulares , Fosfomicina/química , Regulación de la Expresión Génica , Proteínas Hemolisinas/química , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Interacciones Huésped-Patógeno/genética , Humanos , Inflamasomas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neumonía Estafilocócica/microbiología , Neumonía Estafilocócica/patología , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Staphylococcus aureus/genética , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/patogenicidad , Células THP-1 , Factores de Virulencia/química , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Sci Rep ; 9(1): 5339, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30926865

RESUMEN

Staphylococcus aureus pneumonia is associated with high mortality irrespective of antibiotic susceptibility. Both MRSA and MSSA strains produce powerful cytotoxins: alpha-hemolysin(Hla) and up to five leukocidins - LukSF-PV, HlgAB, HlgCB, LukED and LukGH (LukAB) - to evade host innate defense mechanisms. Neutralizing cytotoxins has been shown to provide survival benefit in rabbit S. aureus pneumonia models. We studied the mechanisms of protection of ASN100, a combination of two human monoclonal antibodies (mAbs), ASN-1 and ASN-2, that together neutralize Hla and the five leukocidins, in rabbit MRSA and MSSA pneumonia models. Upon prophylactic passive immunization, ASN100 displayed dose-dependent increase in survival and was fully protective against all S. aureus strains tested at 5 or 20 mg/kg doses. Macroscopic and microscopic lung pathology, edema rate, and bacterial burden were evaluated 12 hours post infection and reduced by ASN100. Pharmacokinetic analysis of ASN100 in bronchoalveolar-lavage fluid from uninfected animals detected efficient penetration to lung epithelial lining fluid reaching peak levels between 24 and 48 hours post dosing that were comparable to the mAb concentration measured in serum. These data confirm that the ASN100 mAbs neutralize the powerful cytotoxins of S. aureus in the lung and prevent damage to the mucosal barrier and innate immune cells.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Inmunoglobulina G/inmunología , Inmunotoxinas/inmunología , Neumonía Estafilocócica/inmunología , Neumonía Estafilocócica/prevención & control , Staphylococcus aureus/inmunología , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/inmunología , Biopsia , Modelos Animales de Enfermedad , Relación Dosis-Respuesta Inmunológica , Inmunización Pasiva , Inmunohistoquímica , Inmunotoxinas/administración & dosificación , Staphylococcus aureus Resistente a Meticilina/inmunología , Neumonía Estafilocócica/mortalidad , Neumonía Estafilocócica/patología , Pronóstico , Conejos
13.
Ann Pharmacother ; 53(6): 627-638, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30600697

RESUMEN

OBJECTIVE: To describe the diagnostic performance characteristics of methicillin-resistant Staphylococcus aureus (MRSA) nasal screening for patients with pneumonia. DATA SOURCES: PubMed and Scopus were searched from 1 January 1990 to 12 December 2018 using terms methicillin-resistant Staphylococcus aureus AND (screening OR active surveillance OR surveillance culture OR targeted surveillance OR chromogenic OR PCR OR polymerase chain reaction OR rapid test) AND (nares OR nasal) AND (pneumonia OR respiratory). STUDY SELECTION AND DATA EXTRACTION: Relevant studies in humans and English were considered. DATA SYNTHESIS: In all, 19 studies, including 21 790 patients, were included. Nasal screening for MRSA had a high negative predictive value (NPV; 76% to 99.4% for relevant studies) across all types of pneumonia. Time from nasal screening to culture varied across studies. Relevance to Patient Care and Clinical Practice: MRSA nasal screening has a high NPV for MRSA involvement in pneumonia. Utilizing this test for antimicrobial stewardship program (ASP) purposes can provide a valuable tool for reducing unwarranted anti-MRSA agents and may provide additional cost benefits. A cutoff of 7 days between nasal swab and culture or infection onset seems most appropriate for use of this test for anti-MRSA agent de-escalation for ASP purposes. CONCLUSIONS: Consideration for the inclusion of the utility of MRSA nasal screening in MRSA pneumonia should be made for future pneumonia and ASP guidelines. Additional studies are warranted to fully evaluate specific pneumonia classifications, culture types, culture timing, and clinical outcomes associated with the use of this test in patients with pneumonia.


