Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Med Virol ; 95(1): e28203, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36217277

RESUMEN

Inducing early apoptosis in alveolar macrophages is one of the strategies influenza A virus (IAV) evolved to subvert host immunity. Correspondingly, the host mitochondrial protein nucleotide-binding oligomerization domain-like receptor (NLR)X1 is reported to interact with virus polymerase basic protein 1-frame 2 (PB1-F2) accessory protein to counteract virus-induced apoptosis. Herein, we report that one of the F-box proteins, FBXO6, promotes proteasomal degradation of NLRX1, and thus facilitates IAV-induced alveolar macrophages apoptosis and modulates both macrophage survival and type I interferon (IFN) signaling. We observed that FBXO6-deficient mice infected with IAV exhibited decreased pulmonary viral replication, alleviated inflammatory-associated pulmonary dysfunction, and less mortality. Analysis of the lungs of IAV-infected mice revealed markedly reduced leukocyte recruitment but enhanced production of type I IFN in Fbxo6-/- mice. Furthermore, increased type I IFN production and decreased viral replication were recapitulated in FBXO6 knockdown macrophages and associated with reduced apoptosis. Through gain- and loss-of-function studies, we found lung resident macrophages but not bone marrow-derived macrophages play a key role in the differences FBXO6 signaling pathway brings in the antiviral immune response. In further investigation, we identified that FBXO6 interacted with and promoted the proteasomal degradation of NLRX1. Together, our results demonstrate that FBXO6 negatively regulates immunity against IAV infection by enhancing the degradation of NLRX1 and thus impairs the survival of alveolar macrophages and antiviral immunity of the host.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Interferón Tipo I , Infecciones por Orthomyxoviridae , Ratones , Animales , Humanos , Macrófagos Alveolares/metabolismo , Antivirales/metabolismo , Macrófagos , Interferón Tipo I/metabolismo , Replicación Viral/fisiología , Inmunidad , Proteínas Mitocondriales/metabolismo
2.
Am J Respir Cell Mol Biol ; 65(1): 30-40, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33761305

RESUMEN

Acute respiratory infections caused by influenza A virus (IAV) spread widely and lead to substantial morbidity and mortality. Host cell induction of type I interferon (IFN-I) plays a fundamental role in eliminating the virus during the innate antiviral response. The potential role of N-myc and STAT interactor (NMI) and its underlying mechanisms of action during IAV infection, however, remain elusive. In this study, we found that the expression of NMI increased after IAV infection. Nmi-knockout mice infected with IAV displayed increased survival rate, decreased weight loss, lower viral replication, and attenuated lung inflammation when compared with wild-type mice. Deficiency of NMI promoted the production of IFN-I and IFN-stimulated genes in vivo and in vitro. Reduced levels of NMI also resulted in an increase of the expression of IFN regulator factor (IRF) 7. Further studies have revealed that NMI could interact with IRF7 after IAV infection, and this interaction involved its NID1 and NID2 domain. In addition, NMI facilitated ubiquitination and proteasome-dependent degradation of IRF7 through recruitment of the E3 ubiquitin ligase TRIM21 (tripartite motif-containing 21) to limit the IAV-triggered innate immunity. Our findings reveal a clearer understanding of the role of NMI in regulating the host innate antiviral response and provide a potential therapeutic target for controlling IAV infection.


Asunto(s)
Inmunidad Innata , Virus de la Influenza A/inmunología , Factor 7 Regulador del Interferón/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Infecciones por Orthomyxoviridae/inmunología , Proteolisis , Ribonucleoproteínas/inmunología , Células A549 , Animales , Perros , Células HEK293 , Humanos , Factor 7 Regulador del Interferón/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Células de Riñón Canino Madin Darby , Ratones , Ratones Noqueados , Infecciones por Orthomyxoviridae/genética , Ribonucleoproteínas/genética
3.
BMC Infect Dis ; 21(1): 1267, 2021 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-34930151

RESUMEN

BACKGROUND: Streptococcus constellatus is a member of Streptococcus anginosus group (SAG) that tends to cause pyogenic infections in various sites. However, Streptococcus constellatus is easily ignored by routine clinical laboratory tests for its prolonged anaerobic culture environment. CASE PRESENTATION: A 71-year-old man was admitted to our hospital due to productive cough, fever, chest pain and shortness of breath for 3 weeks. Chest computed tomography showed patchy opacities and right-sided pleural effusion, so a chest tube was inserted and purulent and hemorrhagic fluid was aspirated. The routine etiological examinations of the pleural effusion were all negative, and next-generation sequencing (NGS) detected Streptococcus constellatus. Intravenous piperacillin-tazobactam 4.5 g every 8 h was used accordingly. The patient recovered and subsequent chest computed tomography confirmed the improvement. CONCLUSIONS: We reported a case of empyema secondary to Streptococcus constellatus infection, which was identified by NGS, instead of bacterial culture. This case highlights the utility of NGS in detecting pathogens negative in traditional bacterial tests.


Asunto(s)
Empiema , Infecciones Estreptocócicas , Streptococcus constellatus , Anciano , Empiema/diagnóstico , Fiebre , Humanos , Laboratorios Clínicos , Masculino , Infecciones Estreptocócicas/diagnóstico , Infecciones Estreptocócicas/tratamiento farmacológico
4.
J Cell Mol Med ; 24(2): 1614-1625, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31829519

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a risk factor for the development of lung cancer. The aim of this study was to identify early diagnosis biomarkers for lung squamous cell carcinoma (SQCC) in COPD patients and to determine the potential pathogenetic mechanisms. The GSE12472 data set was downloaded from the Gene Expression Omnibus database. Differentially co-expressed links (DLs) and differentially expressed genes (DEGs) in both COPD and normal tissues, or in both SQCC + COPD and COPD samples were used to construct a dynamic network associated with high-risk genes for the SQCC pathogenetic process. Enrichment analysis was performed based on Gene Ontology annotations and Kyoto Encyclopedia of Genes and Genomes pathway analysis. We used the gene expression data and the clinical information to identify the co-expression modules based on weighted gene co-expression network analysis (WGCNA). In total, 205 dynamic DEGs, 5034 DLs and one pathway including CDKN1A, TP53, RB1 and MYC were found to have correlations with the pathogenetic progress. The pathogenetic mechanisms shared by both SQCC and COPD are closely related to oxidative stress, the immune response and infection. WGCNA identified 11 co-expression modules, where magenta and black were correlated with the "time to distant metastasis." And the "surgery due to" was closely related to the brown and blue modules. In conclusion, a pathway that includes TP53, CDKN1A, RB1 and MYC may play a vital role in driving COPD towards SQCC. Inflammatory processes and the immune response participate in COPD-related carcinogenesis.


Asunto(s)
Carcinoma de Células Escamosas/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Enfermedad Pulmonar Obstructiva Crónica/genética , Fumar/efectos adversos , Fumar/genética , Análisis por Conglomerados , Ontología de Genes , Redes Reguladoras de Genes , Humanos , Modelos Biológicos , Mapas de Interacción de Proteínas/genética , Reproducibilidad de los Resultados
5.
Artículo en Inglés | MEDLINE | ID: mdl-30509940

RESUMEN

While the inflammatory response to severe pneumonia is paramount in limiting and resolving the infection, excessive inflammation can lead to deleterious effects. We theorized that patients with severe community-acquired pneumonia (CAP) who were treated with macrolides and aspirin would receive benefit beyond that of conventional antibiotic therapy. An observational study was conducted with patients with severe CAP. All patients were admitted to 5 teaching hospitals (in Italy, the United States, Japan, and China), and data were gathered from their electronic medical records. Severe pneumonia was defined according to Infectious Diseases Society of America/American Thoracic Society criteria. Patients were divided into 4 groups, i.e., (i) the aspirin-only group (ASG), (ii) the macrolide-only group (MG), (iii) the aspirin plus macrolide group (ASMG), or (iv) the neither aspirin nor macrolide group (NASMG). Survival rates for the 4 groups were evaluated after adjustment for confounders and after weighting by propensity score. A total of 1,295 patients were included in the analysis. There were 237 patients (18.3%) in the ASG, 294 (22.7%) in the MG, 148 (11.4%) in the ASMG, and 616 (47.6%) in the NASMG. The mortality rate at 30 days was 15.5% in the ASMG, compared to 28.2% in the NASMG, 23.8% in the MG, and 21.1% in the ASG. After propensity score analysis, receipt of aspirin plus macrolide (hazard ratio, 0.71 [95% confidence interval, 0.58 to 0.88]; P = 0.002) was associated with a higher 30-day survival rate. This is a hypothesis-generating study in which data suggest that the combination of aspirin plus a macrolide improves 30-day survival rates for patients with severe CAP. Further randomized studies will need to be undertaken to confirm this phenomenon.


Asunto(s)
Antibacterianos/uso terapéutico , Antiinflamatorios no Esteroideos/uso terapéutico , Aspirina/uso terapéutico , Infecciones Comunitarias Adquiridas/tratamiento farmacológico , Macrólidos/uso terapéutico , Neumonía Bacteriana/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , China , Infecciones Comunitarias Adquiridas/mortalidad , Quimioterapia Combinada , Femenino , Humanos , Italia , Japón , Masculino , Neumonía Bacteriana/mortalidad , Tasa de Supervivencia , Resultado del Tratamiento , Estados Unidos
6.
J Infect Dis ; 218(1): 64-74, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29741644

RESUMEN

Background: Mast cells (MCs) play a key role in immune process response to invading pathogens. Methods: This study assessed the involvement of MCs in controlling Staphylococcus aureus infection in a cutaneous infection model of MC-deficient (KitW-sh/W-sh) mice. Results: KitW-sh/W-sh mice developed significantly larger skin lesions after the cutaneous S. aureus challenge, when compared to wild-type (WT) mice, while MC dysfunction reduced the inflammation response to S. aureus. The levels of tumor necrosis factor (TNF)-α in skin tissues were significantly decreased in KitW-sh/W-sh mice upon infection. Moreover, the exogenous administration of MCs or recombinant TNF-α effectively restored the immune response against S. aureus in KitW-sh/W-sh mice via the recruitment of neutrophils to the infected site. These results indicate that the effects of MC deficiency are largely attributed to the decrease in production of TNF-α in cutaneous S. aureus infection. In addition, S. aureus-induced MC activation was dependent on the c-kit receptor-activated phosphoinositide 3-kinase (PI3K)/AKT/P65-nuclear factor (NF-κB) pathway, which was confirmed by treatment with Masitinib (a c-kit receptor inhibitor), Wortmannin (a PI3K inhibitor), and pyrrolidine dithiocarbamate (a NF-κB inhibitor), respectively. Conclusions: The present study identifies the critical role of MCs in the host defense against S. aureus infection.


Asunto(s)
Mastocitos/inmunología , Infecciones Cutáneas Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones Cutáneas Estafilocócicas/patología
7.
J Cell Mol Med ; 22(11): 5494-5503, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30091835

RESUMEN

Acute lung injury (ALI) is mainly caused by uncontrolled inflammatory response, and it remains without effective therapeutic options. 25-hydroxycholesterol (25HC) has been reported to be a potent regulator of inflammation. The aim of this study was to investigate the effects of 25HC on lipopolysaccharide (LPS)-induced ALI. C57BL/6 mice were pretreated with 25HC intraperitoneally before intratracheal exposure to LPS. Our results showed that 25HC pretreatment improved survival rate, attenuated the pathological changes of the lung and decreased the release of inflammatory cytokines in mice. Consistently, 25HC reduced expression of Toll-like receptor (TLR4)-mediated inflammatory cytokines in vitro. These effects of 25HC were obtained by preventing LPS binding to TLR4 via interaction with myeloid differentiation protein 2 (MD-2). Crystal structure analysis suggested that 25HC could bind MD-2 with high affinity into its hydrophobic pocket. Furthermore, LPS-induced activation of Akt/NF-κB pathway was partially down-regulated by 25HC pretreatment. In summary, this study demonstrates that 25HC could inhibit the overwhelming inflammatory response through MD-2 interaction, which suppresses Akt/NF-κB signalling pathway. These findings suggest 25HC may be a promising candidate for ALI prevention.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Hidroxicolesteroles/administración & dosificación , Antígeno 96 de los Linfocitos/genética , Receptor Toll-Like 4/genética , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Animales , Antiinflamatorios/administración & dosificación , Líquido del Lavado Bronquioalveolar , Modelos Animales de Enfermedad , Humanos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/patología , Lipopolisacáridos/toxicidad , Pulmón/patología , Ratones , FN-kappa B/genética , Sustancias Protectoras , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos
8.
Stem Cells ; 34(7): 1947-56, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26866937

RESUMEN

Effective and specific therapeutic approaches are still needed for treating acute lung injury caused by severe pneumonia. Adipose-derived stem cells (ADSCs) are well-characterized adult stem cells that have antibacterial and anti-inflammatory effects. In this study, we evaluated the therapeutic effect of ADSCs on Staphylococcus aureus-induced acute lung injury in mice. Our results showed that intratracheal injection of ADSCs could attenuate the severity of lung inflammation, and reduce the bacterial load as well as mortality among infected mice. Our experiments also revealed that the secretion of regenerating islet-derived IIIγ (RegIIIγ) is responsible for the protective effect of ADSCs. Moreover, the expression of RegIIIγ requires TLR2, MyD88, and JAK2/STAT3 activation. In conclusion, ADSCs exhibit a direct antimicrobial activity that is mediated primarily by the TLR2-MyD88-JAK2/STAT3-dependent secretion of RegIIIγ. Stem Cells 2016;34:1947-1956.


Asunto(s)
Lesión Pulmonar Aguda/microbiología , Lesión Pulmonar Aguda/terapia , Tejido Adiposo/citología , Proteínas Asociadas a Pancreatitis/metabolismo , Staphylococcus aureus/fisiología , Células Madre/citología , Animales , Antiinfecciosos/metabolismo , Modelos Animales de Enfermedad , Femenino , Ratones Endogámicos C57BL , Modelos Biológicos , Proteínas Recombinantes/metabolismo , Transducción de Señal , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología
9.
J Immunol ; 194(3): 1239-51, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25520401

RESUMEN

A polarized macrophage response is presumed to have a pivotal role in a variety of immunological pathophysiology. However, the molecular mechanism underlying macrophage functional shaping remains largely unknown. In this study, we reveal a pivotal role of miR-127 in macrophage development and thereby the pathogenesis of inflammation and lung injury. In particular, miR-127 was demonstrated to be prominently induced upon TLR engagement and repressed by the M2-prone cytokines. Enforced expression of miR-127 in macrophages resulted in significantly increased production of proinflammatory cytokines, whereas deletion of miR-127 impaired M1 gene expression and led to a M2-biased response. Accordingly, intratracheal administration of miR-127 resulted in an exaggerated pulmonary inflammation and injury. Conversely, antagonizing of miR-127 suppressed production of the proinflammatory cytokines and rendered the mice more refractory to the inflammation-associated pathology. Mechanistically, miR-127 demonstrated to target B cell lymphoma 6 (Bcl6) and remarkably downregulated its expression and subsequently dual specificity phosphatase 1 (Dusp1), which in turn enhanced the activation of JNK kinase and hence the development of proinflammatory macrophages. Thereby, reconstitution with the expression of Bcl6 or Dusp1 or inhibition of JNK activity impaired miR-127-mediated skewing of M1 proinflammatory macrophages, whereas interference of Bcl6 or Dusp1 expression abrogated the anti-inflammatory property of anti-miR-127. Together, these data establish miR-127 as a molecular switch during macrophage development and as a potential target for treatment of inflammatory diseases.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Macrófagos/inmunología , Macrófagos/metabolismo , MicroARNs/genética , Neumonía/genética , Neumonía/metabolismo , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/metabolismo , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Fosfatasa 1 de Especificidad Dual/genética , Fosfatasa 1 de Especificidad Dual/metabolismo , Endotoxinas/efectos adversos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ligandos , Activación de Macrófagos/genética , Activación de Macrófagos/inmunología , Ratones , Neumonía/inmunología , Proteínas Proto-Oncogénicas c-bcl-6 , Interferencia de ARN , Sepsis/genética , Sepsis/inmunología , Sepsis/metabolismo , Receptores Toll-Like/metabolismo , Transcripción Genética , Transcriptoma
10.
J Infect Dis ; 214(4): 625-33, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27330052

RESUMEN

Macrophages can polarize and differentiate to regulate initiation, development, and cessation of inflammation during pulmonary infection with nontypeable Haemophilus influenzae (NTHi). However, the underlying molecular mechanisms driving macrophage phenotypic differentiation are largely unclear. Our study investigated the role of Shp2, a Src homology 2 domain-containing phosphatase, in the regulation of pulmonary inflammation and bacterial clearance. Shp2 levels were increased upon NTHi stimulation. Selective inhibition of Shp2 in mice led to an attenuated inflammatory response by skewing macrophages toward alternatively activated macrophage (M2) polarization. Upon pulmonary NTHi infection, Shp2(-/-) mice, in which the gene encoding Shp2 in monocytes/macrophages was deleted, showed an impaired inflammatory response and decreased antibacterial ability, compared with wild-type controls. In vitro data demonstrated that Shp2 regulated activated macrophage (M1) gene expression via activation of p65-nuclear factor-κB signaling, independent of p38 and extracellular regulated kinase-mitogen-activated proteins kinase signaling pathways. Taken together, our study indicates that Shp2 is required to orchestrate macrophage function and regulate host innate immunity against pulmonary bacterial infection.


Asunto(s)
Infecciones por Haemophilus/inmunología , Infecciones por Haemophilus/patología , Haemophilus influenzae/inmunología , Macrófagos/inmunología , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Femenino , Macrófagos/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados
11.
J Infect Dis ; 208(3): 528-38, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23613163

RESUMEN

Macrophage polarization is critical for dictating host defense against pathogens and injurious agents. Dysregulation of macrophage differentiation has been implicated in infectious and inflammatory diseases. Here, we show that protein kinase B/Akt1 signaling induced by Staphylococcus aureus is essential in shifting macrophages from an antimicrobial phenotype (M1) to a functionally inert signature. Akt1(-/-)mice consistently had enhanced bacterial clearance and greater survival, compared with their wild-type littermates. The blunted M1 macrophage reaction driven by Akt1 was associated with decreased RelA/nuclear factor κB activity. Furthermore, by repression of the expression of suppressor of cytokine signaling 1 (SOCS1), microRNA 155 revealed to promote the transcription of M1 signature genes in macrophages from Akt1(-/-) mice. Accordingly, blocking of microRNA 155 in macrophages from Akt1(-/-)mice or knockdown of SOCS1 in cells from wild-type mice disabled or enabled, respectively, an M1 macrophage shift and antibacterial response. These results thus establish an Akt1-mediated, microRNA-involved circuit that regulates pathogen-driven macrophage polarization and, subsequently, the host response to infection.


Asunto(s)
Macrófagos/inmunología , Neumonía Estafilocócica/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Staphylococcus aureus/inmunología , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Transducción de Señal
12.
Adv Sci (Weinh) ; 11(20): e2307969, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38482752

RESUMEN

Non-antibiotic strategies are desperately needed to treat post-traumatic osteomyelitis (PTO) due to the emergence of superbugs, complex inflammatory microenvironments, and greatly enriched biofilms. Previously, growing evidence indicated that quorum sensing (QS), a chemical communication signal among bacterial cells, can accelerate resistance under evolutionary pressure. This study aims to develop a medical dressing to treat PTO by inhibiting QS and regulating the inflammatory microenvironment, which includes severe oxidative stress and acid abscesses, through a reactive oxygen species (ROS)-responsive bond between N1- (4-borobenzoyl)-N3-(4-borobenzoyl)-the N1, the N1, N3, N3-tetramethylpropane-1,3-diamine (TSPBA) and polyvinyl alcohol (PVA), and the amino side chain of hyperbranched polylysine (HBPL). Physically enclosed QS inhibitors subsequently exerted the antibacterial effects. This hydrogel can scavenge hydrogen peroxide (H2O2), superoxide anion free radical (·O2 -), hydroxyl radicals (·OH) and 2,2-di(4-tert-octylphenyl)-1-picryl-hydrazyl (DPPH) to reduce oxidative stress and inhibit "bacteria-to-bacteria communication", thus clearing planktonic bacteria and biofilms, accelerating bacterial plasmolysis, reducing bacterial virulence and interfering with membrane transport. After in vivo treatment with hydrogel, nearly all bacteria are eliminated, inflammation is effectively inhibited, and osteogenesis and bone repair are promoted to facilitate recovery from PTO. The work demonstrates the clinical translational potential of the hydrogel in the treatment of drug-resistant bacteria induced PTO.


Asunto(s)
Hidrogeles , Osteomielitis , Percepción de Quorum , Percepción de Quorum/efectos de los fármacos , Hidrogeles/química , Hidrogeles/farmacología , Osteomielitis/tratamiento farmacológico , Osteomielitis/microbiología , Animales , Ratones , Modelos Animales de Enfermedad , Antibacterianos/farmacología , Estrés Oxidativo/efectos de los fármacos , Biopelículas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Ratas , Masculino
13.
Medicine (Baltimore) ; 102(1): e32589, 2023 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-36607848

RESUMEN

Most studies on human lung infection have been performed using animal models, formalin or other fixed tissues, and in vitro cultures of established cell lines. However, the experimental data and results obtained from these studies may not completely represent the complicated molecular events that take place in intact human lung tissue in vivo. The newly developed ex vivo short-term tissue culture model can mimic the in vivo microenvironment of humans and allow investigations of different cell types that closely interact with each other in intact human lung tissues. Therefore, this kind of model may be a promising tool for future studies of different human lung infections, owing to its special advantages in providing more realistic events that occur in vivo. In this review, we have summarized the preliminary applications of this novel short-term ex vivo tissue culture model, with a particular emphasis on its applications in some common human lung infections.


Asunto(s)
Pulmón , Animales , Humanos , Pulmón/metabolismo , Línea Celular , Modelos Animales
14.
ACS Nano ; 17(12): 11692-11712, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37310363

RESUMEN

Acute methicillin resistant Staphylococcus aureus (MRSA) pneumonia is one of the most frequently seen lung infection diseases with high morbidity and mortality. It is urgent to explore an efficient antibacterial strategy owing to the increase of drug resistance, virulence, and pathogenicity of MRSA. It was found that Fe3O4 can induce ferroptosis in MRSA, but its effect was inhibited by glutathione (GSH) to a certain extent, while cinnamaldehyde (CA) can enhance ferroptosis by consuming GSH. As a bacterial quorum sensing (QS) inhibitor, CA can suppress the QS system and further exert its antibacterial and antibiofilm effects. Here, we developed an Fe3O4-based ferroptosis inducer to promote ferroptosis in MRSA, interrupt the QS, destroy biofilm, and thus effectively treat acute MRSA pneumonia. We used sodium alginate (SA) to wrap Fe3O4 and CA to form particles, and then coated the surface with a hybrid biomimetic membrane composed of an erythrocyte membrane and platelet membrane to obtain lung targeted antibacterial particles (mFe-CA). Under ultrasonic (US) stimulation, mFe-CA can efficiently release Fe3O4 and CA, thereby synergically inducing MRSA death with the characteristics of ferroptosis, including mass ROS production, lipid peroxidation, GSH depletion, and respiratory chain suppression. Furthermore, mFe-CA + US can inhibit the QS system, remove biofilms, and reduce strain virulence. In the mouse model of MRSA pneumonia, mFe-CA + US treatment markedly advanced the survival rate of the mice, reduced the bacterial load in the lungs, and alleviated the inflammatory damage, but there was no obvious toxicity. This study proposes an antibacterial substitute to induce ferroptosis of MRSA, which may provide a foreground for overcoming microbial drug resistance and fighting biofilm-associated infections and also provides a target and theoretical basis for clinical treatment of acute MRSA pneumonia.


Asunto(s)
Ferroptosis , Staphylococcus aureus Resistente a Meticilina , Neumonía , Animales , Ratones , Biomimética , Antibacterianos/farmacología , Biopelículas , Pruebas de Sensibilidad Microbiana
15.
Rheumatol Int ; 32(7): 2189-93, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20354856

RESUMEN

Although lymph node enlargement is common in active systemic lupus erythematosus (SLE), lymph node examination is frequently ignored in the diagnosis of SLE. Clinical presentation and abnormal laboratory findings are often sufficient for SLE diagnosis, not to mention that the specific histological finding of lymph node necrosis in SLE is rarely seen, and the follicular hyperplasia is usually considered as nonspecific. However, since the late 1990s, a few cases of SLE lymphadenopathy have been reported exhibiting a Castleman's disease (CD) morphology, which was discovered in lymph node biopsies. Here we report a similar case of SLE combined with CD in a 23-year-old girl who displayed systemic symptoms, including systemic lymphadenopathy and abnormal laboratory findings indicating the active phase of SLE. A biopsy of neck lymphnodes showed histopathological features of CD. The patient responded very well to the prednisolone treatment. Based on the related literature review, we would like to stress the possibility of CD in patients with SLE lymphadenopathy.


Asunto(s)
Enfermedad de Castleman/patología , Lupus Eritematoso Sistémico/patología , Ganglios Linfáticos/patología , Biopsia , Enfermedad de Castleman/complicaciones , Enfermedad de Castleman/tratamiento farmacológico , Dolor en el Pecho/diagnóstico , Dolor en el Pecho/tratamiento farmacológico , Femenino , Glucocorticoides/uso terapéutico , Humanos , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/tratamiento farmacológico , Prednisolona/uso terapéutico , Adulto Joven
16.
Cell Biosci ; 12(1): 81, 2022 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-35658939

RESUMEN

BACKGROUND: As important enzymes regulating acetylation, histone deacetylases (HDACs) participate in a series of cell physiological process. However, the mechanisms responsible for individual HDAC family members in regulating innate immunity remained to be elucidated. Here we sought to reveal the mechanism of HDAC3 in regulating the inflammatory response of macrophages. METHODS: RNAseq was done to detect the transcriptional influence of HDAC3 on macrophages. Kyoto Encyclopedia of Genes and Genomes was used to reveal the change of signaling pathways after HDAC3 knockout. CHIPseq was done to detect the deacetylation modification of HDAC3 on chromosome. Western blot, immunofluorescence, and real-time quantitative PCR were used to measure the change of genes and proteins' levels. Mice were intratracheal instillation with lipopolysaccharide or Pseudomonas aeruginosa to determine the influence of HDAC3 on inflammatory response in vivo. RESULTS: HDAC3-deficient macrophages had increased expression of cathepsins resulting from elevated histone acetylation. Over-expressed cathepsins such as cathepsin B (CTSB) caused remarkable degradation of receptor (TNFRSF)-interacting serine-threonine kinase 1 (RIP1), which reduced TNFα mediated NF-κB activation and inflammatory response. Consistently, mice with macrophage specific knockout of HDAC3 were impaired in inflammatory response and thereby susceptible to Pseudomonas aeruginosa infection. CONCLUSION: HDAC3 was required for protecting RIP1 from degrading by CTSB in macrophages. Decreased RIP1 in HDAC3 knockout macrophages impaired TNFα mediated NF-κB activation. Our studies uncovered important roles of HDAC3 in the regulation of cathepsin-mediated lysosomal degradation and RIP1-mediated inflammatory response in macrophages as well as in host defense against bacterial infection.

17.
Cell Rep ; 38(4): 110302, 2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35081346

RESUMEN

It is well known that interferon (IFN)-α/-ß activates the JAK/STAT signaling pathway and suppresses viral replication through the induction of IFN stimulated genes (ISGs). Here, we report that knockout of HDAC3 from macrophages results in the decreased expression of STAT1 and STAT2, leading to defective antiviral immunity in cells and mice. Further studies show that HDAC3 interacts with a conserved transcription factor Forkhead Box K1 (FOXK1), co-localizes with FOXK1 at the promoter of STAT1 and STAT2, and is required for protecting FOXK1 from lysosomal system-mediated degradation. FOXK1-deficient macrophages also show low STAT1 and STAT2 expression with defective responses to viruses. Thus, our studies uncover the biological importance of HDAC3 in regulating the antiviral immunity of macrophages through interacting with FOXK1 to regulate the expression of STAT1 and STAT2.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Histona Desacetilasas/inmunología , Inmunidad Innata/inmunología , Macrófagos/inmunología , Virosis/inmunología , Animales , Factores de Transcripción Forkhead/inmunología , Ratones , Factor de Transcripción STAT1/biosíntesis , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT2/biosíntesis , Factor de Transcripción STAT2/inmunología , Transcripción Genética
18.
Redox Biol ; 41: 101936, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33752110

RESUMEN

Recently, numerous evidence has revealed that excessive reactive oxygen species (ROS) production and mitochondrial disruption during acute lung injury (ALI) and its most severe form, acute respiratory distress syndrome (ARDS) will aggravate the inflammatory process. To identify whether antioxidation can be one of the treatment strategies during this progress, we chose mitoQ, a mitochondria-targeted antioxidant that was proved to be effective in reducing ROS generated in mitochondria, as a ROS scavenger to investigate the role of antioxidation in ALI. We demonstrated that overoxidation occurred during the process of ALI, which could be reduced by mitoQ. In the meantime, apoptosis of endothelial cells of ALI mice, accompanied by hyperpermeability of pulmonary vascular and impaired pulmonary function, was partially reversed following an intraperitoneal injection of mitoQ. Moreover, in in vitro study, lipopolysaccharides (LPS) induced excessive ROS production, mitochondrial dysfunction and apoptosis in human pulmonary microvascular endothelial cells (HPMECs), which were rectified by mitoQ. To explore underlying mechanisms, we proceeded RNA-sequencing and found significantly upregulated expression of musculoaponeurotic fibrosarcoma F (MafF) in mitoQ treated group. Additionally, mitoQ inhibited the degradation and increased nuclear translocation of NF-E2-related factor 2 (Nrf2) and upregulated its downstream antioxidant response elements (AREs), such as heme oxygenase (HO)-1 and NAD(P)H:quinone oxidoreductase (NQO)-1. This effect was abolished by transfecting HPMECs with Nrf2 or Maff siRNA. In Nrf2 deficient mice, the protective effects of mitoQ on LPS model of ALI were largely vanished. Taken together, these results provide insights into how antioxidation exerts beneficial effects on ALI via maintaining mitochondrial hemostasis, inhibiting endothelial cells apoptosis, attenuating the endothelial disruption and regulating lung inflammation via Nrf2-MafF/ARE pathway.


Asunto(s)
Lesión Pulmonar Aguda , Factor 2 Relacionado con NF-E2 , Animales , Antioxidantes/farmacología , Células Endoteliales/metabolismo , Endotelio/metabolismo , Lipopolisacáridos , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , Transducción de Señal
19.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA