Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Animal Model Exp Med ; 6(6): 585-597, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37246733

RESUMEN

BACKGROUND: Staphylococcus aureus can cause serious infections by secreting many superantigen exotoxins in "carrier" or "pathogenic" states. HLA DQ and HLA DR humanized mice have been used as a small animal model to study the role of two molecules during S. aureus infection. However, the contribution of HLA DP to S. aureus infection is unknown yet. METHODS: In this study, we have produced HLA DP401 and HLA DRA0101 humanized mice by microinjection of C57BL/6J zygotes. Neo-floxed IAß+/- mice were crossbred with Ella-Cre and further crossbred with HLA DP401 or HLA-DRA0101 humanized mice. After several rounds of traditional crossbreeding, we finally obtained HLA DP401-IAß-/- and HLA DRA-IAß-/- humanized mice, in which human DP401 or DRA0101 molecule was introduced into IAß-/- mice deficient in endogenous murine MHC class II molecules. A transnasal infection murine model of S. aureus pneumonia was induced in the humanized mice by administering 2 × 108 CFU of S. aureus Newman dropwise into the nasal cavity. The immune responses and histopathology changes were further assessed in lungs in these infected mice. RESULTS: We evaluated the local and systemic effects of S. aureus delivered intranasally in HLA DP401-IAß-/- and HLA DRA-IAß-/- transgenic mice. S. aureus Newman infection significantly increased the mRNA level of IL 12p40 in lungs in humanized mice. An increase in IFN-γ and IL-6 protein was observed in HLA DRA-IAß-/- mice. We observed a declining trend in the percentage of F4/80+ macrophages in lungs in HLA DP401-IAß-/- mice and a decreasing ratio of CD4+ to CD8+ T cells in lungs in IAß-/- mice and HLA DP401-IAß-/- mice. A decreasing ratio of Vß3+ to Vß8+ T cells was also found in the lymph node of IAß-/- mice and HLA DP401-IAß-/- mice. S. aureus Newman infection resulted in a weaker pathological injury in lungs in IAß-/- genetic background mice. CONCLUSION: These humanized mice will be an invaluable mouse model to resolve the pathological mechanism of S. aureus pneumonia and study what role DP molecule plays in S. aureus infection.


Asunto(s)
Genes MHC Clase II , Neumonía Estafilocócica , Ratones , Humanos , Animales , Cadenas alfa de HLA-DR/farmacología , Staphylococcus aureus , Neumonía Estafilocócica/genética , Linfocitos T CD8-positivos , Ratones Endogámicos C57BL , Ratones Transgénicos
2.
Exp Cell Res ; 422(1): 113415, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36379277

RESUMEN

Interferon-inducible protein 204 (IFI204) is an intracellular DNA receptor that can recognize DNA viruses and intracellular bacteria. Extracellular traps (ETs) have been recognized as an indispensable antimicrobial barrier that play an indispensable role in bacterial, fungal, parasitic, and viral infections. However, how ETs form and the mechanisms by which IFI204 function in Staphylococcus aureus pneumonia are still unclear. Moreover, by in vitro experiments, we proved that IFI204 deficiency decreases the formation of ETs induced by Staphylococcus aureus in a NOX-independent manner. More importantly, Deoxyribonuclease I (DNase I) treatment significantly inhibited the formation of ETs. IFI204 contributed to ETs formation by promoting citrullination of histone H3 and the expression of PAD4 (peptidylarginine deiminase 4). Altogether, these findings highlight the potential importance of IFI204 for host defense against S. aureus USA300-TCH1516 infection.


Asunto(s)
Trampas Extracelulares , Neumonía Estafilocócica , Trampas Extracelulares/genética , Trampas Extracelulares/metabolismo , Interferones/metabolismo , Neutrófilos/metabolismo , Neumonía Estafilocócica/genética , Neumonía Estafilocócica/metabolismo , Staphylococcus aureus , Ratones , Animales
3.
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
4.
Pak J Pharm Sci ; 34(2): 493-498, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34275821

RESUMEN

P. petiolosa as a typical Chinese herbal medicine has been generally utilized as Chinese native medicine formulation for treatment of chronic bronchitis, bronchial asthma and pneumoconiosis. The objective of this study was to evaluate the anti-inflammatory and antibacterial activities of P. petiolosa ethyl acetate extract (PPEAE) against S. aureusin mice. In our study, mice were infected pneumonia by S. aureus, colonization of S. aureus in lung tissue was calculated and the number of white blood cells (WBC) in blood was measured. Meanwhile, the hematoxylin-eosin staining (H&E) was observed and the Real-time PCR was employed to determine the relative mRNA expression. The results showed that, after treated with PPEAE the wet/dry (W/D) weight ratio and the number of WBC decreased dramatically, the number of S. aureus was significantly reduced. Furthermore, H&E staining showed that PPEAE obviously relieved the inflammation of infected mice and real-time PCR results indicated that PPEAE significantly down regulated the inflammatory iNOS, TNF-α and up regulated the anti-inflammatory HO-1 mRNA. In summary, our study revealed that application of crude product PPEAE had prominent antibacterial activity against S. aureus. PPEAE significantly reduced the biomass of S. aureus and effectively relieved the inflammation of S. aureus-induced pneumonia.


Asunto(s)
Antibacterianos/farmacología , Antiinflamatorios/farmacología , Medicamentos Herbarios Chinos/farmacología , Pulmón/efectos de los fármacos , Extractos Vegetales/farmacología , Neumonía Estafilocócica/genética , Polypodiaceae , Staphylococcus aureus/efectos de los fármacos , Animales , Hemo-Oxigenasa 1/efectos de los fármacos , Hemo-Oxigenasa 1/genética , Inflamación/genética , Inflamación/metabolismo , Pulmón/metabolismo , Pulmón/microbiología , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/genética , Ratones , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/genética , Neumonía Estafilocócica/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética
5.
Sci Rep ; 11(1): 4837, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33649379

RESUMEN

Acute chest syndrome (ACS) is the most serious complication of sickle cell disease. The pathophysiology of ACS may involve lower respiratory tract infection (LRTI), alveolar hypoventilation and atelectasis, bone infarcts-driven fat embolism, and in situ pulmonary artery thrombosis. One of the most challenging issues for the physicians is to diagnose LRTI as the cause of ACS. The use of a respiratory multiplex PCR (mPCR) for the diagnosis of LRTI has not been assessed in sickle-cell adult patients with ACS. To describe the spectrum of infectious aetiologies of severe ACS, using a diagnostic approach combining conventional tests and mPCR. A non-interventional monocenter prospective study involving all the consecutive sickle-cell adult patients with ACS admitted to the intensive care unit (ICU). Microbiological investigation included conventional tests and a nasopharyngeal swab for mPCR. Altogether, 36 patients were enrolled, of whom 30 (83%) had complete microbiological investigations. A bacterial microorganism, mostly Staphylococcus aureus (n = 8), was identified in 11 patients. There was no pneumonia-associated intracellular bacterial pathogen. A respiratory virus was identified in six patients. Using both conventional tests and nasopharyngeal mPCR, a microbiological documentation was obtained in half of adult ACS patients admitted to the ICU. Pyogenic bacteria, especially S. aureus, predominated.


Asunto(s)
Síndrome Torácico Agudo , Reacción en Cadena de la Polimerasa Multiplex , Neumonía Estafilocócica , Staphylococcus aureus/genética , Síndrome Torácico Agudo/complicaciones , Síndrome Torácico Agudo/genética , Síndrome Torácico Agudo/microbiología , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neumonía Estafilocócica/etiología , Neumonía Estafilocócica/genética , Neumonía Estafilocócica/microbiología , Estudios Prospectivos
6.
Toxins (Basel) ; 12(11)2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33218049

RESUMEN

Pneumonia is an acute pulmonary infection associated with high mortality and an immense financial burden on healthcare systems. Staphylococcus aureus is an opportunistic pathogen capable of inducing S. aureus pneumonia (SAP), with some lineages also showing multidrug resistance. Given the high level of antibiotic resistance, much research has been focused on targeting S. aureus virulence factors, including toxins and biofilm-associated proteins, in an attempt to develop effective SAP therapeutics. Despite several promising leads, many hurdles still remain for S. aureus vaccine research. Here, we review the state-of-the-art SAP therapeutics, highlight their pitfalls, and discuss alternative approaches of potential significance and future perspectives.


Asunto(s)
Neumonía Estafilocócica/terapia , Staphylococcus aureus , Factores de Virulencia , Animales , Vacunas Bacterianas/uso terapéutico , Biopelículas , Genómica , Humanos , Metabolómica , Neumonía Estafilocócica/genética , Neumonía Estafilocócica/metabolismo , Staphylococcus aureus/patogenicidad , Staphylococcus aureus/fisiología
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.
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
9.
Artículo en Inglés | MEDLINE | ID: mdl-31235625

RESUMEN

The use of macrolides against pneumonia has been reported to improve survival; however, little is known about their efficacy against methicillin-resistant Staphylococcus aureus (MRSA) pneumonia. In this study, we investigated the effect of azithromycin (AZM) and compared it with that of vancomycin (VCM) and daptomycin (DAP) in a murine model of MRSA pneumonia. Mice were infected with MRSA by intratracheal injection and then treated with AZM, VCM, or DAP. The therapeutic effect of AZM, in combination or not with the other drugs, was compared in vivo, whereas the effect of AZM on MRSA growth and toxin mRNA expression was evaluated in vitro. In vivo, the AZM-treated group showed significantly longer survival and fewer bacteria in the lungs 24 h after infection than the untreated group, as well as the other anti-MRSA drug groups. No significant decrease in cytokine levels (interleukin-6 [IL-6] and macrophage inflammatory protein-2 [MIP-2]) in bronchoalveolar lavage fluid or toxin expression levels (α-hemolysin [Hla] and staphylococcal protein A [Spa]) was observed following AZM treatment. In vitro, AZM suppressed the growth of MRSA in late log phase but not in stationary phase. No suppressive effect against toxin production was observed following AZM treatment in vitro In conclusion, contrary to the situation in vitro, AZM was effective against MRSA growth in vivo in our pneumonia model, substantially improving survival. The suppressive effect on MRSA growth at the initial stage of pneumonia could underlie the potential mechanism of AZM action against MRSA pneumonia.


Asunto(s)
Antibacterianos/farmacología , Azitromicina/farmacología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Neumonía Estafilocócica/tratamiento farmacológico , Animales , Profilaxis Antibiótica , Toxinas Bacterianas/genética , Daptomicina/farmacología , Modelos Animales de Enfermedad , Quimioterapia Combinada , Neumonía Asociada a la Atención Médica/tratamiento farmacológico , Neumonía Asociada a la Atención Médica/genética , Neumonía Asociada a la Atención Médica/microbiología , Neumonía Asociada a la Atención Médica/prevención & control , Proteínas Hemolisinas/genética , Masculino , Ratones Endogámicos BALB C , Neumonía Estafilocócica/genética , Neumonía Estafilocócica/microbiología , Neumonía Estafilocócica/prevención & control , Vancomicina/farmacología , Factores de Virulencia/genética
10.
Anal Biochem ; 550: 54-60, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29678761

RESUMEN

Pneumonia is one of the most common and potentially lethal infectious conditions worldwide. Streptococcus pneumoniae is the pathogen most frequently associated with bacterial community-acquired pneumonia, while Legionella pneumophila is the major cause for local outbreaks of legionellosis. Both pathogens can be difficult to diagnose since signs and symptoms are nonspecific and do not differ from other causes of pneumonia. Therefore, a rapid diagnosis within a clinically relevant time is essential for a fast onset of the proper treatment. Although methods based on polymerase chain reaction significantly improved the identification of pathogens, they are difficult to conduct and need specialized equipment. We describe a rapid and sensitive test using isothermal recombinase polymerase amplification and detection on a disposable test strip. This method does not require any special instrumentation and can be performed in less than 20 min. The analytical sensitivity in the multiplex assay amplifying specific regions of S. pneumoniae and L. pneumophila simultaneously was 10 CFUs of genomic DNA per reaction. In cross detection studies with closely related strains and other bacterial agents the specificity of the RPA was confirmed. The presented method is applicable for near patient and field testing with a rather simple routine and the possibility for a read out with the naked eye.


Asunto(s)
ADN Bacteriano/genética , Legionella pneumophila/genética , Enfermedad de los Legionarios , Reacción en Cadena de la Polimerasa Multiplex/métodos , Neumonía Estafilocócica , Staphylococcus aureus/genética , Humanos , Enfermedad de los Legionarios/diagnóstico , Enfermedad de los Legionarios/genética , Neumonía Estafilocócica/diagnóstico , Neumonía Estafilocócica/genética
11.
J Infect Dis ; 218(5): 809-813, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-29668950

RESUMEN

Staphylococcus aureus is a predominant cause of fatal pneumonia following influenza A virus (IAV) infection. Herein we investigate the influence of antecedent IAV infection on S. aureus virulence gene expression. Using a murine model, comparing the USA300 and USA300ΔsaeR/S strains, we demonstrate that S. aureus pathogenesis following IAV infection is SaeR/S dependent. Furthermore, we show that IAV modulates the lung environment to rapidly up-regulate S. aureus virulence factors containing the SaeR-binding domain. Data demonstrate that the pathogen response to IAV infection impacts host outcome and provides evidence that the ability of S. aureus to sense and respond to the lung environment determines severity of pneumonia.


Asunto(s)
Proteínas Bacterianas/metabolismo , Infecciones por Orthomyxoviridae/complicaciones , Neumonía Estafilocócica/inmunología , Proteínas Quinasas/metabolismo , Staphylococcus aureus/patogenicidad , Factores de Transcripción/metabolismo , Animales , Proteínas Bacterianas/genética , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Masculino , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/patología , Neumonía Estafilocócica/genética , Proteínas Quinasas/genética , Staphylococcus aureus/genética , Factores de Transcripción/genética
12.
Artículo en Inglés | MEDLINE | ID: mdl-28480385

RESUMEN

BACKGROUND: Alcohol extract from the root of Urena lobata L. (ULL) had broad spectrum antimicrobial activity. Studies in vitro have sho that ULL aqueous extract has antibacterial effect on S. aureusis, and the combination therapy of the ULL aqueous extract with cefazolin sodium showed additive effect. MATERIALS AND METHODS: The mice underwent nasal inhalation with S. aureus, a subset of mice were intra-gastric gavage with ULL and/or intravenous injection cefazolin sodium twice daily. After being exposed to S. aureus for 5 days, 10 days and 14 days respectively, the white blood cells count (WBC), neutrophils absolute value (NEU) and the neutrophil percentage (NEU%) in peripheral blood, as well as the levels of serum immunoglobulin (Ig) G and IgM were determined using commercial kits. The colony count of S. aureus, the levels of interleukin (IL) -6 and IL-10 of mice lung tissue were detected, and the pathological changes of lung tissue were examined using H & E staining. RESULTS: ULL significantly protected against S. aureus pneumonia, as evidenced by the remarkable decrease in the rate of S. aureus colony count/lung weight, WBC, NEU and NEU% in peripheral blood, as well as the attenuation of lung histopathological damage. Additionally, ULL+cefazolin could have markedly reduced the rate of S. aureus colony count/lung weight when compared with cefazolin. Furthermore, ULL and ULL+cefazolin both could significantly decrease the serum levels of IgG and IgM, and the levels of IL-6, IL-10 in mice lung tissue. CONCLUSION: This study first demonstrated that ULL may have potential use as a therapeutic agent for S. aureus pneumonia, and the roles of IgG, IgM, IL-6 and IL-10 in ULL protection against S. aureus pneumonia remain to be further studied.


Asunto(s)
Antibacterianos/administración & dosificación , Medicamentos Herbarios Chinos/administración & dosificación , Malvaceae/química , Neumonía Estafilocócica/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos , Animales , China , Femenino , Humanos , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Masculino , Ratones , Neumonía Estafilocócica/genética , Neumonía Estafilocócica/inmunología , Neumonía Estafilocócica/microbiología , Staphylococcus aureus/fisiología
13.
Mol Ther ; 24(10): 1873-1880, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27456061

RESUMEN

Chronic granulomatous disease (CGD) is a primary immunodeficiency due to a deficiency in one of the subunits of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex. CGD patients are characterized by an increased susceptibility to bacterial and fungal infections, and to granuloma formation due to the excessive inflammatory responses. Several gene therapy approaches with lentiviral vectors have been proposed but there is a lack of in vivo data on the ability to control infections and inflammation. We set up a mouse model of acute infection that closely mimic the airway infection in CGD patients. It involved an intratracheal injection of a methicillin-sensitive reference strain of S. aureus. Gene therapy, with hematopoietic stem cells transduced with regulated lentiviral vectors, restored the functional activity of NADPH oxidase complex (with 20-98% of dihydrorhodamine positive granulocytes and monocytes) and saved mice from death caused by S. aureus, significantly reducing the bacterial load and lung damage, similarly to WT mice even at low vector copy number. When challenged, gene therapy-treated XCGD mice showed correction of proinflammatory cytokines and chemokine imbalance at levels that were comparable to WT. Examined together, our results support the clinical development of gene therapy protocols using lentiviral vectors for the protection against infections and inflammation.


Asunto(s)
Terapia Genética/métodos , Enfermedad Granulomatosa Crónica/complicaciones , Trasplante de Células Madre Hematopoyéticas/métodos , Glicoproteínas de Membrana/genética , NADPH Oxidasas/genética , Neumonía Estafilocócica/terapia , Staphylococcus aureus/fisiología , Animales , Carga Bacteriana , Células Cultivadas , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Vectores Genéticos/administración & dosificación , Enfermedad Granulomatosa Crónica/genética , Células Madre Hematopoyéticas/virología , Humanos , Lentivirus/genética , Ratones , NADPH Oxidasa 2 , Neumonía Estafilocócica/genética , Neumonía Estafilocócica/microbiología
14.
Sci Signal ; 9(418): ra26, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26956485

RESUMEN

In phagocytes, pathogen recognition is followed by Ca(2+) mobilization and NADPH oxidase 2 (NOX2)-mediated "oxidative burst," which involves the rapid production of large amounts of reactive oxygen species (ROS). We showed that ORAI Ca(2+) channels control store-operated Ca(2+) entry, ROS production, and bacterial killing in primary human monocytes. ROS inactivate ORAI channels that lack an ORAI3 subunit. Staphylococcal infection of mice reduced the expression of the gene encoding the redox-sensitive Orai1 and increased the expression of the gene encoding the redox-insensitive Orai3 in the lungs or in bronchoalveolar lavages. A similar switch from ORAI1 to ORAI3 occurred in primary human monocytes exposed to bacterial peptides in culture. These alterations in ORAI1 and ORAI3 abundance shifted the channel assembly toward a more redox-insensitive configuration. Accordingly, silencing ORAI3 increased the redox sensitivity of the channel and enhanced oxidation-induced inhibition of NOX2. We generated a mathematical model that predicted additional features of the Ca(2+)-redox interplay. Our results identified the ORAI-NOX2 feedback loop as a determinant of monocyte immune responses.


Asunto(s)
Canales de Calcio Activados por la Liberación de Calcio/inmunología , Calcio/inmunología , Modelos Biológicos , Monocitos/inmunología , Neumonía Estafilocócica/inmunología , Especies Reactivas de Oxígeno/inmunología , Staphylococcus aureus/inmunología , Animales , Calcio/metabolismo , Canales de Calcio Activados por la Liberación de Calcio/genética , Canales de Calcio Activados por la Liberación de Calcio/metabolismo , Femenino , Humanos , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Ratones , Monocitos/metabolismo , Monocitos/patología , NADPH Oxidasa 2 , NADPH Oxidasas/genética , NADPH Oxidasas/inmunología , NADPH Oxidasas/metabolismo , Oxidación-Reducción , Neumonía Estafilocócica/genética , Neumonía Estafilocócica/metabolismo , Neumonía Estafilocócica/patología , Especies Reactivas de Oxígeno/metabolismo , Staphylococcus aureus/metabolismo
15.
Shock ; 45(4): 375-84, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26863117

RESUMEN

Staphylococcus aureus is a common cause of nosocomial pneumonia frequently resulting in acute respiratory distress syndrome (ARDS). Surfactant protein B (SP-B) gene expresses two proteins involved in lowering surface tension and host defense. Genotyping studies demonstrate a significant association between human SP-B genetic variants and ARDS. Curcumins have been shown to attenuate host inflammation in many sepsis models. Our hypothesis is that functional differences of SP-B variants and treatment with curcumin (CMC2.24) modulate lung injury in bacterial pneumonia. Humanized transgenic mice, expressing either SP-B T or C allele without mouse SP-B gene, were used. Bioluminescent labeled S. aureus Xen 36 (50 µL) was injected intratracheally to cause pneumonia. Infected mice received daily CMC2.24 (40 mg/kg) or vehicle alone by oral gavage. Dynamic changes of bacteria were monitored using in vivo imaging system. Histological, cellular, and molecular indices of lung injury were studied in infected mice 48 h after infection. In vivo imaging analysis revealed total flux (bacterial number) was higher in the lung of infected SP-B-C mice compared with infected SP-B-T mice (P < 0.05). Infected SP-B-C mice demonstrated increased mortality, lung injury, apoptosis, and NF-κB expression compared with infected SP-B-T mice. Compared with controls, CMC2.24 treatment significantly reduced the following: mortality, total bacterial flux and lung tissue apoptosis, inflammatory cells, NF-κB expression (P < 0.05), and MMPs-2, -9, -12 activities (P < 0.05). We conclude that mice with SP-B-C allele are more susceptible to S. aureus pneumonia than mice with SP-B-T allele, and that CMC2.24 attenuates lung injury thus reducing mortality.


Asunto(s)
Curcumina/farmacología , Predisposición Genética a la Enfermedad , Variación Genética , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/genética , Neumonía Estafilocócica/tratamiento farmacológico , Neumonía Estafilocócica/genética , Proteína B Asociada a Surfactante Pulmonar/genética , Staphylococcus aureus , Alelos , Animales , Humanos , Lesión Pulmonar/metabolismo , Ratones , Ratones Transgénicos , Neumonía Estafilocócica/metabolismo , Proteína B Asociada a Surfactante Pulmonar/metabolismo
16.
Shock ; 46(2): 164-72, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26849628

RESUMEN

UNLABELLED: Staphylococcus aureus pneumonia is an important cause of sepsis which causes gut injury, inflammation, and apoptosis. The surfactant proteins surfactant protein A (SP-A) and surfactant protein D (SP-D) bind bacterial pathogens and facilitate clearance of pathogens, apoptotic bodies, and modulate immune responses. SP-A and SP-D are expressed in both lung and gut epithelia. We hypothesize SP-A and SP-D regulate pneumonia severity and gut injury during pneumonia. METHODS: Wild-type (WT) and SP-A and SP-D double knockout (SP-A/D KO) mice were subjected to S. aureus or sham pneumonia. Bronchoalveolar lavage and tissue harvest were performed 24 h later. Pneumonia severity, gut mucosal injury, inflammation, and apoptosis were measured using a combination of histology, immunohistochemistry, cytokine assay, TUNEL assay, quantitative real-time polymerase chain reaction, and Western blot analyses. RESULTS: Pneumonia increased gut inflammation, apoptosis, and mucosal injury in both groups. Pneumonia histology and bacterial growth in bronchoalveolar lavage fluid demonstrate more severe infection in SP-A/D KO mice compared with WT controls. SP-A/D KO mice with pneumonia also demonstrate more severe histologic gut mucosal injury, increased gut apoptosis, elevated caspase-3 levels, and Bax/Bcl-2 mRNA expression compared with WT pneumonia mice. Nuclear factor κB (NF-κB) p65 expression and its nuclear translocation, gut levels of tumor necrosis factor α and interleukin-1ß were all increased in SP-A/D KO mice with pneumonia compared with WT controls. CONCLUSIONS: These data provide evidence SP-A and SP-D attenuate S. aureus pneumonia severity resulting in decreased intestinal mucosal injury, apoptosis, and inflammation. Improved pulmonary clearance of S. aureus decreased caspase-3 and Bax/Bcl-2 expressions and decreased activation of the NF-κB signaling pathway in intestine represent potential mechanisms for the effects of SP-A and SP-D on gut injury during pneumonia.


Asunto(s)
Neumonía Estafilocócica/inmunología , Neumonía Estafilocócica/patología , Proteína A Asociada a Surfactante Pulmonar/metabolismo , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Animales , Apoptosis/fisiología , Líquido del Lavado Bronquioalveolar/microbiología , Caspasa 3/genética , Caspasa 3/metabolismo , Modelos Animales de Enfermedad , Femenino , Intestinos/citología , Intestinos/inmunología , Pulmón/citología , Pulmón/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía Estafilocócica/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína A Asociada a Surfactante Pulmonar/genética , Proteína D Asociada a Surfactante Pulmonar/genética , Transducción de Señal , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
18.
Respir Res ; 16: 10, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25651926

RESUMEN

BACKGROUND: Influenza is a common respiratory virus and Staphylococcus aureus frequently causes secondary pneumonia during influenza infection, leading to increased morbidity and mortality. Influenza has been found to attenuate subsequent Type 17 immunity, enhancing susceptibility to secondary bacterial infections. IL-27 is known to inhibit Type 17 immunity, suggesting a potential critical role for IL-27 in viral and bacterial co-infection. METHODS: A murine model of influenza and Staphylococcus aureus infection was used to mimic human viral, bacterial co-infection. C57BL/6 wild-type, IL-27 receptor α knock-out, and IL-10 knock-out mice were infected with Influenza H1N1 (A/PR/8/34) or vehicle for 6 days followed by challenge with Staphylococcus aureus or vehicle for 24 hours. Lung inflammation, bacterial burden, gene expression, and cytokine production were determined. RESULTS: IL-27 receptor α knock-out mice challenged with influenza A had increased morbidity compared to controls, but no change in viral burden. IL-27 receptor α knock-out mice infected with influenza displayed significantly decreased IL-10 production compared to wild-type. IL-27 receptor α knock-out mice co-infected with influenza and S. aureus had improved bacterial clearance compared to wild-type controls. Importantly, there were significantly increased Type 17 responses and decreased IL-10 production in IL-27 receptor α knock-out mice. Dual infected IL-10-/- mice had significantly less bacterial burden compared to dual infected WT mice. CONCLUSIONS: These data reveal that IL-27 regulates enhanced susceptibility to S. aureus pneumonia following influenza infection, potentially through the induction of IL-10 and suppression of IL-17.


Asunto(s)
Coinfección , Subtipo H1N1 del Virus de la Influenza A/inmunología , Interleucinas/inmunología , Pulmón/inmunología , Infecciones por Orthomyxoviridae/inmunología , Neumonía Estafilocócica/inmunología , Staphylococcus aureus/inmunología , Animales , Carga Bacteriana , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno , Inmunidad Celular , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Interleucina-10/deficiencia , Interleucina-10/genética , Interleucina-17/inmunología , Pulmón/microbiología , Pulmón/virología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/virología , Neumonía Estafilocócica/genética , Neumonía Estafilocócica/microbiología , Receptores de Citocinas/deficiencia , Receptores de Citocinas/genética , Receptores de Interleucina , Staphylococcus aureus/patogenicidad , Células Th17/inmunología , Células Th17/microbiología , Células Th17/virología , Factores de Tiempo , Carga Viral
19.
Am J Respir Cell Mol Biol ; 52(1): 14-24, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24918472

RESUMEN

Treatment of primary or immortalized human airway epithelial cells (16HBE14o-, S9) or alveolar cancer cells (A549) with recombinant hemolysin A (rHla), a major virulence-associated factor of Staphylococcus aureus, induces alterations in cell shape and formation of paracellular gaps in the cell layer. Semiquantitative Western blotting using extracts of freshly isolated airway tissue (nasal epithelium) or 16HBE14o- model cells revealed that phosphorylation levels of focal adhesion kinase (Fak) and paxillin were altered upon treatment of tissue or cells with rHla. Immune fluorescence analyses showed that rHla treatment of 16HBE14o- cells results in losses of vinculin and paxillin from focal contacts and a net reduction in the number of focal contacts. The actin cytoskeleton was strongly remodeled. We concluded that treatment of cells with rHla activates Fak signaling, which accelerates focal contact turnover and prevents newly formed focal contacts (focal complexes) from maturation to focal adhesions. The inability of rHla-treated cells to form stable focal adhesions may be one factor that contributes to gap formation in the cell layer. In vivo, such changes may disturb the defensive barrier function of the airway epithelium and may facilitate lung infections by S. aureus.


Asunto(s)
Células Epiteliales/metabolismo , Matriz Extracelular/metabolismo , Neumonía Estafilocócica/metabolismo , Mucosa Respiratoria/metabolismo , Staphylococcus aureus/metabolismo , Toxinas Bacterianas/genética , Adhesión Celular/genética , Línea Celular , Células Epiteliales/patología , Matriz Extracelular/genética , Femenino , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Adhesiones Focales/genética , Adhesiones Focales/metabolismo , Adhesiones Focales/patología , Proteínas Hemolisinas/genética , Humanos , Masculino , Paxillin/genética , Paxillin/metabolismo , Fosforilación/genética , Neumonía Estafilocócica/genética , Neumonía Estafilocócica/patología , Mucosa Respiratoria/patología , Transducción de Señal/genética , Staphylococcus aureus/genética , Vinculina/genética , Vinculina/metabolismo
20.
Mucosal Immunol ; 7(6): 1366-74, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24736233

RESUMEN

Staphylococcus aureus is a major cause of severe pneumonia. Multiple mechanisms of proinflammatory signaling are activated to recruit immune cells into the airway in response to S. aureus. We found that interleukin-16 (IL-16), a T cell cytokine that binds CD4, is potently activated by S. aureus, specifically by protein A (SpA), and to a much greater extent than by Gram-negative pathogens or lipopolysaccharide. IL-16 production involved multiple signals including ligation of tumor necrosis factor receptor (TNFR) family members or epidermal growth factor receptor, both receptors for SpA and generation of Ca(2+) fluxes to activate calpains and caspase-3. Although human airway epithelial cells, vascular endothelial cells, THP-1 and Jurkat T cells released IL-16 in response to S. aureus in vitro, in a murine model of pneumonia, CD4(+) cells were the major source of IL-16 suggesting the involvement of an autocrine signaling pathway. The production of IL-16 contributed to lung damage as neutralization of IL-16 enhanced S. aureus clearance and resulted in diminished lung pathology in S. aureus pneumonia. Our results suggest that the ability of S. aureus to activate TNFR1 and Ca(2+)/calpain signaling contribute to T cell activation and excessive inflammation in the setting of acute pneumonia.


Asunto(s)
Señalización del Calcio/inmunología , Calpaína/inmunología , Caspasas/inmunología , Interleucina-16/inmunología , Staphylococcus aureus Resistente a Meticilina/inmunología , Neumonía Estafilocócica/inmunología , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Mucosa Respiratoria/inmunología , Enfermedad Aguda , Animales , Señalización del Calcio/genética , Calpaína/genética , Caspasas/genética , Humanos , Interleucina-16/genética , Interleucina-16/metabolismo , Ratones , Ratones Noqueados , Neumonía Estafilocócica/genética , Neumonía Estafilocócica/metabolismo , Neumonía Estafilocócica/patología , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA