Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
1.
Infect Immun ; 91(10): e0016623, 2023 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-37768067

RESUMO

In this study, we examined the effect of Il9 deletion on macrophages in methicillin-resistant Staphylococcus aureus (MRSA) infection. MRSA-infected mice were employed for the in vivo experiments, and RAW264.7 cells were stimulated with MRSA for the in vitro experiments. Macrophage polarization was determined by flow cytometry and quantitative real-time PCR; macrophage phagocytosis was assessed by flow cytometry and laser scanning confocal microscopy; cell apoptosis was assessed by flow cytometry and western blotting. Il9 deletion markedly elevated macrophage phagocytosis and M2 macrophages in MRSA infection, which was accompanied by elevated expression of Il10 and Arg1 and reduced expression of Inos, tumor necrosis factor-α (Tnfα), and Il6. Il9 deletion also inhibited macrophage apoptosis in MRSA infection, which was manifested by elevated B-cell lymphoma 2 (BCL-2) protein level and reduced protein levels of cleaved cysteine protease 3 (CASPASE-3) and BCL2-Associated X (BAX). Both the in vivo and in vitro experiments further showed the activation of phosphoinositide 3-kinase (PI3K)/AKT (also known as protein kinase B, PKB) signaling pathway in MRSA infection and that the regulation of Il9 expression may be dependent on Toll-like receptor (TLR) 2/PI3K pathway. The above results showed that Il9 deletion exhibited a protective role against MRSA infection by promoting M2 polarization and phagocytosis of macrophages and the regulation of Il9 partly owing to the activation of TLR2/PI3K pathway, proposing a novel therapeutic strategy for MRSA-infected pneumonia.


Assuntos
Interleucina-9 , Staphylococcus aureus Resistente à Meticilina , Fagocitose , Pneumonia Estafilocócica , Animais , Camundongos , Interleucina-9/genética , Interleucina-9/metabolismo , Macrófagos/metabolismo , Staphylococcus aureus Resistente à Meticilina/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Pneumonia Estafilocócica/tratamento farmacológico , Pneumonia Estafilocócica/imunologia
2.
JCI Insight ; 7(4)2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-34990413

RESUMO

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.


Assuntos
Fibrose Pulmonar Idiopática/imunologia , Imunidade Inata/imunologia , Macrófagos Alveolares/metabolismo , Staphylococcus aureus Resistente à Meticilina/isolamento & purificação , Neutrófilos/patologia , Pneumonia Estafilocócica/patologia , Animais , Modelos Animais de Doenças , Fibrose Pulmonar Idiopática/complicações , Macrófagos Alveolares/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/metabolismo , Fagocitose , Pneumonia Estafilocócica/etiologia , Pneumonia Estafilocócica/imunologia
3.
J Immunol ; 207(5): 1371-1376, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34380647

RESUMO

Inflammatory cytokine storm is a known cause for acute respiratory distress syndrome. In this study, we have investigated the role of IFN-γ in lethal lung inflammation using a mouse model of postinfluenza methicillin-resistant Staphylococcus aureus (MRSA) pneumonia. To mimic the clinical scenario, animals were treated with antibiotics for effective bacterial control following MRSA superinfection. However, antibiotic therapy alone is not sufficient to improve survival of wild-type animals in this lethal acute respiratory distress syndrome model. In contrast, antibiotics induce effective protection in mice deficient in IFN-γ response. Mechanistically, we show that rather than inhibiting bacterial clearance, IFN-γ promotes proinflammatory cytokine response to cause lethal lung damage. Neutralization of IFN-γ after influenza prevents hyperproduction of TNF-α, and thereby protects against inflammatory lung damage and animal mortality. Taken together, the current study demonstrates that influenza-induced IFN-γ drives a stepwise propagation of inflammatory cytokine response, which ultimately results in fatal lung damage during secondary MRSA pneumonia, despite of antibiotic therapy.


Assuntos
Antibacterianos/uso terapêutico , Inflamação/imunologia , Vírus da Influenza A/fisiologia , Influenza Humana/imunologia , Interferon gama/metabolismo , Pulmão/imunologia , Infecções por Orthomyxoviridae/imunologia , Pneumonia Estafilocócica/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/fisiologia , Animais , Células Cultivadas , Humanos , Influenza Humana/complicações , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/complicações , Pneumonia Estafilocócica/complicações , Infecções Estafilocócicas/complicações , Superinfecção , Fator de Necrose Tumoral alfa
4.
Front Immunol ; 12: 636861, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34135886

RESUMO

STING (Stimulator of interferon genes) is known as an important adaptor protein or direct sensor in the detection of nucleotide originating from pathogens or the host. The implication of STING during pulmonary microbial infection remains unknown to date. Herein, we showed that STING protected against pulmonary S.aureus infection by suppressing necroptosis. STING deficiency resulted in increased mortality, more bacteria burden in BALF and lungs, severe destruction of lung architecture, and elevated inflammatory cells infiltration and inflammatory cytokines secretion. STING deficiency also had a defect in bacterial clearance, but did not exacerbate pulmonary inflammation during the early stage of infection. Interestingly, TUNEL staining and LDH release assays showed that STING-/- mice had increased cell death than WT mice. We further demonstrated that STING-/- mice had decreased number of macrophages accompanied by increased dead macrophages. Our in vivo and in vitro findings further demonstrated this cell death as necroptosis. The critical role of necroptosis was detected by the fact that MLKL-/- mice exhibited decreased macrophage death and enhanced host defense to S.aureus infection. Importantly, blocking necroptosis activation rescued host defense defect against S.aureus pneumonia in STING-/- mice. Hence, these results reveal an important role of STING in suppressing necroptosis activation to facilitate early pathogen control during pulmonary S.aureus infection.


Assuntos
Pulmão/patologia , Macrófagos/imunologia , Proteínas de Membrana/metabolismo , Pneumonia Estafilocócica/imunologia , Proteínas Quinases/metabolismo , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/fisiologia , Animais , Carga Bacteriana , Células Cultivadas , Citocinas/metabolismo , Imunidade , Mediadores da Inflamação/metabolismo , Pulmão/microbiologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Necroptose , Proteínas Quinases/genética
5.
Am J Respir Cell Mol Biol ; 64(5): 569-578, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33571420

RESUMO

Pneumonia-induced lung injury and acute respiratory distress syndrome can develop because of an inappropriate inflammatory response to acute infections, leading to a compromised alveolar barrier. Recent work suggests that hospitalized patients with allergies/asthma are less likely to die of pulmonary infections and that there is a correlation between survival from acute respiratory distress syndrome and higher eosinophil counts; thus, we hypothesized that eosinophils associated with a type 2 immune response may protect against pneumonia-induced acute lung injury. To test this hypothesis, mice were treated with the type 2-initiating cytokine IL-33 intratracheally 3 days before induction of pneumonia with airway administration of a lethal dose of Staphylococcus aureus. Interestingly, IL-33 pretreatment promoted survival by inhibiting acute lung injury: amount of BAL fluid proinflammatory cytokines and pulmonary edema were both reduced, with an associated increase in oxygen saturation. Pulmonary neutrophilia was also reduced, whereas eosinophilia was strongly increased. This eosinophilia was key to protection; eosinophil reduction eliminated both IL-33-mediated protection against mortality and inhibition of neutrophilia and pulmonary edema. Together, these data reveal a novel role for eosinophils in protection against lung injury and suggest that modulation of pulmonary type 2 immunity may represent a novel therapeutic strategy.


Assuntos
Lesão Pulmonar Aguda/imunologia , Eosinófilos/imunologia , Interleucina-33/imunologia , Pneumonia Estafilocócica/imunologia , Edema Pulmonar/imunologia , Síndrome do Desconforto Respiratório/imunologia , Staphylococcus aureus/patogenicidade , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/microbiologia , Lesão Pulmonar Aguda/prevenção & controle , Animais , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Toxina Diftérica/farmacologia , Modelos Animais de Doenças , Eosinófilos/efeitos dos fármacos , Feminino , Expressão Gênica , Humanos , Interleucina-33/genética , Interleucina-33/farmacologia , Interleucina-5/deficiência , Interleucina-5/genética , Interleucina-5/imunologia , Contagem de Leucócitos , Procedimentos de Redução de Leucócitos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Pneumonia Estafilocócica/complicações , Pneumonia Estafilocócica/microbiologia , Pneumonia Estafilocócica/mortalidade , Edema Pulmonar/complicações , Edema Pulmonar/microbiologia , Edema Pulmonar/mortalidade , Síndrome do Desconforto Respiratório/etiologia , Síndrome do Desconforto Respiratório/microbiologia , Síndrome do Desconforto Respiratório/prevenção & controle , Staphylococcus aureus/imunologia , Análise de Sobrevida
6.
J Infect Dis ; 224(5): 903-913, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-33417695

RESUMO

BACKGROUND: Staphylococcus aureus causes community- and hospital-acquired pneumonia linked to a high mortality rate. The emergence and rapid transmission of multidrug-resistant S. aureus strains has become a serious health concern, highlighting the challenges associated with the development of a vaccine to combat S. aureus pneumonia. METHODS: This study evaluated the effects of intrapulmonary immunization on the immune response and protection against S. aureus lung infection in a respiratory mouse model using a subunit vaccine. RESULTS: Compared with the intranasal immunized mice, the intrapulmonarily immunized mice had lower levels of pulmonary bacterial colonization and lethality, accompanied by alleviated lung inflammation with reduced proinflammatory cytokines and increased levels of interleukin-10 and antimicrobial peptide following intrapulmonary challenge. Optimal protection was associated with increased pulmonary antibodies and resident memory T cells. Moreover, intrapulmonary immunization provided long-lasting pulmonary protection for at least 6 months, with persistent cellular and humoral immunity in the lungs. CONCLUSIONS: Vaccine reaching the deep lung by intrapulmonary immunization plays a significant role in the induction of efficacious and long-lasting immunity against S. aureus in the lung parenchyma. Hence, intrapulmonary immunization can be a strategy for the development of a vaccine against S. aureus pneumonia.Immunization through the intrapulmonary route with a subunit of S. aureus vaccine elicited tissue resident memory T cells and antigen-specific antibodies in the lungs, and provided optimal and long-term protection against S. aureus pneumonia.


Assuntos
Células T de Memória , Pneumonia Estafilocócica/prevenção & controle , Staphylococcus aureus/imunologia , Vacinação , Animais , Peptídeos Antimicrobianos , Pulmão , Staphylococcus aureus Resistente à Meticilina , Camundongos , Pneumonia Estafilocócica/imunologia
7.
Infect Immun ; 88(10)2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32690637

RESUMO

Staphylococcus aureus is a leading cause of bacterial pneumonia, and we have shown previously that type I interferon (IFN) contributes to the pathogenesis of this disease. In this study, we screened 75 S. aureus strains for their ability to induce type I and III IFN. Both cytokine pathways were differentially stimulated by various S. aureus strains independently of their isolation sites or methicillin resistance profiles. These induction patterns persisted over time, and type I and III IFN generation differentially correlated with tumor necrosis factor alpha production. Investigation of one isolate, strain 126, showed a significant defect in type I IFN induction that persisted over several time points. The lack of induction was not due to differential phagocytosis, subcellular location, or changes in endosomal acidification. A correlation between reduced type I IFN induction levels and decreased autolysis and lysostaphin sensitivity was found between strains. Strain 126 had a decreased rate of autolysis and increased resistance to lysostaphin degradation and host cell-mediated killing. This strain displayed decreased virulence in a murine model of acute pneumonia compared to USA300 (current epidemic strain and commonly used in research) and had reduced capacity to induce multiple cytokines. We observed this isolate to be a vancomycin-intermediate S. aureus (VISA) strain, and reduced Ifnb was observed with a defined mutation in walK that induces a VISA phenotype. Overall, this study demonstrates the heterogeneity of IFN induction by S. aureus and uncovered an interesting property of a VISA strain in its inability to induce type I IFN production.


Assuntos
Citocinas/imunologia , Interferon Tipo I/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/patogenicidade , Animais , Antibacterianos/farmacologia , Carga Bacteriana , Proteínas de Bactérias/genética , Células Cultivadas , Farmacorresistência Bacteriana/genética , Lisostafina/farmacologia , Camundongos , Testes de Sensibilidade Microbiana , Mutação , Pneumonia Estafilocócica/imunologia , Pneumonia Estafilocócica/microbiologia , Transdução de Sinais , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/isolamento & purificação , Vancomicina/farmacologia , Virulência
8.
J Leukoc Biol ; 108(3): 967-981, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32531864

RESUMO

Staphylococcus aureus causes necrotizing pneumonia by secreting toxins such as leukocidins that target front-line immune cells. The mechanism by which leukocidins kill innate immune cells and trigger inflammation during S. aureus lung infection, however, remains unresolved. Here, we explored human-induced pluripotent stem cell-derived macrophages (hiPSC-dMs) to study the interaction of the leukocidins Panton-Valentine leukocidin (PVL) and LukAB with lung macrophages, which are the initial leukocidin targets during S. aureus lung invasion. hiPSC-dMs were susceptible to the leukocidins PVL and LukAB and both leukocidins triggered NLPR3 inflammasome activation resulting in IL-1ß secretion. hiPSC-dM cell death after LukAB exposure, however, was only temporarily dependent of NLRP3, although NLRP3 triggered marked cell death after PVL treatment. CRISPR/Cas9-mediated deletion of the PVL receptor, C5aR1, protected hiPSC-dMs from PVL cytotoxicity, despite the expression of other leukocidin receptors, such as CD45. PVL-deficient S. aureus had reduced ability to induce lung IL-1ß levels in human C5aR1 knock-in mice. Unexpectedly, inhibiting NLRP3 activity resulted in increased wild-type S. aureus lung burdens. Our findings suggest that NLRP3 induces macrophage death and IL-1ß secretion after PVL exposure and controls S. aureus lung burdens.


Assuntos
Proteínas de Bactérias/antagonistas & inibidores , Toxinas Bacterianas/antagonistas & inibidores , Exotoxinas/antagonistas & inibidores , Células-Tronco Pluripotentes Induzidas/citologia , Leucocidinas/antagonistas & inibidores , Macrófagos/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Receptor da Anafilatoxina C5a/efeitos dos fármacos , Staphylococcus aureus , Animais , Antígeno CD11b/imunologia , Sistemas CRISPR-Cas , Diferenciação Celular , Células Cultivadas , Exotoxinas/deficiência , Técnicas de Introdução de Genes , Humanos , Interleucina-1beta/metabolismo , Antígenos Comuns de Leucócito/fisiologia , Pulmão/imunologia , Pulmão/microbiologia , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/citologia , Fragmentos de Peptídeos/imunologia , Pneumonia Estafilocócica/imunologia , Subunidades Proteicas , Receptor da Anafilatoxina C5a/deficiência , Receptor da Anafilatoxina C5a/genética , Receptor da Anafilatoxina C5a/fisiologia , Proteínas Recombinantes/metabolismo , Staphylococcus aureus/fisiologia
9.
J Infect Dis ; 222(8): 1400-1404, 2020 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-32386322

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Endopeptidase Clp/metabolismo , Pneumonia Estafilocócica/microbiologia , Staphylococcus aureus/patogenicidade , Animais , Proteínas de Bactérias/genética , Modelos Animais de Doenças , Endopeptidase Clp/genética , Feminino , Interações Hospedeiro-Patógeno , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Viabilidade Microbiana , Mutação , Neutrófilos/imunologia , Pneumonia Estafilocócica/imunologia , Pneumonia Estafilocócica/patologia , Staphylococcus aureus/enzimologia , Staphylococcus aureus/genética
10.
Immunol Cell Biol ; 98(7): 577-594, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32333812

RESUMO

Staphylococcus aureus is found in the nasal cavity of up to 30% of the human population. Persistent nasal carriage of S. aureus is a risk factor for influenza virus-induced secondary bacterial pneumonia. There is limited understanding of the factors that cause S. aureus to shift from the upper to the lower respiratory tract and convert from a commensal organism to an invasive pathogen. Here we show that neutrophils actively prevent S. aureus dissemination. Establishment of a mouse model of localized S. aureus nasal carriage revealed variations in the longevity of persistence of S. aureus isolates. Improved persistence within this site was associated with reduced nasal inflammation, less neutrophil egress into the airways and reduced neutrophil-bacteria association. Neutrophil depletion of mice with localized S. aureus nasal carriage triggered the development of an invasive S. aureus infection. Moreover, utilizing a model of influenza-induced staphylococcal pneumonia we showed that treatment with granulocyte-colony-stimulating factor, a potent enhancer of neutrophil number and function, significantly reduced bacterial loads in the lung and improved disease outcomes. These data reveal that neutrophils play an important and active role in confining S. aureus to the upper respiratory tract and highlight the use of approaches that improve neutrophil function as effective strategies to attenuate morbidity associated with staphylococcal pneumonia.


Assuntos
Pulmão/microbiologia , Neutrófilos/imunologia , Pneumonia Estafilocócica/imunologia , Animais , Camundongos , Nariz/microbiologia , Infecções por Orthomyxoviridae , Staphylococcus aureus
11.
J Infect Dis ; 221(2): 267-275, 2020 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-31504652

RESUMO

Staphylococcus aureus is a common pathogen causing infections in humans with various degrees of severity, with pneumonia being one of the most severe infections. In as much as staphylococcal pneumonia is a disease driven in large part by α-hemolysin (Hla) and Panton-Valentine leukocidin (PVL), we evaluated whether active immunization with attenuated forms of Hla (HlaH35L/H48L) alone, PVL components (LukS-PVT28F/K97A/S209A and LukF-PVK102A) alone, or combination of all 3 toxoids could prevent lethal challenge in a rabbit model of necrotizing pneumonia caused by the USA300 community-associated methicillin-resistant S. aureus (MRSA). Rabbits vaccinated with Hla toxoid alone or PVL components alone were only partially protected against lethal pneumonia, whereas those vaccinated with all 3 toxoids had 100% protection against lethality. Vaccine-mediated protection correlated with induction of polyclonal antibody response that neutralized not only α-hemolysin and PVL, but also other related toxins, produced by USA300 and other epidemic MRSA clones.


Assuntos
Toxinas Bacterianas/imunologia , Exotoxinas/imunologia , Proteínas Hemolisinas/imunologia , Leucocidinas/imunologia , Pneumonia Necrosante/prevenção & controle , Pneumonia Estafilocócica/prevenção & controle , Animais , Toxinas Bacterianas/administração & dosagem , Modelos Animais de Doenças , Exotoxinas/administração & dosagem , Proteínas Hemolisinas/administração & dosagem , Humanos , Leucocidinas/administração & dosagem , Staphylococcus aureus Resistente à Meticilina , Pneumonia Necrosante/imunologia , Pneumonia Estafilocócica/imunologia , Coelhos , Vacinação
12.
Clin Exp Rheumatol ; 38(2): 245-256, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31498077

RESUMO

OBJECTIVES: To provide evidence-based recommendations for vaccination against influenza virus and S. pneumoniae in patients with autoimmune rheumatic diseases (ARDs). METHODS: A Consensus Committee including physicians with expertise in rheumatic and infectious diseases was established by two Italian scientific societies, Società Italiana di Reumatologia (SIR) and Società Italiana di Malattie Infettive e Tropicali (SIMIT). The experts were invited to develop evidence-based recommendations concerning vaccinations in ARDs patients, based on their clinical status before and after undergoing immunosuppressive treatments. Key clinical questions were formulated for the systematic literature reviews, based on the clinical pathway. A search was made in Medline (via PubMed) according to the original MeSH strategy from October 2009 and a keyword strategy from January 2016 up to December 2017, updating existing EULAR recommendations. Specific recommendations were separately voted and scored from 0 (no agreement with) to 100 (maximal agreement) and supporting evidence graded. The mean and standard deviation of the scores were calculated to determine the level of agreement among the experts' panel for each recommendation. Total cumulative agreement ≥70 defined consensus for each statement. RESULTS: Nine recommendations, based on 6 key clinical questions addressed by the expert committee, were proposed. The aim of this work is to integrate the 2011 EULAR recommendations on vaccination against influenza and S. pneumoniae in ARDs patients. An implementation plan was proposed to improve the vaccination status of these patients and their safety during immunosuppressive treatments. CONCLUSIONS: Influenza and pneumococcus vaccinations are effective and safe in patients with ARDs. More efforts should be made to translate the accumulated evidence into practice.


Assuntos
Vacinas contra Influenza/administração & dosagem , Vacinas Pneumocócicas/administração & dosagem , Doenças Reumáticas/imunologia , Vacinação , Adulto , Doenças Autoimunes/complicações , Doenças Autoimunes/tratamento farmacológico , Consenso , Medicina Baseada em Evidências/métodos , Feminino , Humanos , Imunossupressores/efeitos adversos , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Itália , Masculino , Vacinas Pneumocócicas/imunologia , Pneumonia Estafilocócica/imunologia , Pneumonia Estafilocócica/prevenção & controle , Doenças Reumáticas/complicações , Doenças Reumáticas/tratamento farmacológico , Vacinação/normas
13.
Int Immunopharmacol ; 78: 106009, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31771815

RESUMO

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.


Assuntos
Células da Medula Óssea/imunologia , Citocinas/metabolismo , Neutrófilos/imunologia , Pneumonia Estafilocócica/imunologia , Staphylococcus aureus/imunologia , Administração Intranasal , Animais , Medula Óssea/imunologia , Medula Óssea/patologia , Células da Medula Óssea/metabolismo , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Citocinas/imunologia , Enterotoxinas/administração & dosagem , Enterotoxinas/imunologia , Humanos , Contagem de Leucócitos , Pulmão/citologia , Pulmão/imunologia , Pulmão/patologia , Masculino , Camundongos , Infiltração de Neutrófilos/imunologia , Neutrófilos/metabolismo , Pneumonia Estafilocócica/microbiologia , Pneumonia Estafilocócica/patologia , Staphylococcus aureus/metabolismo
14.
Cell Prolif ; 53(1): e12721, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31782850

RESUMO

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.


Assuntos
Vírus da Influenza A/imunologia , Macrófagos/imunologia , Infecções por Orthomyxoviridae/imunologia , Pneumonia Estafilocócica/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/deficiência , Staphylococcus aureus/imunologia , Animais , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Macrófagos/patologia , Camundongos , Camundongos Transgênicos , NF-kappa B/genética , NF-kappa B/imunologia , Infecções por Orthomyxoviridae/complicações , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/patologia , Pneumonia Estafilocócica/etiologia , Pneumonia Estafilocócica/genética , Pneumonia Estafilocócica/patologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/imunologia
15.
JCI Insight ; 4(14)2019 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-31341107

RESUMO

Postinfluenza bacterial superinfections cause increased morbidity and mortality compared with singular infection with influenza during both pandemics and seasonal epidemics. Vaccines and current treatments provide limited benefit, a rationale to conduct studies utilizing alternative therapies. FY1 and an optimized version, MEDI8852, anti-influenza HA mAbs, have been shown to neutralize influenza virus during singular influenza infection. MEDI4893*, an anti-Staphylococcus aureus α-toxin mAb, has been shown to improve survival when administered prophylactically prior to S. aureus pneumonia. Our objective was to determine if mAbs can improve survival during postinfluenza bacterial pneumonia. We administered FY1 in a murine model of postinfluenza methicillin-resistant S. aureus (MRSA) pneumonia and observed improved survival rates when given early during the course of influenza infection. Our findings indicate decreased lung injury and increased uptake and binding of bacteria by macrophages in the mice that received FY1 earlier in the course of influenza infection, corresponding to decreased bacterial burden. We also observed improved survival when mice were treated with a combination of FY1 and MEDI4893* late during the course of postinfluenza MRSA pneumonia. In conclusion, both FY1 and MEDI4893* prolong survival when used in a murine model of postinfluenza MRSA pneumonia, suggesting pathogen-specific mAbs as a possible therapeutic in the context of bacterial superinfection.


Assuntos
Antibacterianos/uso terapêutico , Antivirais/uso terapêutico , Influenza Humana/tratamento farmacológico , Pneumonia Estafilocócica/tratamento farmacológico , Superinfecção/tratamento farmacológico , Animais , Antibacterianos/farmacologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Antivirais/farmacologia , Anticorpos Amplamente Neutralizantes/farmacologia , Anticorpos Amplamente Neutralizantes/uso terapêutico , Modelos Animais de Doenças , Quimioterapia Combinada/métodos , Humanos , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H1N1/patogenicidade , Influenza Humana/imunologia , Influenza Humana/mortalidade , Influenza Humana/virologia , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/virologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Staphylococcus aureus Resistente à Meticilina/imunologia , Staphylococcus aureus Resistente à Meticilina/patogenicidade , Camundongos , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Pneumonia Estafilocócica/imunologia , Pneumonia Estafilocócica/microbiologia , Pneumonia Estafilocócica/mortalidade , Superinfecção/imunologia , Superinfecção/microbiologia , Superinfecção/mortalidade , Análise de Sobrevida , Resultado do Tratamento
16.
Front Immunol ; 10: 474, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30936875

RESUMO

Interferon-inducible protein (IFI204) (p204, the murine homolog of human IFI16) is known as a cytosolic DNA sensor to recognize DNA viruses and intracellular bacteria. However, little is known about its role during extracellular bacterial infection. Here we show that IFI204 is required for host defense against the infection of Staphylococcus aureus, an extracellular bacterial pathogen. IFI204 deficiency results in decreased survival, increased bacterial loads, severe organs damage, and decreased recruitment of neutrophils and macrophages. Production of several inflammatory cytokines/chemokines including IFN-ß and KC is markedly decreased, as well as the related STING-IRF3 and NF-κB pathways are impaired. However, exogenous administration of recombinant KC or IFN-ß is unable to rescue the susceptibility of IFI204-deficient mice, suggesting that other mechanisms rather than KC and IFN-ß account for IFI204-mediated host defense. IFI204 deficiency leads to a defect in extracellular bacterial killing in macrophages and neutrophils, although bacterial engulf, and intracellular killing activity are normal. Moreover, the defect of bactericidal activity is mediated by decreased extracellular trap formation in the absence of IFI204. Adoptively transferred WT bone marrow cells significantly protect WT and IFI204-deficient recipients against Staphylococcus infection compared with transferred IFI204-deficient bone marrow cells. Hence, this study suggests that IFI204 is essential for the host defense against Staphylococcus infection.


Assuntos
DNA Bacteriano/imunologia , Macrófagos/imunologia , Neutrófilos/microbiologia , Proteínas Nucleares/imunologia , Fosfoproteínas/imunologia , Infecções Estafilocócicas/imunologia , Animais , Bacteriemia/imunologia , Bacteriemia/microbiologia , Carga Bacteriana , Transplante de Medula Óssea , Quimiocina CXCL1/biossíntese , Quimiocina CXCL1/imunologia , Quimiocina CXCL1/farmacologia , Citocinas/biossíntese , Feminino , Fator Regulador 3 de Interferon/fisiologia , Interferon beta/biossíntese , Interferon beta/imunologia , Interferon beta/farmacologia , Macrófagos/microbiologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/microbiologia , Masculino , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/fisiologia , Neutrófilos/imunologia , Proteínas Nucleares/deficiência , Fosfoproteínas/deficiência , Pneumonia Estafilocócica/imunologia , Pneumonia Estafilocócica/microbiologia , Quimera por Radiação , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Infecções Estafilocócicas/microbiologia
17.
Int Immunopharmacol ; 72: 40-47, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30959370

RESUMO

Staphylococcus aureus is a major causative microorganism in community- and healthcare-acquired pneumonia. CD5L is an important protein in the control of immune homeostasis. In this study, we found that patients with S. aureus pneumonia displayed increased levels of circulating CD5L. Likewise, mice with S. aureus pneumonia had elevated CD5L levels in the lungs. Anti-CD5L antibody protected mice from lethal pneumonia induced by methicillin-resistant S. aureus. The survival benefit obtained with antibody against CD5L was associated with an improvement of bacterial clearance and a reduction of pulmonary inflammatory cytokines and chemokines. Conversely, co-injection of recombinant CD5L and S. aureus markedly increased the lethality of S. aureus pneumonia. These findings suggest that CD5L contributed to the immunopathology of S. aureus pneumonia.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Pneumonia Estafilocócica/imunologia , Receptores Depuradores Classe B/imunologia , Animais , Anticorpos/uso terapêutico , Proteínas Reguladoras de Apoptose/imunologia , Criança , Pré-Escolar , Citocinas/imunologia , Feminino , Humanos , Pulmão/imunologia , Macrófagos/imunologia , Masculino , Camundongos Endogâmicos C57BL , Neutrófilos/imunologia , Pneumonia Estafilocócica/sangue , Pneumonia Estafilocócica/tratamento farmacológico , Receptores Imunológicos/imunologia , Receptores Depuradores , Receptores Depuradores Classe B/sangue
18.
Int Immunopharmacol ; 72: 195-203, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30991161

RESUMO

The bacterial pneumonia caused by methicillin-resistant Staphylococcus aureus (MRSA) is a potentially fatal disease, featured with extensive infection, inflammation, and airway dysfunction. With the increasing emerging of drug-resistant strains, new therapeutic strategies beyond canonical antibiotic treatment are pressingly needed. Daphnetin (DAPH) is a natural coumarin derivative with anti-inflammation, anti-microorganism and anti-oxidative properties. However, the protective effect of DAPH on S. aureus-caused pneumonia and the mechanism involved are never explored. Here we show that DAPH treatment conferred substantial protection against S. aureus-induced pneumonia, characterized by the reduced inflammatory responses, the augmented bacterial clearance and the alleviated tissue damage. Our study indicates that DAPH significantly enhanced mTOR-dependent autophagic pathway, leading to the boosted microphage bactericidal activity and the suppressed inflammatory responses. Inhibition of autophagic pathway therefore largely abolished DAPH-elicited repression of inflammatory response and macrophage anti-bacterial capability. Together, we herein not only identify a novel, natural agent to combat bacterial pneumonia, but also underscore the significance of autophagic pathway in orchestrating antimicrobial and anti-inflammatory responses, which may have important implication for the treatment of the infectious diseases, particularly that caused by obstinate, antibiotic-resistant pathogens such as MRSA.


Assuntos
Antibacterianos/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Autofagia/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina , Pneumonia Estafilocócica/tratamento farmacológico , Umbeliferonas/uso terapêutico , Animais , Antibacterianos/farmacologia , Anti-Inflamatórios/farmacologia , Citocinas/genética , Citocinas/imunologia , Feminino , Macrófagos/imunologia , Macrófagos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia Estafilocócica/imunologia , Células RAW 264.7 , Umbeliferonas/farmacologia
19.
Sci Rep ; 9(1): 5339, 2019 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-30926865

RESUMO

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.


Assuntos
Anticorpos Neutralizantes/imunologia , Imunoglobulina G/imunologia , Imunotoxinas/imunologia , Pneumonia Estafilocócica/imunologia , Pneumonia Estafilocócica/prevenção & controle , Staphylococcus aureus/imunologia , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/imunologia , Biópsia , Modelos Animais de Doenças , Relação Dose-Resposta Imunológica , Imunização Passiva , Imuno-Histoquímica , Imunotoxinas/administração & dosagem , Staphylococcus aureus Resistente à Meticilina/imunologia , Pneumonia Estafilocócica/mortalidade , Pneumonia Estafilocócica/patologia , Prognóstico , Coelhos
20.
Microbes Infect ; 21(1): 33-39, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30026066

RESUMO

Interleukin (IL)-17 is a key member of the Th17 cytokines and has been reported to be involved in the pathomechanisms underlying various diseases, including infectious diseases. Infections with community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) have garnered worldwide attention, and the representative USA300 strain is known to cause pneumonia in healthy people, which can be lethal. However, little is known about the role of IL-17 in CA-MRSA pneumonia. In this study, we investigated the role of IL-17 in a CA-MRSA pneumonia animal model. Mortality was higher and occurred at an earlier stage of infection in the IL-17A-knockout mice than in the wild-type (P < 0.01) and IL-17A/F-knockout mice (P < 0.05); however, no significant difference in the intrapulmonary bacterial counts was observed among the three groups of mice. Moreover, the IL-17A-knockout group showed significantly higher levels of IL-17F and granulocyte-colony stimulating factor (G-CSF) and a significantly higher neutrophil count in the bronchoalveolar lavage fluid than the other groups. These results confirmed that G-CSF expression significantly increased, and significant neutrophilic inflammation occurred under conditions of IL-17A deficiency in the murine CA-MRSA pneumonia model.


Assuntos
Interleucina-17/deficiência , Interleucina-17/imunologia , Staphylococcus aureus Resistente à Meticilina/imunologia , Pneumonia Estafilocócica/imunologia , Animais , Modelos Animais de Doenças , Expressão Gênica , Fator Estimulador de Colônias de Granulócitos/genética , Interleucina-17/genética , Pulmão/imunologia , Pulmão/microbiologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Knockout , Neutrófilos/imunologia , Pneumonia Estafilocócica/microbiologia , Pneumonia Estafilocócica/mortalidade , Taxa de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...