Asunto(s)
Tamizaje Masivo/métodos , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Neumonía Estafilocócica/diagnóstico , Infecciones Estafilocócicas/diagnóstico , Femenino , Humanos , Masculino , Meticilina , Persona de Mediana Edad , Neumonía Estafilocócica/patología , Infecciones Estafilocócicas/patología
14.
Clin Infect Dis ; 68(3): 365-372, 2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-29893805

RESUMEN

Background: Coinfection with influenza virus and methicillin-resistant Staphylococcus aureus (MRSA) causes life-threatening necrotizing pneumonia in children. Sporadic incidence precludes evaluation of antimicrobial efficacy. We assessed the clinical characteristics and outcomes of critically ill children with influenza-MRSA pneumonia and evaluated antibiotic use. Methods: We enrolled children (<18 years) with influenza infection and respiratory failure across 34 pediatric intensive care units 11/2008-5/2016. We compared baseline characteristics, clinical courses, and therapies in children with MRSA coinfection, non-MRSA bacterial coinfection, and no bacterial coinfection. Results: We enrolled 170 children (127 influenza A, 43 influenza B). Children with influenza-MRSA pneumonia (N = 30, 87% previously healthy) were older than those with non-MRSA (N = 61) or no (N = 79) bacterial coinfections. Influenza-MRSA was associated with increased leukopenia, acute lung injury, vasopressor use, extracorporeal life support, and mortality than either group (P ≤ .0001). Influenza-related mortality was 40% with MRSA compared to 4.3% without (relative risk [RR], 9.3; 95% confidence interval [CI], 3.8-22.9). Of 29/30 children with MRSA who received vancomycin within the first 24 hours of hospitalization, mortality was 12.5% (N = 2/16) if treatment also included a second anti-MRSA antibiotic compared to 69.2% (N = 9/13) with vancomycin monotherapy (RR, 5.5; 95% CI, 1.4, 21.3; P = .003). Vancomycin dosing did not influence initial trough levels; 78% were <10 µg/mL. Conclusions: Influenza-MRSA coinfection is associated with high fatality in critically ill children. These data support early addition of a second anti-MRSA antibiotic to vancomycin in suspected severe cases.


Asunto(s)
Antibacterianos/uso terapéutico , Coinfección/tratamiento farmacológico , Enfermedad Crítica , Gripe Humana/complicaciones , Staphylococcus aureus Resistente a Meticilina/aislamiento & purificación , Neumonía Estafilocócica/tratamiento farmacológico , Vancomicina/uso terapéutico , Adolescente , Niño , Preescolar , Coinfección/microbiología , Coinfección/mortalidad , Coinfección/patología , Femenino , Humanos , Lactante , Recién Nacido , Gripe Humana/mortalidad , Gripe Humana/patología , Masculino , Neumonía Estafilocócica/microbiología , Neumonía Estafilocócica/mortalidad , Neumonía Estafilocócica/patología , Estudios Prospectivos , Análisis de Supervivencia , Resultado del Tratamiento
15.
Acta Cir Bras ; 33(11): 983-990, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30517325

RESUMEN

PURPOSE: To investigate the efficacy and mechanisms of root tuber of Polygonum ciliinerve (Nakai) ohwi (rPC) which has been used to treat bacterial infection in traditional Chinese medicine. METHODS: With the mouse model of Staphylococcus aureus (S. aureus) pneumonia, the phenotype of rPC treated mice, including body weight, mortality, lung slices and bacterial burden were evaluated. Furthermore, inflammatory factors in bronchoalveolar lavage (BAL) were determined by ELISA and the distribution of T cells in lung was assessed by immunofluorescence assay. RESULTS: rPC treatment could dose-dependently reduce weight loss and mortality in S. aureus-infected mice. Upon 10 mg/ml rPC treatment, S. aureus-infected mice showed about 8 grams increase in body weight (P<0.001) and 50% enhancement in mortality. The integrity of lung tissue and bacterial burden were also improved by rPC treatment. Moreover, rPC was found to modulate the immune response in infection. CONCLUSION: rPC has therapeutic potential for S. aureus infections and pneumonia with immunomodulatory functions.


Asunto(s)
Antibacterianos/farmacología , Medicamentos Herbarios Chinos/farmacología , Inmunomodulación/efectos de los fármacos , Neumonía Estafilocócica/prevención & control , Polygonum/química , Sustancias Protectoras/farmacología , Staphylococcus aureus/efectos de los fármacos , Animales , Líquido del Lavado Bronquioalveolar/química , Quimiocina CCL2/análisis , Recuento de Colonia Microbiana , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , Interleucina-6/análisis , Pulmón/efectos de los fármacos , Pulmón/patología , Ratones Endogámicos C57BL , Neumonía Estafilocócica/tratamiento farmacológico , Neumonía Estafilocócica/patología , Reproducibilidad de los Resultados , Factores de Tiempo , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/análisis
16.
Acta cir. bras ; 33(11): 983-990, Nov. 2018. graf
Artículo en Inglés | LILACS | ID: biblio-973479

RESUMEN

Abstract Purpose: To investigate the efficacy and mechanisms of root tuber of Polygonum ciliinerve (Nakai) ohwi (rPC) which has been used to treat bacterial infection in traditional Chinese medicine. Methods: With the mouse model of Staphylococcus aureus (S. aureus) pneumonia, the phenotype of rPC treated mice, including body weight, mortality, lung slices and bacterial burden were evaluated. Furthermore, inflammatory factors in bronchoalveolar lavage (BAL) were determined by ELISA and the distribution of T cells in lung was assessed by immunofluorescence assay. Results: rPC treatment could dose-dependently reduce weight loss and mortality in S. aureus-infected mice. Upon 10 mg/ml rPC treatment, S. aureus-infected mice showed about 8 grams increase in body weight (P<0.001) and 50% enhancement in mortality. The integrity of lung tissue and bacterial burden were also improved by rPC treatment. Moreover, rPC was found to modulate the immune response in infection. Conclusion: rPC has therapeutic potential for S. aureus infections and pneumonia with immunomodulatory functions.


Asunto(s)
Animales , Neumonía Estafilocócica/prevención & control , Staphylococcus aureus/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Sustancias Protectoras/farmacología , Polygonum/química , Inmunomodulación/efectos de los fármacos , Antibacterianos/farmacología , Neumonía Estafilocócica/patología , Neumonía Estafilocócica/tratamiento farmacológico , Factores de Tiempo , Ensayo de Inmunoadsorción Enzimática , Líquido del Lavado Bronquioalveolar/química , Inmunohistoquímica , Recuento de Colonia Microbiana , Reproducibilidad de los Resultados , Interleucina-6/análisis , Factor de Necrosis Tumoral alfa/análisis , Resultado del Tratamiento , Quimiocina CCL2/análisis , Pulmón/efectos de los fármacos , Pulmón/patología , Ratones Endogámicos C57BL
17.
Cell Host Microbe ; 23(5): 594-606.e7, 2018 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-29706505

RESUMEN

Staphylococcus aureus is a commensal bacterium that can asymptomatically colonize its host but also causes invasive infections. Quorum sensing regulates S. aureus virulence and the transition from a commensal to a pathogenic organism. However, little is known about how host innate immunity affects interbacterial communication. We show that nitric oxide suppresses staphylococcal virulence by targeting the Agr quorum sensing system. Nitric oxide-mediated inhibition occurs through direct modification of cysteine residues C55, C123, and C199 of the AgrA transcription factor. Cysteine modification decreases AgrA promoter occupancy as well as transcription of the agr operon and quorum sensing-activated toxin genes. In a staphylococcal pneumonia model, mice lacking inducible nitric oxide synthase develop more severe disease with heightened mortality and proinflammatory cytokine responses. In addition, staphylococcal α-toxin production increases in the absence of nitric oxide or nitric oxide-sensitive AgrA cysteine residues. Our findings demonstrate an anti-virulence mechanism for nitric oxide in innate immunity.


Asunto(s)
Comunicación Celular/inmunología , Comunicación Celular/fisiología , Interacciones Huésped-Patógeno/inmunología , Interacciones Huésped-Patógeno/fisiología , Óxido Nítrico/antagonistas & inhibidores , Staphylococcus/efectos de los fármacos , Staphylococcus/patogenicidad , Animales , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Cisteína , Citocinas/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Proteínas Hemolisinas/metabolismo , Inmunidad Innata , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/genética , Operón , Neumonía Estafilocócica/metabolismo , Neumonía Estafilocócica/patología , Regiones Promotoras Genéticas/efectos de los fármacos , Percepción de Quorum/efectos de los fármacos , Percepción de Quorum/genética , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/patogenicidad , Transactivadores/metabolismo , Factores de Transcripción/efectos de los fármacos , Virulencia/efectos de los fármacos
18.
World J Microbiol Biotechnol ; 34(5): 64, 2018 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-29671126

RESUMEN

Staphylococcus aureus (S. aureus) is a common pathogenic bacterium that causes various diseases in both humans and animals. With the increased prevalence of methicillin-resistant S. aureus, the therapeutic effects of commonly used antibiotics are limited against S. aureus infection. Novel treatment strategies and new antibiotics are needed urgently to address this concern. Many studies have shown that virulence factors secreted from S. aureus play vital roles in their pathogenic processes. Alpha-hemolysin (Hla), an important exotoxin in S. aureus, is one such virulence factor that increases sensitivity of multiple host cells to S. aureus resulting in various diseases. Eriodictyol is a flavonoid compound that exists in many fruits and vegetables. In this study, eriodictyol was demonstrated to inhibit the expression of Hla by hemolysis assays, western blotting, and RT-qPCR at the sub-minimal inhibitory concentration. In live/dead and cytotoxicity assays, the results showed that eriodictyol protected A549 cells against Hla-induced injury in a dose-dependent manner. The minimal inhibitory concentration of eriodictyol against S. aureus was 512 µg/mL. Eriodictyol can downregulate S. aureus Hla at both the expressional and transcriptional levels without affecting S. aureus growth. In addition, cell assays had proved that eriodictyol could protect A549 cells against Hla damage. Eriodictyol could therefore have the potential to treat S. aureus infection targeting Hla.


Asunto(s)
Antibacterianos/farmacología , Toxinas Bacterianas/toxicidad , Flavanonas/farmacología , Proteínas Hemolisinas/efectos de los fármacos , Proteínas Hemolisinas/toxicidad , Lesión Pulmonar/prevención & control , Staphylococcus aureus/efectos de los fármacos , Células A549/efectos de los fármacos , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Hemólisis , Humanos , Lesión Pulmonar/microbiología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/metabolismo , Pruebas de Sensibilidad Microbiana , Neumonía Estafilocócica/microbiología , Neumonía Estafilocócica/patología , Neumonía Estafilocócica/prevención & control , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidad , Factores de Virulencia/metabolismo
19.
Cell Death Dis ; 9(3): 362, 2018 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-29500427

RESUMEN

Given its high resistance, enhanced virulence, and high transmissibility, community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) pneumonia is highly associated with high morbidity and mortality. Anti-virulence therapy is a promising strategy that bypasses the evolutionary pressure on the bacterium to develop resistance. RNAIII-inhibiting peptide (RIP), as an accessory gene regulator (agr)-specific inhibitor, significantly restricts the virulence of S. aureus and protects infected mice from death by blocking the agr quorum sensing system. The protective effects of RIP on the neutropenic mice completely disappeared in a neutrophil-deleted mouse infection model, but not in the macrophage-deleted mice. This result confirmed that the in vivo antibacterial activity of RIP is highly associated with neutrophil function. Phenol-soluble modulins (PSMs), as major leukocyte lysis toxins of CA-MRSA, are directly regulated by the agr system. In this experiment, PSMα1, 2, and 3 significantly induced neutrophil necroptosis by activating mixed lineage kinase-like protein (MLKL) phosphorylation and increasing lactate dehydrogenase release. The S. aureus supernatants harvested from the agr or psmα mutant strains both decreased the phosphorylation level of MLKL and cell lysis. PSMα1-mediated neutrophil lysis was significantly inhibited by necrosulfonamide, necrostatin-1, TNFα antibody, and WRW4. These results showed PSMα1 induced necroptosis depends on formylpeptide receptor 2 (FPR2)-mediated autocrine TNFα. Moreover, the neutrophil necroptosis induced by S. aureus was significantly suppressed and pneumonia was effectively prevented by the blockage of agrA and psmα expression levels. These findings indicate that PSMα-induced necroptosis is a major cause of lung pathology in S. aureus pneumonia and suggest that interfering with the agr quorum sensing signaling pathway is a potential therapeutic strategy.


Asunto(s)
Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Pulmón/patología , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Neutrófilos/metabolismo , Neumonía Estafilocócica/patología , Transactivadores/metabolismo , Animales , Anticuerpos/metabolismo , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos BALB C , Necrosis , Fosforilación , Proteínas Quinasas/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Virulencia
20.
J Infect Dis ; 218(4): 659-668, 2018 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-29378030

RESUMEN

Staphylococcus aureus is a major cause of both community- and healthcare-acquired pneumonias. Inducible costimulator (ICOS) is part of the CD28 family of proteins and is a target for immune checkpoint therapy. We found ICOS highly expressed on activated CD4 cells in response to S. aureus. In the absence of ICOS, mice had improved survival in a pneumonia model with the methicillin-resistant Staphylococcus aureus (MRSA) strain USA300 and significant reductions in bacterial burden in a nonlethal acute pneumonia model. Infected Icos-/- mice had major reductions in several proinflammatory cytokines, neutrophils, inflammatory monocytes, and eosinophils compared to infected wild-type mice, while there was improved expression of CD11c and macrophage receptor with collagenous structure on the surface of alveolar macrophages. Early during infection infected Icos-/- mice had increased numbers of alveolar macrophages and expression of several surface markers on alveolar macrophages and neutrophils. ICOS signaling also contributed to the pathogenesis of the airway pathogens Klebsiella pneumoniae, Pseudomonas aeruginosa, and Streptococcus pneumoniae, and neutralizing antibody to ICOS led to improved clearance of S. aureus from the airway. Our results indicate that ICOS plays a significant role in orchestrating the innate immune response to S. aureus and other airway pathogens, and could be a potential immunomodulatory target to attenuate S. aureus-related immunopathology.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Interacciones Huésped-Patógeno , Inmunidad Innata , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Staphylococcus aureus Resistente a Meticilina/crecimiento & desarrollo , Neumonía Estafilocócica/patología , Animales , Carga Bacteriana , Modelos Animales de Enfermedad , Factores Inmunológicos/análisis , Proteína Coestimuladora de Linfocitos T Inducibles/deficiencia , Infecciones por Klebsiella/patología , Pulmón/patología , Macrófagos Alveolares/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones Neumocócicas/patología , Neumonía Estafilocócica/microbiología , Infecciones por Pseudomonas/patología , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...