Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 961
Filtrar
1.
J Immunol ; 210(8): 1086-1097, 2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36883861

RESUMEN

Fatty acid-binding protein 4 (FABP4) is a critical immune-metabolic modulator, mainly expressed in adipocytes and macrophages, secreted from adipocytes in association with lipolysis, and plays essential pathogenic roles in cardiovascular and metabolic diseases. We previously reported Chlamydia pneumoniae infecting murine 3T3-L1 adipocytes and causing lipolysis and FABP4 secretion in vitro. However, it is still unknown whether C. pneumoniae intranasal lung infection targets white adipose tissues (WATs), induces lipolysis, and causes FABP4 secretion in vivo. In this study, we demonstrate that C. pneumoniae lung infection causes robust lipolysis in WAT. Infection-induced WAT lipolysis was diminished in FABP4-/- mice or FABP4 inhibitor-pretreated wild-type mice. Infection by C. pneumoniae in wild-type but not FABP4-/- mice induces the accumulation of TNF-α- and IL-6-producing M1-like adipose tissue macrophages in WAT. Infection-induced WAT pathology is augmented by endoplasmic reticulum (ER) stress/the unfolded protein response (UPR), which is abrogated by treatment with azoramide, a modulator of the UPR. C. pneumoniae lung infection is suggested to target WAT and induce lipolysis and FABP4 secretion in vivo via ER stress/UPR. FABP4 released from infected adipocytes may be taken up by other neighboring intact adipocytes or adipose tissue macrophages. This process can further induce ER stress activation and trigger lipolysis and inflammation, followed by FABP4 secretion, leading to WAT pathology. A better understanding of the role of FABP4 in C. pneumoniae infection-induced WAT pathology will provide the basis for rational intervention measures directed at C. pneumoniae infection and metabolic syndrome, such as atherosclerosis, for which robust epidemiologic evidence exists.


Asunto(s)
Tejido Adiposo Blanco , Infecciones por Chlamydophila , Proteínas de Unión a Ácidos Grasos , Neumonía Bacteriana , Animales , Ratones , Tejido Adiposo Blanco/patología , Chlamydophila pneumoniae , Proteínas de Unión a Ácidos Grasos/metabolismo , Pulmón/microbiología , Pulmón/patología , Infecciones por Chlamydophila/patología , Neumonía Bacteriana/patología
2.
Shock ; 58(5): 400-407, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36166827

RESUMEN

ABSTRACT: Traumatic brain injury is one of the main causes of death and disability worldwide, and results in multisystem complications. However, the mechanism of mild traumatic brain injury (MTBI) on lung injury remains unclear. In this study, we used a murine model of MTBI and pneumonia ( Pseudomonas aeruginosa ;) to explore the relationship between these conditions and the underlying mechanism. Methods: Mice (n = 104) were divided into control, MTBI, pneumonia, and MTBI + pneumonia groups. MTBI was induced by the weight-drop method. Pneumonia was induced by intratracheal injection with P. aeruginosa Xen5 strain. Animals were killed 24 h after bacterial challenging. Histological, cellular, and molecular indices of brain and lung injury were assessed using various methods. Results: Mice in both the MTBI and pneumonia groups had more Fluoro-Jade C-positive neurons than did the controls ( P < 0.01), but mice in the MTBI + pneumonia group had fewer Fluoro-Jade C-positive cells than did the pneumonia group ( P < 0.01). The MTBI + pneumonia mice showed decreased bacterial load ( P < 0.05), reduced lung injury score and pulmonary permeability ( P < 0.01), less inflammatory cells, and lower levels of proinflammatory cytokines (TNF-α and IL-1ß; P < 0.01) when compared with the pneumonia group. Molecular analysis indicated lower levels of phosphorylated nuclear factor-κB in the lung of MTBI + pneumonia mice compared with the pneumonia group ( P < 0.01). Furthermore, alveolar macrophages from MTBI mice exhibited enhanced bactericidal capacity compared with those from controls ( P < 0.01). Moreover, MTBI + pneumonia mice exhibited less CD86-positive M1 macrophages compared with the pneumonia group ( P < 0.01). Conclusions: MTBI attenuates pneumonia-induced acute lung injury through the modulation of alveolar macrophage bactericidal capacity and M1 polarization in bacterial pneumonia model.


Asunto(s)
Lesión Pulmonar Aguda , Conmoción Encefálica , Neumonía Bacteriana , Animales , Ratones , Lesión Pulmonar Aguda/inducido químicamente , Conmoción Encefálica/patología , Lipopolisacáridos , Pulmón/patología , Macrófagos Alveolares/patología , FN-kappa B , Neumonía Bacteriana/patología
3.
Shock ; 58(4): 295-303, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-36018281

RESUMEN

ABSTRACT: Objectives: Nosocomial pneumonia is a common complication in critically ill patients. The goal of this study was to examine the efficacy of the Toll-like receptor 4 agonist 3-deacyl phosphorylated hexacyl disaccharide (3D PHAD), in a clinically relevant murine model of pneumonia, and assess the cellular mechanisms that mediate the protective response. Design: Mice received intrapulmonary 3D PHAD (20 µg) or vehicle for 2 consecutive days before challenge with intrapulmonary Klebsiella pneumoniae (2.3 × 10 3 colony-forming units). Mice were followed for 14-day survival, pulmonary K. pneumoniae burden, lung leukocyte profile, leukocyte phagocytic capacity, and cytokine production. Pneumonia severity and leukocyte recruitment were further assessed by histological evaluation. Setting: Research laboratory. Subjects: Wild-type, male C57BL/6 J mice. Interventions: Intrapulmonary treatment with 20 µg 3D PHAD for 2 consecutive days. Measurements and main results: Intrapulmonary treatment with 3D PHAD decreased lung K. pneumoniae colony-forming units and pneumonia severity with an associated improvement in survival compared with mice treated with vehicle. The numbers of neutrophils, monocytes, and macrophages in the lungs of 3D PHAD-treated mice were higher than those in vehicle-treated mice before infection but were not significantly different from vehicle-treated mice at 48 h after K. pneumoniae challenge. Lung innate leukocytes from 3D PHAD-treated mice had increased phagocytic capacity. Treatment with 3D PHAD alone increased cytokines in the lungs but decreased cytokines in plasma during K. pneumoniae pneumonia as compared with control. Conclusions: Intrapulmonary treatment with 3D PHAD augments innate immunity in the lung and facilitates resistance to K. pneumoniae pneumonia.


Asunto(s)
Infecciones por Klebsiella , Neumonía Bacteriana , Masculino , Ratones , Animales , Klebsiella pneumoniae , Receptor Toll-Like 4 , Ratones Endogámicos C57BL , Neumonía Bacteriana/patología , Citocinas , Pulmón/patología , Disacáridos
4.
JCI Insight ; 7(4)2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35050901

RESUMEN

Lung alveolar type 2 (AT2) cells are progenitors for alveolar type 1 (AT1) cells. Although many factors regulate AT2 cell plasticity, the role of mitochondrial calcium (mCa2+) uptake in controlling AT2 cells remains unclear. We previously identified that the miR-302 family supports lung epithelial progenitor cell proliferation and less differentiated phenotypes during development. Here, we report that a sustained elevation of miR-302 in adult AT2 cells decreases AT2-to-AT1 cell differentiation during the Streptococcus pneumoniae-induced lung injury repair. We identified that miR-302 targets and represses the expression of mitochondrial Ca2+ uptake 1 (MICU1), which regulates mCa2+ uptake through the mCa2+ uniporter channel by acting as a gatekeeper at low cytosolic Ca2+ levels. Our results reveal a marked increase in MICU1 protein expression and decreased mCa2+ uptake during AT2-to-AT1 cell differentiation in the adult lung. Deletion of Micu1 in AT2 cells reduces AT2-to-AT1 cell differentiation during steady-state tissue maintenance and alveolar epithelial regeneration after bacterial pneumonia. These studies indicate that mCa2+ uptake is extensively modulated during AT2-to-AT1 cell differentiation and that MICU1-dependent mCa2+ uniporter channel gating is a prominent mechanism modulating AT2-to-AT1 cell differentiation.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Proteínas de Unión al Calcio/genética , Calcio/metabolismo , Regulación de la Expresión Génica , Proteínas de Transporte de Membrana Mitocondrial/genética , Neumonía Bacteriana/genética , ARN/genética , Regeneración/genética , Células Epiteliales Alveolares/patología , Animales , Transporte Biológico , Proteínas de Unión al Calcio/biosíntesis , Diferenciación Celular , Plasticidad de la Célula , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Transporte de Membrana Mitocondrial/biosíntesis , Neumonía Bacteriana/metabolismo , Neumonía Bacteriana/patología
5.
PLoS One ; 17(1): e0262052, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35061767

RESUMEN

The COVID-19 epidemic has a catastrophic impact on global well-being and public health. More than 27 million confirmed cases have been reported worldwide until now. Due to the growing number of confirmed cases, and challenges to the variations of the COVID-19, timely and accurate classification of healthy and infected patients is essential to control and treat COVID-19. We aim to develop a deep learning-based system for the persuasive classification and reliable detection of COVID-19 using chest radiography. Firstly, we evaluate the performance of various state-of-the-art convolutional neural networks (CNNs) proposed over recent years for medical image classification. Secondly, we develop and train CNN from scratch. In both cases, we use a public X-Ray dataset for training and validation purposes. For transfer learning, we obtain 100% accuracy for binary classification (i.e., Normal/COVID-19) and 87.50% accuracy for tertiary classification (Normal/COVID-19/Pneumonia). With the CNN trained from scratch, we achieve 93.75% accuracy for tertiary classification. In the case of transfer learning, the classification accuracy drops with the increased number of classes. The results are demonstrated by comprehensive receiver operating characteristics (ROC) and confusion metric analysis with 10-fold cross-validation.


Asunto(s)
COVID-19/diagnóstico por imagen , Aprendizaje Profundo , Interpretación de Imagen Asistida por Computador/métodos , Neumonía Bacteriana/diagnóstico por imagen , COVID-19/patología , COVID-19/virología , Estudios de Casos y Controles , Bases de Datos Factuales , Diagnóstico Diferencial , Femenino , Humanos , Masculino , Neumonía Bacteriana/patología , Neumonía Bacteriana/virología , Curva ROC , Radiografía Torácica , SARS-CoV-2/patogenicidad
6.
JCI Insight ; 7(2)2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-34908534

RESUMEN

Acute respiratory distress syndrome (ARDS) is a life-threatening syndrome, constituted by respiratory failure and diffuse alveolar damage that results from dysregulated local and systemic immune activation, causing pulmonary vascular, parenchymal, and alveolar damage. SARS-CoV-2 infection has become the dominant cause of ARDS worldwide, and emerging evidence implicates neutrophils and their cytotoxic arsenal of effector functions as central drivers of immune-mediated lung injury in COVID-19 ARDS. However, key outstanding questions are whether COVID-19 drives a unique program of neutrophil activation or effector functions that contribute to the severe pathogenesis of this pandemic illness and whether this unique neutrophil response can be targeted to attenuate disease. Using a combination of high-dimensional single-cell analysis and ex vivo functional assays of neutrophils from patients with COVID-19 ARDS, compared with those with non-COVID ARDS (caused by bacterial pneumonia), we identified a functionally distinct landscape of neutrophil activation in COVID-19 ARDS that was intrinsically programmed during SARS-CoV-2 infection. Furthermore, neutrophils in COVID-19 ARDS were functionally primed to produce high amounts of neutrophil extracellular traps. Surprisingly, this unique pathological program of neutrophil priming escaped conventional therapy with dexamethasone, thereby revealing a promising target for adjunctive immunotherapy in severe COVID-19.


Asunto(s)
COVID-19/inmunología , Trampas Extracelulares/inmunología , Activación Neutrófila , Neutrófilos/inmunología , Síndrome de Dificultad Respiratoria/inmunología , SARS-CoV-2/inmunología , Adulto , Anciano , Anciano de 80 o más Años , COVID-19/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neutrófilos/patología , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/patología , Síndrome de Dificultad Respiratoria/patología , Índice de Severidad de la Enfermedad
7.
J Biol Chem ; 298(1): 101482, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34896150

RESUMEN

Patients who recover from nosocomial pneumonia oftentimes exhibit long-lasting cognitive impairment comparable with what is observed in Alzheimer's disease patients. We previously hypothesized that the lung endothelium contributes to infection-related neurocognitive dysfunction, because bacteria-exposed endothelial cells release a form(s) of cytotoxic tau that is sufficient to impair long-term potentiation in the hippocampus. However, the full-length lung and endothelial tau isoform(s) have yet to be resolved and it remains unclear whether the infection-induced endothelial cytotoxic tau triggers neuronal tau aggregation. Here, we demonstrate that lung endothelial cells express a big tau isoform and three additional tau isoforms that are similar to neuronal tau, each containing four microtubule-binding repeat domains, and that tau is expressed in lung capillaries in vivo. To test whether infection elicits endothelial tau capable of causing transmissible tau aggregation, the cells were infected with Pseudomonas aeruginosa. The infection-induced tau released from endothelium into the medium-induced neuronal tau aggregation in reporter cells, including reporter cells that express either the four microtubule-binding repeat domains or the full-length tau. Infection-induced release of pathological tau variant(s) from endothelium, and the ability of the endothelial-derived tau to cause neuronal tau aggregation, was abolished in tau knockout cells. After bacterial lung infection, brain homogenates from WT mice, but not from tau knockout mice, initiated tau aggregation. Thus, we conclude that bacterial pneumonia initiates the release of lung endothelial-derived cytotoxic tau, which is capable of propagating a neuronal tauopathy.


Asunto(s)
Enfermedades Pulmonares , Neumonía Bacteriana , Tauopatías , Proteínas tau , Animales , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/microbiología , Disfunción Cognitiva/patología , Células Endoteliales/metabolismo , Células Endoteliales/microbiología , Células Endoteliales/patología , Humanos , Pulmón/irrigación sanguínea , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/microbiología , Enfermedades Pulmonares/patología , Ratones , Neumonía Bacteriana/metabolismo , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/patología , Isoformas de Proteínas , Pseudomonas aeruginosa , Tauopatías/genética , Tauopatías/metabolismo , Tauopatías/patología , Proteínas tau/química , Proteínas tau/genética , Proteínas tau/metabolismo
8.
Sci Rep ; 11(1): 21210, 2021 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-34707211

RESUMEN

Pleural fibrosis (PF) is a chronic and progressive lung disease which affects approximately 30,000 people per year in the United States. Injury and sustained inflammation of the pleural space can result in PF, restricting lung expansion and impairing oxygen exchange. During the progression of pleural injury, normal pleural mesothelial cells (PMCs) undergo a transition, termed mesothelial mesenchymal transition (MesoMT). While multiple components of the fibrinolytic pathway have been investigated in pleural remodeling and PF, the role of the urokinase type plasminogen activator receptor (uPAR) is unknown. We found that uPAR is robustly expressed by pleural mesothelial cells in PF. Downregulation of uPAR by siRNA blocked TGF-ß mediated MesoMT. TGF-ß was also found to significantly induce uPA expression in PMCs undergoing MesoMT. Like uPAR, uPA downregulation blocked TGF-ß mediated MesoMT. Further, uPAR is critical for uPA mediated MesoMT. LRP1 downregulation likewise blunted TGF-ß mediated MesoMT. These findings are consistent with in vivo analyses, which showed that uPAR knockout mice were protected from S. pneumoniae-mediated decrements in lung function and restriction. Histological assessments of pleural fibrosis including pleural thickening and α-SMA expression were likewise reduced in uPAR knockout mice compared to WT mice. These studies strongly support the concept that uPAR targeting strategies could be beneficial for the treatment of PF.


Asunto(s)
Transición Epitelial-Mesenquimal , Neumonía Bacteriana/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Infecciones Estreptocócicas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Actinas/metabolismo , Animales , Células Cultivadas , Epitelio/metabolismo , Epitelio/patología , Fibrosis , Humanos , Ratones , Ratones Endogámicos C57BL , Pleura/metabolismo , Pleura/patología , Neumonía Bacteriana/patología , Infecciones Estreptocócicas/patología , Activador de Plasminógeno de Tipo Uroquinasa/genética
9.
Am J Physiol Lung Cell Mol Physiol ; 321(5): L827-L836, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34524905

RESUMEN

We previously reported that extracellular vesicles (EVs) released during Escherichia coli (E. coli) bacterial pneumonia were inflammatory, and administration of high molecular weight hyaluronic acid (HMW HA) suppressed several indices of acute lung injury (ALI) from E. coli pneumonia by binding to these inflammatory EVs. The current study was undertaken to study the therapeutic effects of HMW HA in ex vivo perfused human lungs injured with Pseudomonas aeruginosa (PA)103 bacterial pneumonia. For lungs with baseline alveolar fluid clearance (AFC) <10%/h, HMW HA 1 or 2 mg was injected intravenously after 1 h (n = 4-9), and EVs released during PA pneumonia were collected from the perfusate over 6 h. For lungs with baseline AFC > 10%/h, HMW HA 2 mg was injected intravenously after 1 h (n = 6). In vitro experiments were conducted to evaluate the effects of HA on inflammation and bacterial phagocytosis. For lungs with AFC < 10%/h, administration of HMW HA intravenously significantly restored AFC and numerically decreased protein permeability and alveolar inflammation from PA103 pneumonia but had no effect on bacterial counts at 6 h. However, HMW HA improved bacterial phagocytosis by human monocytes and neutrophils and suppressed the inflammatory properties of EVs released during pneumonia on monocytes. For lungs with AFC > 10%/h, administration of HMW HA intravenously improved AFC from PA103 pneumonia but had no significant effects on protein permeability, inflammation, or bacterial counts. In the presence of impaired alveolar epithelial transport capacity, administration of HMW HA improved the resolution of pulmonary edema from Pseudomonas PA103 bacterial pneumonia.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Ácido Hialurónico/farmacología , Neumonía Bacteriana/tratamiento farmacológico , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/efectos de los fármacos , Edema Pulmonar/tratamiento farmacológico , Lesión Pulmonar Aguda/microbiología , Lesión Pulmonar Aguda/patología , Adulto , Vesículas Extracelulares/patología , Femenino , Humanos , Pulmón/efectos de los fármacos , Pulmón/microbiología , Pulmón/patología , Masculino , Persona de Mediana Edad , Monocitos/inmunología , Neutrófilos/inmunología , Técnicas de Cultivo de Órganos , Fagocitosis/efectos de los fármacos , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/patología , Edema Pulmonar/microbiología , Edema Pulmonar/patología , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/microbiología , Síndrome de Dificultad Respiratoria/patología
10.
Food Funct ; 12(19): 9466-9475, 2021 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-34473137

RESUMEN

Chlorogenic acid (CGA) possesses a wide variety of bioactive properties, such as antioxidation, anti-inflammation and anti-bacteria. This study was aimed at exploring the effects of CGA of anti-inflammation and anti-bacteria on mouse pneumonia prepared by immunosuppressed mice infected with Klebsiella pneumoniae (K. pneumoniae) in vivo and the cellular inflammasomes through lipopolysaccharide (LPS) and adenosine triphosphate (ATP)-induced RAW 264.7 murine macrophages in vitro. Mice received CGA treatment (30 and 90 mg kg-1) for 8 consecutive days and on the fourth day immunosuppression in mice was induced by cyclophosphamide (40 mg kg-1) for 5 days before inoculation of K. pneumoniae. Immunosuppressed mice infected with K. pneumoniae developed severe pneumonia, with marked interstitial vascular congestion, widened alveolar intervals, infiltration of monocytes, lymphocytes and macrophages as well as the damage of epithelial architecture, with growing mortality and count forming unit (CFU). CGA treatment significantly decreased the ratio of lung/body weight, reduced the severity of pneumonia induced by K. pneumoniae, decreased the lung injury, inflammatory cell infiltration scores and CD68 protein expression, inhibited the expression of interleukin (IL)-6, IL-8, tumor necrosis factor (TNF)-α, and elevated the expression of IL-10. Meanwhile, we investigated the mechanism of CGA to counter K. pneumoniae-induced pneumonia and found that CGA remarkably repressed the activation of nucleotide-binding domain like receptor protein 3 (NLRP3) inflammasome. Altogether, our results indicate that the dietary intake of CGA or its rich foods ameliorates K. pneumonia-induced pneumonia by inhibiting the activation of NLRP3 inflammasomes.


Asunto(s)
Ácido Clorogénico/uso terapéutico , Tolerancia Inmunológica , Inflamasomas/metabolismo , Infecciones por Klebsiella/tratamiento farmacológico , Klebsiella pneumoniae , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neumonía Bacteriana/tratamiento farmacológico , Animales , Ácido Clorogénico/farmacología , Citocinas/genética , Citocinas/metabolismo , Infecciones por Klebsiella/inmunología , Infecciones por Klebsiella/metabolismo , Infecciones por Klebsiella/patología , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/metabolismo , Neumonía Bacteriana/patología , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos
11.
PLoS One ; 16(8): e0256166, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34383849

RESUMEN

As electronic cigarette (E-cig) use, also known as "vaping", has rapidly increased in popularity, data regarding potential pathologic effects are recently emerging. Recent associations between vaping and lung pathology have led to an increased need to scrutinize E-cigs for adverse health impacts. Our previous work (and others) has associated vaping with Ca2+-dependent cytotoxicity in cultured human airway epithelial cells. Herein, we develop a vaped e-liquid pulmonary exposure mouse model to evaluate vaping effects in vivo. Using this model, we demonstrate lung pathology through the use of preclinical measures, that is, the lung wet: dry ratio and lung histology/H&E staining. Further, we demonstrate that acute vaping increases macrophage chemotaxis, which was ascertained using flow cytometry-based techniques, and inflammatory cytokine production, via Luminex analysis, through a Ca2+-dependent mechanism. This increase in macrophage activation appears to exacerbate pulmonary pathology resulting from microbial infection. Importantly, modulating Ca2+ signaling may present a therapeutic direction for treatment against vaping-associated pulmonary inflammation.


Asunto(s)
Calcio/metabolismo , Mezclas Complejas/efectos adversos , Infecciones por Klebsiella/etiología , Klebsiella pneumoniae/patogenicidad , Neumonía Bacteriana/etiología , Vapeo/efectos adversos , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/inmunología , Quimiotaxis/inmunología , Sistemas Electrónicos de Liberación de Nicotina , Expresión Génica , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Infecciones por Klebsiella/inmunología , Infecciones por Klebsiella/microbiología , Infecciones por Klebsiella/patología , Klebsiella pneumoniae/fisiología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/patología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
12.
Mol Med ; 27(1): 79, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34271850

RESUMEN

BACKGROUND: High mobility group box 1 protein (HMGB1) is an alarmin following its release by immune cells upon cellular activation or stress. High levels of extracellular HMGB1 play a critical role in impairing the clearance of invading pulmonary pathogens and dying neutrophils in the injured lungs of cystic fibrosis (CF) and acute respiratory distress syndrome (ARDS). A heparin derivative, 2-O, 3-O desulfated heparin (ODSH), has been shown to inhibit HMGB1 release from a macrophage cell line and is efficacious in increasing bacterial clearance in a mouse model of pneumonia. Thus, we hypothesized that ODSH can attenuate the bacterial burden and inflammatory lung injury in CF and we conducted experiments to determine the underlying mechanisms. METHODS: We determined the effects of ODSH on lung injury produced by Pseudomonas aeruginosa (PA) infection in CF mice with the transmembrane conductance regulator gene knockout (CFTR-/-). Mice were given ODSH or normal saline intraperitoneally, followed by the determination of the bacterial load and lung injury in the airways and lung tissues. ODSH binding to HMGB1 was determined using surface plasmon resonance and in silico docking analysis of the interaction of the pentasaccharide form of ODSH with HMGB1. RESULTS: CF mice given 25 mg/kg i.p. of ODSH had significantly lower PA-induced lung injury compared to mice given vehicle alone. The CF mice infected with PA had decreased levels of nitric oxide (NO), increased levels of airway HMGB1 and HMGB1-impaired macrophage phagocytic function. ODSH partially attenuated the PA-induced alteration in the levels of NO and airway HMGB1 in CF mice. In addition, ODSH reversed HMGB1-impaired macrophage phagocytic function. These effects of ODSH subsequently decreased the bacterial burden in the CF lungs. In a surface plasmon resonance assay, ODSH interacted with HMGB1 with high affinity (KD = 3.89 × 10-8 M) and induced conformational changes that may decrease HMGB1's binding to its membrane receptors, thus attenuating HMGB1-induced macrophage dysfunction. CONCLUSIONS: The results suggest that ODSH can significantly decrease bacterial infection-induced lung injury in CF mice by decreasing both HMGB1-mediated impairment of macrophage function and the interaction of HMGB1 with membrane receptors. Thus, ODSH could represent a novel approach for treating CF and ARDS patients that have HMGB1-mediated lung injury.


Asunto(s)
Fibrosis Quística/complicaciones , Fibrosis Quística/metabolismo , Proteína HMGB1/genética , Heparina/análogos & derivados , Macrófagos/inmunología , Macrófagos/metabolismo , Neumonía Bacteriana/etiología , Neumonía Bacteriana/metabolismo , Animales , Carga Bacteriana , Biomarcadores , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Proteína HMGB1/química , Proteína HMGB1/metabolismo , Heparina/química , Heparina/metabolismo , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Modelos Moleculares , Óxido Nítrico/metabolismo , Fagocitosis/inmunología , Neumonía Bacteriana/patología , Unión Proteica , Células RAW 264.7 , Relación Estructura-Actividad
13.
Microbiol Spectr ; 9(1): e0020721, 2021 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-34319124

RESUMEN

Tenascin C (TNC) is an extracellular matrix protein with immunomodulatory properties that plays a major role during tissue injury and repair. TNC levels are increased in patients with pneumonia and pneumosepsis, and they are associated with worse outcomes. Methicillin-resistant Staphylococcus aureus (MRSA) is a Gram-positive bacterium that is a major causative pathogen in nosocomial pneumonia and a rising cause of community-acquired pneumonia. To study the role of TNC during MRSA-induced pneumonia, TNC sufficient (TNC+/+) and TNC-deficient (TNC-/-) mice were infected with MRSA via the airways and euthanized after 6, 24, and 48 h for analysis. Pulmonary transcription of TNC peaked at 6 h, while immunohistochemistry revealed higher protein levels at later time points. Although TNC deficiency was not associated with changes in bacterial clearance, TNC-/- mice showed increased levels of TNF-α and IL-6 in bronchoalveolar lavage fluid during the acute phase of infection when compared with TNC+/+ mice. In addition, TNC-/- mice showed more severe pulmonary pathology at 6, but not at 24 or 48 h, after infection. Together, these data suggest that TNC plays a moderate protective role against tissue pathology during the acute inflammatory phase, but not during the bacterial clearance phase, of MRSA-induced pneumonia. These results argue against an important role of TNC on disease outcome during MRSA-induced pneumonia. IMPORTANCE Recently, the immunomodulatory properties of TNC have drawn substantial interest. However, to date most studies made use of sterile models of inflammation. In this study, we examine the pathobiology of MRSA-induced pneumonia in a model of TNC-sufficient and TNC-deficient mice. We have studied the immune response and tissue pathology both during the initial insult and also during the resolution phase. We demonstrate that MRSA-induced pneumonia upregulates pulmonary TNC expression at the mRNA and protein levels. However, the immunomodulatory role of TNC during bacterial pneumonia is distinct from models of sterile inflammation, indicating that the function of TNC is context dependent. Contrary to previous descriptions of TNC as a proinflammatory mediator, TNC-deficient mice seem to suffer from enhanced tissue pathology during the acute phase of infection. Nonetheless, besides its role during the acute phase response, TNC does not seem to play a major role in disease outcome during MRSA-induced pneumonia.


Asunto(s)
Pulmón/microbiología , Staphylococcus aureus Resistente a Meticilina/fisiología , Neumonía Bacteriana/metabolismo , Infecciones Estafilocócicas/metabolismo , Tenascina/metabolismo , Animales , Femenino , Humanos , Pulmón/metabolismo , Pulmón/patología , Masculino , Staphylococcus aureus Resistente a Meticilina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía Bacteriana/genética , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/patología , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología , Tenascina/genética
14.
mBio ; 12(3): e0326920, 2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34061598

RESUMEN

Influenza A virus (IAV) infection predisposes the host to secondary bacterial pneumonia, known as a major cause of morbidity and mortality during influenza virus epidemics. Analysis of interactions between IAV-infected human epithelial cells and Streptococcus pneumoniae revealed that infected cells ectopically exhibited the endoplasmic reticulum chaperone glycoprotein 96 (GP96) on the surface. Importantly, efficient pneumococcal adherence to epithelial cells was imparted by interactions with extracellular GP96 and integrin αV, with the surface expression mediated by GP96 chaperone activity. Furthermore, abrogation of adherence was gained by chemical inhibition or genetic knockout of GP96 as well as addition of RGD peptide, an inhibitor of integrin-ligand interactions. Direct binding of extracellular GP96 and pneumococci was shown to be mediated by pneumococcal oligopeptide permease components. Additionally, IAV infection induced activation of calpains and Snail1, which are responsible for degradation and transcriptional repression of junctional proteins in the host, respectively, indicating increased bacterial translocation across the epithelial barrier. Notably, treatment of IAV-infected mice with the GP96 inhibitor enhanced pneumococcal clearance from lung tissues and ameliorated lung pathology. Taken together, the present findings indicate a viral-bacterial synergy in relation to disease progression and suggest a paradigm for developing novel therapeutic strategies tailored to inhibit pneumococcal colonization in an IAV-infected respiratory tract. IMPORTANCE Secondary bacterial pneumonia following an influenza A virus (IAV) infection is a major cause of morbidity and mortality. Although it is generally accepted that preceding IAV infection leads to increased susceptibility to secondary bacterial infection, details regarding the pathogenic mechanism during the early stage of superinfection remain elusive. Here, we focused on the interaction of IAV-infected cells and Streptococcus pneumoniae, which revealed that human epithelial cells infected with IAV exhibit a cell surface display of GP96, an endoplasmic reticulum chaperon. Notably, extracellular GP96 was shown to impart efficient adherence for secondary infection by S. pneumoniae, and GP96 inhibition ameliorated lung pathology of superinfected mice, indicating it to be a useful target for development of therapeutic strategies for patients with superinfection.


Asunto(s)
Virus de la Influenza A/patogenicidad , Gripe Humana/complicaciones , Glicoproteínas de Membrana/genética , Neumonía Bacteriana/virología , Streptococcus pneumoniae/patogenicidad , Brote de los Síntomas , Células A549 , Animales , Adhesión Bacteriana , Coinfección/complicaciones , Coinfección/microbiología , Coinfección/virología , Células Epiteliales/microbiología , Femenino , Humanos , Gripe Humana/virología , Pulmón/microbiología , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/microbiología , Infecciones por Orthomyxoviridae/virología , Neumonía Bacteriana/etiología , Neumonía Bacteriana/patología
15.
J Trauma Acute Care Surg ; 90(6): 924-934, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34016916

RESUMEN

BACKGROUND: Forty percent of critically ill trauma patients will develop an infectious complication. Pneumonia is the most common cause of death of trauma patients surviving their initial insult. We previously demonstrated that polytrauma (PT), defined as two or more severe injuries in at least two areas of the body, induces emergency hematopoiesis characterized by accelerated myelopoiesis in the bone marrow and increased myeloid cell frequency in the peripheral tissues. We hypothesized that PT alone induces priming of neutrophils, resulting in hyperactivation upon secondary exposure to bacteria and causing acute lung injury and increased susceptibility to secondary exposure to Pseudomonas aeruginosa pneumonia. METHODS: C57BL/6 mice were subjected to PT consisting of a lower extremity pseudofracture, liver crush injury, and 15% blood-volume hemorrhage. Pneumonia was induced by intratracheal injection of 5 × 106 CFU live P. aeruginosa or 1 × 107 of heat-killed P. aeruginosa (HKPA). For reactive oxygen species (ROS), studies polymorphonuclear neutrophils (PMNs) were isolated by immunomagnetic bead negative selection and stimulated ex-vivo with HKPA. Reactive oxygen species production was measured by immunofluorescence. For histology, lung sections were stained by hematoxylin-eosin and analyzed by a blinded grader. RESULTS: Polytrauma induced persistent changes in immune function at baseline and to secondary infection. Pneumonia after injury resulted in increased mortality (60% vs. 5% p < 0.01). Blood neutrophils from PT mice had higher resting (unstimulated) ROS production than in naive animals (p < 0.02) demonstrating priming of the neutrophils following PT. After intratracheal HKPA injection, bronchoalveolar lavage PMNs from injured mice had higher ROS production compared with naive mice (p < 0.01), demonstrating an overexuberant immunopathologic response of neutrophils following PT. CONCLUSION: Polytrauma primes neutrophils and causes immunopathologic PMN ROS production, increased lung injury and susceptibility to secondary bacterial pneumonia. These results suggest that trauma-induced immune dysfunction can cause immunopathologic response to secondary infection and suggests neutrophil-mediated pulmonary damage as a therapeutic target for posttrauma pneumonia.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Traumatismo Múltiple/complicaciones , Neutrófilos/inmunología , Neumonía Bacteriana/inmunología , Infecciones por Pseudomonas/inmunología , Lesión Pulmonar Aguda/sangre , Lesión Pulmonar Aguda/microbiología , Lesión Pulmonar Aguda/patología , Animales , Modelos Animales de Enfermedad , Humanos , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Masculino , Ratones , Traumatismo Múltiple/sangre , Traumatismo Múltiple/diagnóstico , Traumatismo Múltiple/inmunología , Neutrófilos/metabolismo , Neumonía Bacteriana/sangre , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/patología , Infecciones por Pseudomonas/sangre , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa/inmunología , Especies Reactivas de Oxígeno/metabolismo , Índices de Gravedad del Trauma
16.
mSphere ; 6(3)2021 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-34011681

RESUMEN

Staphylococcus aureus is both a commensal and a pathogenic bacterium for humans. Its ability to induce severe infections is based on a wide range of virulence factors. S. aureus community-acquired pneumonia (SA-CAP) is rare and severe, and the contribution of certain virulence factors in this disease has been recognized over the past 2 decades. First, the factors involved in metabolism adaptation are crucial for S. aureus survival in the lower respiratory tract, and toxins and enzymes are required for it to cross the pulmonary epithelial barrier. S. aureus subsequently faces host defense mechanisms, including the epithelial barrier, but most importantly the immune system. Here, again, S. aureus uses myriad virulence factors to successfully escape from the host's defenses and takes advantage of them. The impact of S. aureus virulence, combined with the collateral damage caused by an overwhelming immune response, leads to severe tissue damage and adverse clinical outcomes. In this review, we summarize step by step all of the S. aureus factors implicated in CAP and described to date, and we provide an outlook for future research.


Asunto(s)
Neumonía Bacteriana/inmunología , Enfermedades Respiratorias/microbiología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Animales , Infecciones Comunitarias Adquiridas/inmunología , Infecciones Comunitarias Adquiridas/microbiología , Humanos , Ratones , Neumonía Bacteriana/patología , Enfermedades Respiratorias/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/genética , Staphylococcus aureus/inmunología , Virulencia , Factores de Virulencia
18.
Front Immunol ; 12: 654649, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33868297

RESUMEN

Extracellular traps released by neutrophils (NETs) are essential for the clearance of Pseudomonas aeruginosa. Alkaline protease (AprA) secreted by P. aeruginosa negatively correlates with clinical improvement. Moreover, anti-AprA in patients with cystic fibrosis (CF) can help identify patients with aggressive forms of chronic infection. However, the mechanism underlying the clinical outcomes remains unclear. We demonstrated that aprA deficiency in P. aeruginosa decreased the bacterial burden and reduced lung infection. AprA degraded NET components in vitro and in vivo but did not affect NET formation. Importantly, antibodies induced by AprA acted as an agonist and directly enhanced the degrading activities of AprA. Moreover, antisera from patients with P. aeruginosa infection exhibited antibody-dependent enhancement (ADE) similar to that of the antibodies we prepared. Our further investigations showed that the interaction between AprA and the specific antibodies might make the enzyme active sites better exposed, and subsequently enhance the recognition of substrates and accelerate the degradation. Our findings revealed that AprA secreted by P. aeruginosa may aggravate infection by destroying formed NETs, an effect that was further enhanced by its antibodies.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Endopeptidasas/inmunología , Trampas Extracelulares/inmunología , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/inmunología , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Modelos Animales de Enfermedad , Endopeptidasas/genética , Endopeptidasas/metabolismo , Trampas Extracelulares/enzimología , Femenino , Interacciones Huésped-Patógeno/inmunología , Humanos , Ratones , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/metabolismo , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/patología , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa/enzimología , Pseudomonas aeruginosa/patogenicidad
19.
Am J Respir Cell Mol Biol ; 64(5): 641-643, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33929292
20.
BMC Infect Dis ; 21(1): 364, 2021 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-33865326

RESUMEN

BACKGROUND: Parvimonas micra (P. micra) is a gram-positive anaerobic coccus that is detected widely on the skin, in the oral mucosa and in the gastrointestinal tract. In certain circumstances, P. micra can cause abdominal abscesses, bacteraemia and other infections. To the best of our knowledge, there have been no case reports describing the biological characteristics of P. micra-related pneumonia. These bacteria do not always multiply in an aerobic organ, such as the lung, and they could be easily overlooked because of the clinical mindset. CASE PRESENTATION: A 35-year-old pregnant woman was admitted to the emergency department 4 weeks prior to her due date who was exhibiting 5 points on the Glasgow coma scale. A computed tomography (CT) scan showed a massive haemorrhage in her left basal ganglia. She underwent a caesarean section and brain surgery before being admitted to the ICU. She soon developed severe pneumonia and hypoxemia. Given that multiple sputum cultures were negative, the patient's bronchoalveolar lavage fluid was submitted for next-generation sequencing (NGS) to determine the pathogen responsible for the pneumonia; as a result, P. micra was determined to be the causative pathogen. Accordingly the antibiotic therapy was altered and the pneumonia improved. CONCLUSION: In this case, we demonstrated severe pneumonia caused by the anaerobic organism P. micra, and the patient benefited from receiving the correct antibiotic. NGS was used as a method of quick diagnosis when sputum culture failed to distinguish the pathogen.


Asunto(s)
Firmicutes , Infecciones por Bacterias Grampositivas/complicaciones , Neumonía Bacteriana/microbiología , Complicaciones Infecciosas del Embarazo , Adulto , Antibacterianos/uso terapéutico , Cesárea , Coma/diagnóstico , Coma/microbiología , Coma/cirugía , Femenino , Firmicutes/aislamiento & purificación , Firmicutes/patogenicidad , Infecciones por Bacterias Grampositivas/diagnóstico , Infecciones por Bacterias Grampositivas/microbiología , Infecciones por Bacterias Grampositivas/cirugía , Humanos , Hemorragias Intracraneales/diagnóstico , Hemorragias Intracraneales/microbiología , Hemorragias Intracraneales/cirugía , Neumonía Bacteriana/diagnóstico , Neumonía Bacteriana/patología , Neumonía Bacteriana/cirugía , Embarazo , Complicaciones Infecciosas del Embarazo/diagnóstico , Complicaciones Infecciosas del Embarazo/microbiología , Tercer Trimestre del Embarazo , Índice de Severidad de la Enfermedad , Tomografía Computarizada por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...