Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 400
Filtrar
1.
Front Immunol ; 14: 1268078, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37954603

RESUMO

Introduction: Alveolar epithelial regeneration depends on the activity of resident quiescent progenitor cells. Alveolar epithelial type II (AT2) cells are known as the alveolar epithelial progenitor cells. They exit quiescent state, proliferate rapidly in response to injury and differentiate into alveolar epithelial type I (AT1) cells to regenerate the damaged alveolar epithelium. Although AT2 cell plasticity has been a very intense field of research, the role of CD8 T cell response and their released cytokine IFN-γ, in regulating AT2 cell plasticity and alveolar epithelial repair and regeneration after injury remains largely unknown. Methods: We used flow cytometry to quantify the amount of CD8 T cells in mouse lungs after bacterial pneumonia caused by Streptococcus pneumoniae. To determine whether CD8 T cells and their released cytokine IFN-γ are necessary for AT2 cell activity during alveolar epithelial regeneration, we performed loss of function studies using anti-CD8 or anti-IFN-γ monoclonal antibody (mAb) treatment in vivo. We assessed the effects of CD8 T cells and cytokine IFN-γ on AT2 cell differentiation capacity using the AT2- CD8 T cell co-culture system in vitro. Results: We detected a transient wave of accumulation of CD8 T cells in mouse lungs, which coincided with the burst of AT2 cell proliferation during alveolar epithelial repair and regeneration in mice following bacterial pneumonia caused by Streptococcus pneumoniae. Depletion of CD8 T cells or neutralization of cytokine IFN-γ using anti-CD8 or anti-IFN-γ monoclonal antibody significantly reduced AT2 cell proliferation and differentiation into AT1 cells in mice after bacterial pneumonia. Furthermore, co-culture of CD8 T cells or cytokine IFN-γ with AT2 cells promoted AT2-to-AT1 cell differentiation in both murine and human systems. Conversely, blockade of IFN-γ signaling abrogated the increase in AT2-to-AT1 cell differentiation in the AT2- CD8 T cell co-culture system. Discussion: Our data demonstrate that CD8 T-cell response and cytokine IFN-γ are necessary for promoting AT2 cell activity during alveolar epithelial repair and regeneration after acute lung injury caused by bacterial pneumonia.


Assuntos
Lesão Pulmonar Aguda , Pneumonia Bacteriana , Animais , Humanos , Camundongos , Lesão Pulmonar Aguda/metabolismo , Células Epiteliais Alveolares/metabolismo , Anticorpos Monoclonais/farmacologia , Citocinas/metabolismo , Interferon gama/metabolismo , Pneumonia Bacteriana/metabolismo
2.
Am J Physiol Lung Cell Mol Physiol ; 325(3): L277-L287, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37431588

RESUMO

Failure of the lung's endothelial barrier underlies lung injury, which causes the high mortality acute respiratory distress syndrome (ARDS). Multiple organ failure predisposes to the mortality, but mechanisms are poorly understood. Here, we show that mitochondrial uncoupling protein 2 (UCP2), a component of the mitochondrial inner membrane, plays a role in the barrier failure. Subsequent lung-liver cross talk mediated by neutrophil activation causes liver congestion. We intranasally instilled lipopolysaccharide (LPS). Then, we viewed the lung endothelium by real-time confocal imaging of the isolated, blood-perfused mouse lung. LPS caused alveolar-capillary transfer of reactive oxygen species and mitochondrial depolarization in lung venular capillaries. The mitochondrial depolarization was inhibited by transfection of alveolar Catalase and vascular knockdown of UCP2. LPS instillation caused lung injury as indicated by increases in bronchoalveolar lavage (BAL) protein content and extravascular lung water. LPS or Pseudomonas aeruginosa instillation also caused liver congestion, quantified by liver hemoglobin and plasma aspartate aminotransferase (AST) increases. Genetic inhibition of vascular UCP2 prevented both lung injury and liver congestion. Antibody-mediated neutrophil depletion blocked the liver responses, but not lung injury. Knockdown of lung vascular UCP2 mitigated P. aeruginosa-induced mortality. Together, these data suggest a mechanism in which bacterial pneumonia induces oxidative signaling to lung venular capillaries, known sites of inflammatory signaling in the lung microvasculature, depolarizing venular mitochondria. Successive activation of neutrophils induces liver congestion. We conclude that oxidant-induced UCP2 expression in lung venular capillaries causes a mechanistic sequence leading to liver congestion and mortality. Lung vascular UCP2 may present a therapeutic target in ARDS.NEW & NOTEWORTHY We report that mitochondrial injury in lung venular capillaries underlies barrier failure in pneumonia, and venular capillary uncoupling protein 2 (UCP2) causes neutrophil-mediated liver congestion. Using in situ imaging, we found that epithelial-endothelial transfer of H2O2 activates UCP2, depolarizing mitochondria in venular capillaries. The conceptual advance from our findings is that mitochondrial depolarization in lung capillaries mediates liver cross talk through circulating neutrophils. Pharmacologic blockade of UCP2 could be a therapeutic strategy for lung injury.


Assuntos
Lesão Pulmonar , Pneumonia Bacteriana , Síndrome do Desconforto Respiratório , Camundongos , Animais , Lipopolissacarídeos/farmacologia , Lipopolissacarídeos/metabolismo , Capilares/metabolismo , Peróxido de Hidrogênio , Fígado/metabolismo , Mitocôndrias/metabolismo , Síndrome do Desconforto Respiratório/metabolismo , Lesão Pulmonar/metabolismo , Pneumonia Bacteriana/metabolismo , Proteínas Mitocondriais/metabolismo
3.
Sci Rep ; 13(1): 4283, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36922640

RESUMO

Bacterial pulmonary infections are a major cause of morbidity and mortality in neonates, with less severity in older children. Previous studies demonstrated that the DNA of CD4+ T cells in the mouse lung, whose primary responsibility is to coordinate the immune response to foreign pathogens, is differentially methylated in neonates compared with juveniles. Nevertheless, the effect of this differential DNA methylation on CD4+ T cell gene expression and response to infection remains unclear. Here we treated E. coli-infected neonatal (4-day-old) and juvenile (13-day-old) mice with decitabine (DAC), a DNA methyltransferase inhibitor with broad-spectrum DNA demethylating activity, and performed simultaneous genome-wide DNA methylation and transcriptional profiling on lung CD4+ T cells. We show that juvenile and neonatal mice experienced differential demethylation in response to DAC treatment, with larger methylation differences observed in neonates. By cross-filtering differentially expressed genes between juveniles and neonates with those sites that were demethylated in neonates, we find that interferon-responsive genes such as Ifit1 are the most down-regulated methylation-sensitive genes in neonatal mice. DAC treatment shifted neonatal lung CD4+ T cells toward a gene expression program similar to that of juveniles. Following lung infection with E. coli, lung CD4+ T cells in neonatal mice exhibit epigenetic repression of important host defense pathways, which are activated by inhibition of DNA methyltransferase activity to resemble a more mature profile.


Assuntos
Infecções por Escherichia coli , Pneumonia Bacteriana , Animais , Camundongos , Linfócitos T/metabolismo , Escherichia coli/genética , Animais Recém-Nascidos , Pulmão/metabolismo , Pneumonia Bacteriana/metabolismo , Metilases de Modificação do DNA/genética , Infecções por Escherichia coli/genética , Metilação de DNA , Linfócitos T CD4-Positivos , Expressão Gênica
4.
Mol Immunol ; 153: 60-74, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36444819

RESUMO

Mycoplasma pneumoniae (M. pneumoniae) is the most common pathogen of respiratory tract infections in both children and adults. M. pneumoniae P1 adhesin plays an important role in the pathogenesis of M. pneumoniae infection by mediating the attachment of pathogen to host cells. The inoculation of C-terminal residuals of P1 (P1-C) showed a protective role from M. pneumoniae infection. Accumulated evidence suggests that the Wnt/ß-Catenin signaling is implicated in regulation of inflammatory responses to bacterial infections. However, mechanisms underlying the regulatory roles of Wnt signaling in host cells in response to M. pneumoniae infections are incompletely understood. In the present study, the impact and molecular mechanism of Wnt/ß-catenin signaling in immune responses induced by M. pneumoniae P1-C were investigated. The results demonstrated that the P1-C could activate Wnt/ß-catenin and Toll-like receptor (TLR) signaling in primary mouse airway epithelial cells cultured in an air-liquid interface (ALI) state. Interestingly, the inhibition of Wnt/ß-catenin signaling by an adenovirus-mediated Wnt inhibitor Dickkopf-1 (Dkk1) gene transduction alleviated the P1-C induced inflammation fibrosis in mouse lung, accompanied by the reduced expression of epithelial mesenchymal transition (EMT) markers. Mechanistical analysis further demonstrated that the Dkk1 could suppress the expression of JAK2/STAT1-STAT3 and Caspase3, 8/Bax signaling in mouse lung tissues. In vitro study further revealed that XAV939, a small molecule of Wnt/ß-catenin inhibitor, inhibited the P1-C-activated TLR4/MyD88 signaling and cytokine productions in primary mouse airway ALI epithelial cells. This study thus provides an insight into the function of Wnt/ß-catenin signaling in regulation of the pathogenesis of M. pneumoniae infection, suggesting that targeting Wnt/ß-catenin signaling by gene transduction of Dkk1, or pharmacological molecules of inhibitor may be a promised approach that worthy of further investigation in the treatment of M. pneumoniae pneumonia.


Assuntos
Mycoplasma pneumoniae , Pneumonia Bacteriana , Via de Sinalização Wnt , Animais , Camundongos , beta Catenina/metabolismo , Células Epiteliais/metabolismo , Inflamação/metabolismo , Pulmão/metabolismo , Mycoplasma pneumoniae/metabolismo , Pneumonia Bacteriana/metabolismo
5.
Cells ; 11(23)2022 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-36497202

RESUMO

Acute respiratory distress syndrome (ARDS) due to pulmonary infections is associated with high morbidity and mortality. Upon inflammation, the alarmin S100A8/A9 is released and stimulates neutrophil recruitment mainly via binding to Toll-like receptor 4 (TLR4). TLR4 is also expressed on platelets, which modulate the immune response through direct interaction with leukocytes. In a murine model of Klebsiella pneumoniae-induced pulmonary inflammation, global S100A9 deficiency resulted in diminished neutrophil recruitment into the lung alveoli and neutrophil accumulation in the intravascular space, indicating an impaired neutrophil migration. A lack of TLR4 on platelets resulted in reduced neutrophil counts in the whole lung, emphasising the impact of TLR4-mediated platelet activity on neutrophil behaviour. Flow cytometry-based analysis indicated elevated numbers of platelet-neutrophil complexes in the blood of S100A9-/- mice. Intravital microscopy of the murine cremaster muscle confirmed these findings and further indicated a significant increase in neutrophil-platelet complex formation in S100A9-/- mice, which was reversed by administration of the S100A8/A9 tetramer. An in vitro bilayer assay simulated the murine alveolar capillary barrier during inflammation and validated significant differences in transmigration behaviour between wild-type and S100A9-/- neutrophils. This study demonstrates the role of S100A8/A9 during platelet-neutrophil interactions and neutrophil recruitment during pulmonary inflammation.


Assuntos
Calgranulina A , Calgranulina B , Neutrófilos , Pneumonia Bacteriana , Animais , Camundongos , Alarminas/metabolismo , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Inflamação/metabolismo , Infiltração de Neutrófilos , Neutrófilos/metabolismo , Camundongos Knockout , Pneumonia Bacteriana/metabolismo
6.
Cells ; 11(15)2022 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-35892600

RESUMO

Severe epithelial dysfunction is one major hallmark throughout the pathophysiological progress of bacterial pneumonia. Junctional and cellular adhesion molecules (e.g., JAMA-A, ICAM-1), cytokines (e.g., TNFα), and growth factors (e.g., TGFα), controlling proper lung barrier function and leukocyte recruitment, are proteolytically cleaved and released into the extracellular space through a disintegrin and metalloproteinase (ADAM) 17. In cell-based assays, we could show that the protein expression, maturation, and activation of ADAM17 is upregulated upon infection of lung epithelial cells with Pseudomonas aeruginosa and Exotoxin A (ExoA), without any impact of infection by Streptococcus pneumoniae. The characterization of released extracellular vesicles/exosomes and the comparison to heat-inactivated bacteria revealed that this increase occurred in a cell-associated and toxin-dependent manner. Pharmacological targeting and gene silencing of ADAM17 showed that its activation during infection with Pseudomonas aeruginosa was critical for the cleavage of junctional adhesion molecule A (JAM-A) and epithelial cell survival, both modulating barrier integrity, epithelial regeneration, leukocyte adhesion and transepithelial migration. Thus, site-specific targeting of ADAM17 or blockage of the activating toxins may constitute a novel anti-infective therapeutic option in Pseudomonas aeruginosa lung infection preventing severe epithelial and organ dysfunctions and stimulating future translational studies.


Assuntos
Proteína ADAM17 , Pneumonia Bacteriana , Infecções por Pseudomonas , Proteína ADAM17/metabolismo , Células Epiteliais/metabolismo , Humanos , Pulmão/microbiologia , Pneumonia Bacteriana/metabolismo , Pseudomonas aeruginosa
7.
Phytother Res ; 36(4): 1822-1835, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35233841

RESUMO

Bacterial pneumonia is one of the most important causes of mortality in the United States. The bacteria Klebsiella pneumoniae (KP) accounts for a significant proportion of community and hospital-acquired infections. Here, we determine that the holy basil (Ocimum sanctum) extract improves cell viability and dampens the proinflammatory cytokine response in an in vitro model of pneumonia. For this, A549, a human alveolar basal epithelial cell line, was subjected to a lethal KP model following a 24-hr pretreatment with basil extract. Bacteremia, cell viability, apoptosis, MTT assay, phagocytic capacity, cytokines, and Khe gene expression were assessed in these cells following pneumonia. Cell morphology analysis showed that holy basil protected A549 cells from KP infection-mediated effects by inhibiting cell death due to apoptosis. Additionally, in the presence of basil, A549 cells demonstrated significantly higher bactericidal capacity and phagocytosis. Administration of holy basil led to reduced expression of hypoxia-inducible factor-1/2a, nuclear factor kappa B, and Khe in the KP-infected cells while increasing interferon (IFN)-γ expression. Our results suggest that basil significantly reduced cell death in the setting of KP infection, likely via attenuation of cytokine and IFN-γ mediated signaling pathways. Holy basil is a promising therapeutic agent for managing and treating bacterial pneumonia based on its potency.


Assuntos
Óleos Voláteis , Pneumonia Bacteriana , Células Epiteliais Alveolares/metabolismo , Humanos , Interferon gama/uso terapêutico , NF-kappa B/metabolismo , Ocimum sanctum , Óleos Voláteis/farmacologia , Óleos Voláteis/uso terapêutico , Pneumonia Bacteriana/tratamento farmacológico , Pneumonia Bacteriana/metabolismo , Pneumonia Bacteriana/microbiologia
8.
Infect Immun ; 90(3): e0049121, 2022 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-35130455

RESUMO

Neutrophils are capable of extruding neutrophil extracellular traps (NETs), a network of granule proteins and chromatin material, upon activation. NETs provide defense against extracellular microbes, but histones in NETs can also induce cytotoxicity and activate inflammatory responses. The relevance of NETs to bacterial pneumonias is beginning to be defined. In the present study, we found that the extracellular concentration of citrullinated histone H3, a component of NETs, was elevated in bronchoalveolar lavage fluid recovered from mice with diverse bacterial pneumonias and correlated with neutrophil infiltration and cell death in the lungs as well as levels of H4. Because the histone H4 component of NETs is sufficient to stimulate inflammation, we tested its effects in the air spaces of the lungs. Recombinant histone H4 in the noninflamed lung produced only modest effects, but in the setting of neutrophilic inflammation, H4 substantially increased pulmonary neutrophils, NETs, necrosis, and edema. However, blockade of histone H4 with a monoclonal antibody during pneumonia did not significantly alter measures of lung damage. Taken together, these results implicate NETs and extracellular histone H4 in exacerbating the lung injury resulting from bacterial pneumonia.


Assuntos
Armadilhas Extracelulares , Pneumonia Bacteriana , Animais , Armadilhas Extracelulares/metabolismo , Histonas/metabolismo , Inflamação/metabolismo , Camundongos , Neutrófilos , Pneumonia Bacteriana/metabolismo
9.
JCI Insight ; 7(4)2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35050901

RESUMO

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.


Assuntos
Células Epiteliais Alveolares/metabolismo , Proteínas de Ligação ao Cálcio/genética , Cálcio/metabolismo , Regulação da Expressão Gênica , Proteínas de Transporte da Membrana Mitocondrial/genética , Pneumonia Bacteriana/genética , RNA/genética , Regeneração/genética , Células Epiteliais Alveolares/patologia , Animais , Transporte Biológico , Proteínas de Ligação ao Cálcio/biossíntese , Diferenciação Celular , Plasticidade Celular , Células Cultivadas , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Pneumonia Bacteriana/metabolismo , Pneumonia Bacteriana/patologia
10.
J Biol Chem ; 298(1): 101482, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34896150

RESUMO

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.


Assuntos
Pneumopatias , Pneumonia Bacteriana , Tauopatias , Proteínas tau , Animais , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/microbiologia , Disfunção Cognitiva/patologia , Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Células Endoteliais/patologia , Humanos , Pulmão/irrigação sanguínea , Pneumopatias/metabolismo , Pneumopatias/microbiologia , Pneumopatias/patologia , Camundongos , Pneumonia Bacteriana/metabolismo , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/patologia , Isoformas de Proteínas , Pseudomonas aeruginosa , Tauopatias/genética , Tauopatias/metabolismo , Tauopatias/patologia , Proteínas tau/química , Proteínas tau/genética , Proteínas tau/metabolismo
11.
Int J Mol Sci ; 22(21)2021 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-34768890

RESUMO

Gram-negative (G-) bacteria are the leading cause of hospital-acquired pneumonia in the United States. The devastating damage caused by G- bacteria results from the imbalance of bactericidal effects and overwhelming inflammation. Despite decades of research, the underlying mechanisms by which runaway inflammation is developed remain incompletely understood. Clara Cell Protein 16 (CC16), also known as uteroglobin, is the major protein secreted by Clara cells and the most abundant protein in bronchoalveolar lavage fluid (BALF). However, the regulation and functions of CC16 during G- bacterial infection are unknown. In this study, we aimed to assess the regulation of CC16 in response to Klebsiella pneumoniae (K. pneu) and to investigate the role of CC16 in bronchial epithelial cells. After K. pneu infection, we found that CC16 mRNA expression was significantly decreased in bronchial epithelial cells. Our data also showed that K. pneu infection upregulated cytokine and chemokine genes, including IL-1ß, IL-6, and IL-8 in BEAS-2B cells. Endogenously overexpressed CC16 in BEAS-2B cells provided an anti-inflammatory effect by reducing these markers. We also observed that endogenous CC16 can repress NF-κB reporter activity. In contrast, the recombinant CC16 (rCC16) did not show an anti-inflammatory effect in K. pneu-infected cells or suppression of NF-κB promoter activity. Moreover, the overexpression of CC16 reduced reactive oxygen species (ROS) levels and protected BEAS-2B cells from K. pneu-induced apoptosis.


Assuntos
Inflamação/metabolismo , Pneumonia Bacteriana/metabolismo , Uteroglobina , Apoptose , Brônquios/citologia , Brônquios/microbiologia , Líquido da Lavagem Broncoalveolar/química , Citocinas/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Imunidade Inata , Klebsiella pneumoniae , Pulmão/microbiologia , Pulmão/patologia , NF-kappa B/metabolismo , Uteroglobina/genética , Uteroglobina/metabolismo
12.
Sci Rep ; 11(1): 22725, 2021 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-34811449

RESUMO

We previously reported that flagellin-expressing Pseudomonas aeruginosa (Pa) provokes NEU1 sialidase-mediated MUC1 ectodomain (MUC1-ED) desialylation and MUC1-ED shedding from murine lungs in vivo. Here, we asked whether Pa in the lungs of patients with ventilator-associated pneumonia might also increase MUC1-ED shedding. The levels of MUC1-ED and Pa-expressed flagellin were dramatically elevated in bronchoalveolar lavage fluid (BALF) harvested from Pa-infected patients, and each flagellin level, in turn, predicted MUC1-ED shedding in the same patient. Desialylated MUC1-ED was only detected in BALF of Pa-infected patients. Clinical Pa strains increased MUC1-ED shedding from cultured human alveolar epithelia, and FlaA and FlaB flagellin-expressing strains provoked comparable levels of MUC1-ED shedding. A flagellin-deficient isogenic mutant generated dramatically reduced MUC1-ED shedding compared with the flagellin-expressing wild-type strain, and purified FlaA and FlaB recapitulated the effect of intact bacteria. Pa:MUC1-ED complexes were detected in the supernatants of alveolar epithelia exposed to wild-type Pa, but not to the flagellin-deficient Pa strain. Finally, human recombinant MUC1-ED dose-dependently disrupted multiple flagellin-driven processes, including Pa motility, Pa biofilm formation, and Pa adhesion to human alveolar epithelia, while enhancing human neutrophil-mediated Pa phagocytosis. Therefore, shed desialylated MUC1-ED functions as a novel flagellin-targeting, Pa-responsive decoy receptor that participates in the host response to Pa at the airway epithelial surface.


Assuntos
Flagelina/metabolismo , Pulmão/metabolismo , Mucina-1/metabolismo , Pneumonia Bacteriana/metabolismo , Pneumonia Associada à Ventilação Mecânica/metabolismo , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/metabolismo , Células A549 , Idoso , Biomarcadores/metabolismo , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/microbiologia , Feminino , Flagelina/genética , Interações Hospedeiro-Patógeno , Humanos , Pulmão/microbiologia , Masculino , Pessoa de Meia-Idade , Mutação , Neuraminidase/metabolismo , Pneumonia Bacteriana/diagnóstico , Pneumonia Bacteriana/microbiologia , Pneumonia Associada à Ventilação Mecânica/diagnóstico , Pneumonia Associada à Ventilação Mecânica/microbiologia , Infecções por Pseudomonas/diagnóstico , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidade
13.
Sci Rep ; 11(1): 21210, 2021 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-34707211

RESUMO

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.


Assuntos
Transição Epitelial-Mesenquimal , Pneumonia Bacteriana/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Infecções Estreptocócicas/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Actinas/metabolismo , Animais , Células Cultivadas , Epitélio/metabolismo , Epitélio/patologia , Fibrose , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Pleura/metabolismo , Pleura/patologia , Pneumonia Bacteriana/patologia , Infecções Estreptocócicas/patologia , Ativador de Plasminogênio Tipo Uroquinase/genética
14.
Lab Invest ; 101(11): 1467-1474, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34504306

RESUMO

The mortality rates among patients who initially survive sepsis are, in part, associated with a high risk of secondary lung infections and respiratory failure. Given that phagolysosomes are important for intracellular killing of pathogenic microbes, we investigated how severe lung infections associated with post-sepsis immunosuppression affect phagolysosome biogenesis. In mice with P. aeruginosa-induced pneumonia, we found a depletion of both phagosomes and lysosomes, as evidenced by decreased amounts of microtubule associated protein light chain 3-II (LC3-II) and lysosomal-associated membrane protein (LAMP1). We also found a loss of transcription factor E3 (TFE3) and transcription factor EB (TFEB), which are important activators for transcription of genes encoding autophagy and lysosomal proteins. These events were associated with increased expression of ZKSCAN3, a repressor for transcription of genes encoding autophagy and lysosomal proteins. Zkscan3-/- mice had increased expression of genes involved in the autophagy-lysosomal pathway along with enhanced killing of P. aeruginosa in the lungs, as compared to wild-type mice. These findings highlight the involvement of ZKSCAN3 in response to severe lung infection, including susceptibility to secondary bacterial infections due to immunosuppression.


Assuntos
Fagossomos/fisiologia , Pneumonia Bacteriana/complicações , Infecções por Pseudomonas/complicações , Sepse/imunologia , Fatores de Transcrição/deficiência , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Tolerância Imunológica , Pulmão/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Pneumonia Bacteriana/metabolismo , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa , Sepse/microbiologia
15.
Food Funct ; 12(19): 9466-9475, 2021 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-34473137

RESUMO

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.


Assuntos
Ácido Clorogênico/uso terapêutico , Tolerância Imunológica , Inflamassomos/metabolismo , Infecções por Klebsiella/tratamento farmacológico , Klebsiella pneumoniae , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Pneumonia Bacteriana/tratamento farmacológico , Animais , Ácido Clorogênico/farmacologia , Citocinas/genética , Citocinas/metabolismo , Infecções por Klebsiella/imunologia , Infecções por Klebsiella/metabolismo , Infecções por Klebsiella/patologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/metabolismo , Pneumonia Bacteriana/patologia , Células RAW 264.7 , Transdução de Sinais/efeitos dos fármacos
16.
Biochem Pharmacol ; 192: 114753, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34474040

RESUMO

Methicillin-resistant Staphylococcus aureus is one of the leading causes of community and nosocomial infections, which has created the urgent need for innovative anti-infective agents to control MRSA-associated infections. A conserved serine protease, caseinolytic peptidase P (ClpP) in Staphylococcus aureus is highly associated with pathogenicity and has been claimed to be a novel antimicrobial target. We aim to search suitable inhibitors of ClpP to attenuate the virulence of MRSA and combat their infections in vivo. Over 500 natural compounds were pre-screened via fluorescence resonance energy transfer using the Suc-LY-AMC substrate. The binding of myricetin to ClpP was determined and the mechanism of action was elucidated by thermal shift assay, surface plasmon resonance, and molecular dynamics simulations. The therapeutic effects of myricetin on S. aureus infection were further investigated using a S. aureus-induced pneumonia model. We revealed that myricetin could effectively block the activity of ClpP without disturbing the growth of the bacteria and the Gln-47 and Met-31 residues were necessary for myricetin binding to ClpP. Importantly, myricetin attenuated the pathogenicity of S. aureus in vivo, while improving the efficacy of the traditional antibiotic oxacillin against MRSA infection and protecting mice from fatal lung infections caused by MRSA. These findings indicate that myricetin has the potential to be applied in the pharmaceutical industry as a promising therapeutic agent.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Endopeptidase Clp/metabolismo , Flavonoides/metabolismo , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Pneumonia Bacteriana/metabolismo , Infecções Estafilocócicas/metabolismo , Animais , Relação Dose-Resposta a Droga , Feminino , Flavonoides/administração & dosagem , Células Hep G2 , Humanos , Staphylococcus aureus Resistente à Meticilina/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia Bacteriana/prevenção & controle , Infecções Estafilocócicas/prevenção & controle
17.
Mol Med ; 27(1): 79, 2021 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-34271850

RESUMO

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.


Assuntos
Fibrose Cística/complicações , Fibrose Cística/metabolismo , Proteína HMGB1/genética , Heparina/análogos & derivados , Macrófagos/imunologia , Macrófagos/metabolismo , Pneumonia Bacteriana/etiologia , Pneumonia Bacteriana/metabolismo , Animais , Carga Bacteriana , Biomarcadores , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Proteína HMGB1/química , Proteína HMGB1/metabolismo , Heparina/química , Heparina/metabolismo , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Modelos Moleculares , Óxido Nítrico/metabolismo , Fagocitose/imunologia , Pneumonia Bacteriana/patologia , Ligação Proteica , Células RAW 264.7 , Relação Estrutura-Atividade
18.
Microbiol Spectr ; 9(1): e0020721, 2021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34319124

RESUMO

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.


Assuntos
Pulmão/microbiologia , Staphylococcus aureus Resistente à Meticilina/fisiologia , Pneumonia Bacteriana/metabolismo , Infecções Estafilocócicas/metabolismo , Tenascina/metabolismo , Animais , Feminino , Humanos , Pulmão/metabolismo , Pulmão/patologia , Masculino , Staphylococcus aureus Resistente à Meticilina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumonia Bacteriana/genética , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/patologia , Infecções Estafilocócicas/genética , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Tenascina/genética
19.
PLoS One ; 16(6): e0252378, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34086721

RESUMO

Diagnosis of microbial disease etiology in community-acquired pneumonia (CAP) remains challenging. We undertook a large-scale metabolomics study of serum samples in hospitalized CAP patients to determine if host-response associated metabolites can enable diagnosis of microbial etiology, with a specific focus on discrimination between the major CAP pathogen groups S. pneumoniae, atypical bacteria, and respiratory viruses. Targeted metabolomic profiling of serum samples was performed for three groups of hospitalized CAP patients with confirmed microbial etiologies: S. pneumoniae (n = 48), atypical bacteria (n = 47), or viral infections (n = 30). A wide range of 347 metabolites was targeted, including amines, acylcarnitines, organic acids, and lipids. Single discriminating metabolites were selected using Student's T-test and their predictive performance was analyzed using logistic regression. Elastic net regression models were employed to discover metabolite signatures with predictive value for discrimination between pathogen groups. Metabolites to discriminate S. pneumoniae or viral pathogens from the other groups showed poor predictive capability, whereas discrimination of atypical pathogens from the other groups was found to be possible. Classification of atypical pathogens using elastic net regression models was associated with a predictive performance of 61% sensitivity, 86% specificity, and an AUC of 0.81. Targeted profiling of the host metabolic response revealed metabolites that can support diagnosis of microbial etiology in CAP patients with atypical bacterial pathogens compared to patients with S. pneumoniae or viral infections.


Assuntos
Infecções Comunitárias Adquiridas/metabolismo , Metaboloma/fisiologia , Idoso , Bactérias/patogenicidade , Doenças Transmissíveis/metabolismo , Doenças Transmissíveis/microbiologia , Doenças Transmissíveis/virologia , Infecções Comunitárias Adquiridas/microbiologia , Infecções Comunitárias Adquiridas/virologia , Feminino , Hospitalização , Humanos , Masculino , Metabolômica , Pessoa de Meia-Idade , Infecções Pneumocócicas/metabolismo , Infecções Pneumocócicas/microbiologia , Pneumonia Bacteriana/metabolismo , Pneumonia Bacteriana/microbiologia , Streptococcus pneumoniae/patogenicidade , Vírus/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA