Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 345
Filter
Add more filters

Publication year range
1.
Am J Physiol Lung Cell Mol Physiol ; 327(2): L141-L149, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38772909

ABSTRACT

Neutrophils are the first leukocytes to be recruited to sites of inflammation in response to chemotactic factors released by activated macrophages and pulmonary epithelial and endothelial cells in bacterial pneumonia, a common cause of acute respiratory distress syndrome (ARDS). Although neutrophilic inflammation facilitates the elimination of pathogens, neutrophils also may cause bystander tissue injury. Even though the presence of neutrophils in alveolar spaces is a key feature of acute lung injury and ARDS especially from pneumonia, their contribution to the pathogenesis of lung injury is uncertain. The goal of this study was to elucidate the role of neutrophils in a clinically relevant model of bacterial pneumonia. We investigated the effect of reducing neutrophils in a mouse model of pneumococcal pneumonia treated with antibiotics. Neutrophils were reduced with anti-lymphocyte antigen 6 complex locus G6D (Ly6G) monoclonal antibody 24 h before and immediately preceding infection. Mice were inoculated intranasally with Streptococcus pneumoniae and received ceftriaxone 12 h after bacterial inoculation. Neutrophil reduction in mice treated with ceftriaxone attenuated hypoxemia, alveolar permeability, epithelial injury, pulmonary edema, and inflammatory biomarker release induced by bacterial pneumonia, even though bacterial loads in the distal air spaces of the lung were modestly increased as compared with antibiotic treatment alone. Thus, when appropriate antibiotics are administered, lung injury in the early phase of bacterial pneumonia is mediated in part by neutrophils. In the early phase of bacterial pneumonia, neutrophils contribute to the severity of lung injury, although they also participate in host defense.NEW & NOTEWORTHY Neutrophil accumulation is a key feature of ARDS, but their contribution to the pathogenesis is still uncertain. We investigated the effect of reducing neutrophils in a clinically relevant mouse model of pneumococcal pneumonia treated with antibiotics. When appropriate antibiotics were administered, neutrophil reduction with Ly6G antibody markedly attenuated lung injury and improved oxygenation. In the early phase of bacterial pneumonia, neutrophils contribute to the severity of lung injury, although they also participate in host defense.


Subject(s)
Mice, Inbred C57BL , Neutrophils , Pneumonia, Pneumococcal , Animals , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/pathology , Pneumonia, Pneumococcal/drug therapy , Pneumonia, Pneumococcal/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Mice , Streptococcus pneumoniae/pathogenicity , Acute Lung Injury/pathology , Acute Lung Injury/immunology , Acute Lung Injury/drug therapy , Acute Lung Injury/microbiology , Disease Models, Animal , Lung/pathology , Lung/immunology , Lung/metabolism , Lung/drug effects , Lung Injury/pathology , Lung Injury/immunology , Lung Injury/drug therapy , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/immunology , Male
2.
BMC Infect Dis ; 24(1): 414, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38641804

ABSTRACT

BACKGROUND: Lobar pneumonia caused by Mycoplasma pneumoniae is a relatively difficult-to-treat pneumonia in children. The time of radiographic resolution after treatment is variable, a long recovery time can result in several negative effects, and it has attracted our attention. Therefore, exploring factors associated with delayed radiographic resolution will help to identify these children at an early stage and prepare for early intervention. METHODS: The data of 339 children with lobar pneumonia caused by Mycoplasma pneumoniae were collected from the Department of Pediatrics of Fu Yang People's Hospital, China from January 2021 to June 2022. After discharge, the children were regularly followed up in the outpatient department and on the WeChat platform for > 8 weeks. According to whether pulmonary imaging (chest radiography or plain chest computed tomography) returned to normal within 8 weeks, the children were divided into the delayed recovery group (DRG) (n = 69) and the normal recovery group (NRG) (n = 270). The children's general information, laboratory examination findings, bronchoscopy results, and imaging findings were retrospectively analyzed. Single-factor analysis was performed to identify the risk factors for delayed radiographic resolution of lobar pneumonia caused by Mycoplasma pneumoniae, and the factors with statistically significant differences underwent multiple-factor logistic regression analysis. Receiver operating characteristic (ROC) analysis was then performed to calculate the cutoff value of early predictive indicators of delayed radiographic resolution. RESULTS: Single-factor analysis showed that the following were significantly greater in the DRG than NRG: total fever duration, the hospitalization time, C-reactive protein (CRP) level, lactate dehydrogenase (LDH) level, D-dimer level, pulmonary lesions involving two or more lobes, a large amount of pleural effusion, the time to interventional bronchoscopy, and mucus plugs formation. Multivariate logistic regression analysis showed that the hospitalization time, CRP level, LDH level, pulmonary lesions involving two or more lobes, and a large amount of pleural effusion were independent risk factors for delayed radiographic resolution of lobar pneumonia caused by Mycoplasma pneumoniae. The cutoff values on the receiver operating characteristic curve were a hospitalization time of ≥ 10.5 days, CRP level of ≥ 25.92 mg/L, and LDH level of ≥ 378 U/L. CONCLUSION: If patients with lobar pneumonia caused by Mycoplasma pneumoniae have a hospitalization time of ≥ 10.5 days, CRP level of ≥ 25.92 mg/L, and LDH level ≥ 378 U/L, the time of radiographic resolution is highly likely to exceed 8 weeks. Pediatricians must maintain a high level of vigilance for these factors, control the infection as early as possible, strengthen airway management, and follow up closely to avoid complications and sequelae of Mycoplasma pneumoniae pneumonia.


Subject(s)
Pleural Effusion , Pneumonia, Mycoplasma , Pneumonia, Pneumococcal , Child , Humans , Mycoplasma pneumoniae , Retrospective Studies , Pneumonia, Mycoplasma/complications , Lung/diagnostic imaging , Lung/pathology , Pneumonia, Pneumococcal/pathology , Pleural Effusion/complications
3.
J Immunol ; 205(12): 3390-3399, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33158955

ABSTRACT

Macrophage-inducible C-type lectin (Mincle)-dependent sensing of pathogens triggers proinflammatory immune responses in professional phagocytes that contribute to protecting the host against pathogen invasion. In this study, we examined whether overexpression of Mincle that is designed to improve early pathogen sensing by professional phagocytes would improve lung-protective immunity against Streptococcus pneumoniae in mice. Proteomic profiling of alveolar macrophages of Mincle transgenic (tg) mice stimulated with the Mincle-specific pneumococcal ligand glucosyl-diacylglycerol (Glc-DAG) revealed increased Nlrp3 inflammasome activation and downstream IL-1ß cytokine release that was not observed in Glc-DAG-stimulated Mincle knockout or Nlrp3 knockout macrophages. Along this line, Mincle tg mice also responded with a stronger Nlrp3 expression and early proinflammatory cytokine release after challenge with S. pneumoniae, ultimately leading to fatal pneumonia in the Mincle tg mice. Importantly, Nlrp3 inhibitor treatment of Mincle tg mice significantly mitigated the observed hyperinflammatory response to pneumococcal challenge. Together, we show that overexpression of the pattern recognition receptor Mincle triggers increased Glc-DAG-dependent Nlrp3 inflammasome activation in professional phagocytes leading to fatal pneumococcal pneumonia in mice that is amenable to Nlrp3 inhibitor treatment. These data show that ectopic expression of the Mincle receptor confers increased susceptibility rather than resistance to S. pneumoniae in mice, thus highlighting the importance of an inducible Mincle receptor expression in response to microbial challenge.


Subject(s)
Lectins, C-Type/immunology , Macrophages, Alveolar/immunology , Membrane Proteins/immunology , Pneumonia, Pneumococcal/immunology , Streptococcus pneumoniae/immunology , Animals , Inflammasomes/genetics , Inflammasomes/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Lectins, C-Type/genetics , Macrophages, Alveolar/pathology , Membrane Proteins/genetics , Mice , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Pneumonia, Pneumococcal/genetics , Pneumonia, Pneumococcal/pathology
4.
Am J Respir Cell Mol Biol ; 64(4): 477-491, 2021 04.
Article in English | MEDLINE | ID: mdl-33600743

ABSTRACT

Streptococcus pneumoniae is the leading cause of hospital community-acquired pneumonia. Patients with pneumococcal pneumonia may develop complicated parapneumonic effusions or empyema that can lead to pleural organization and subsequent fibrosis. The pathogenesis of pleural organization and scarification involves complex interactions between the components of the immune system, coagulation, and fibrinolysis. EPCR (endothelial protein C receptor) is a critical component of the protein C anticoagulant pathway. The present study was performed to evaluate the role of EPCR in the pathogenesis of S. pneumoniae infection-induced pleural thickening and fibrosis. Our studies show that the pleural mesothelium expresses EPCR. Intrapleural instillation of S. pneumoniae impairs lung compliance and lung volume in wild-type and EPCR-overexpressing mice but not in EPCR-deficient mice. Intrapleural S. pneumoniae infection induces pleural thickening in wild-type mice. Pleural thickening is more pronounced in EPCR-overexpressing mice, whereas it is reduced in EPCR-deficient mice. Markers of mesomesenchymal transition are increased in the visceral pleura of S. pneumoniae-infected wild-type and EPCR-overexpressing mice but not in EPCR-deficient mice. The lungs of wild-type and EPCR-overexpressing mice administered intrapleural S. pneumoniae showed increased infiltration of macrophages and neutrophils, which was significantly reduced in EPCR-deficient mice. An analysis of bacterial burden in the pleural lavage, the lungs, and blood revealed a significantly lower bacterial burden in EPCR-deficient mice compared with wild-type and EPCR-overexpressing mice. Overall, our data provide strong evidence that EPCR deficiency protects against S. pneumoniae infection-induced impairment of lung function and pleural remodeling.


Subject(s)
Endothelial Protein C Receptor/deficiency , Lung/metabolism , Pleura/metabolism , Pleural Effusion/metabolism , Pleurisy/metabolism , Pneumonia, Pneumococcal/metabolism , Streptococcus pneumoniae/pathogenicity , Animals , Bacterial Load , Cells, Cultured , Disease Models, Animal , Endothelial Protein C Receptor/genetics , Female , Fibrosis , Host-Pathogen Interactions , Humans , Lung/microbiology , Lung/pathology , Lung/physiopathology , Macrophages/metabolism , Macrophages/microbiology , Male , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Neutrophils/metabolism , Neutrophils/microbiology , Pleura/microbiology , Pleura/pathology , Pleural Effusion/microbiology , Pleural Effusion/pathology , Pleural Effusion/physiopathology , Pleurisy/microbiology , Pleurisy/pathology , Pleurisy/physiopathology , Pneumonia, Pneumococcal/microbiology , Pneumonia, Pneumococcal/pathology , Pneumonia, Pneumococcal/physiopathology
5.
Am J Respir Cell Mol Biol ; 64(4): 492-503, 2021 04.
Article in English | MEDLINE | ID: mdl-33513310

ABSTRACT

Pleural organization may occur after empyema or complicated parapneumonic effusion and can result in restrictive lung disease with pleural fibrosis (PF). Pleural mesothelial cells (PMCs) may contribute to PF through acquisition of a profibrotic phenotype, mesothelial-mesenchymal transition (MesoMT), which is characterized by increased expression of α-SMA (α-smooth muscle actin) and other myofibroblast markers. Although MesoMT has been implicated in the pathogenesis of PF, the role of the reactive oxygen species and the NOX (nicotinamide adenine dinucleotide phosphate oxidase) family in pleural remodeling remains unclear. Here, we show that NOX1 expression is enhanced in nonspecific human pleuritis and is induced in PMCs by THB (thrombin). 4-Hydroxy-2-nonenal, an indicator of reactive oxygen species damage, was likewise increased in our mouse model of pleural injury. NOX1 downregulation blocked THB- and Xa (factor Xa)-mediated MesoMT, as did pharmacologic inhibition of NOX1 with ML-171. NOX1 inhibition also reduced phosphorylation of Akt, p65, and tyrosine 216-GSK-3ß, signaling molecules previously shown to be implicated in MesoMT. Conversely, ML-171 did not reverse established MesoMT. NOX4 downregulation attenuated TGF-ß- and THB-mediated MesoMT. However, NOX1 downregulation did not affect NOX4 expression. NOX1- and NOX4-deficient mice were also protected in our mouse model of Streptococcus pneumoniae-mediated PF. These data show that NOX1 and NOX4 are critical determinants of MesoMT.


Subject(s)
Epithelial-Mesenchymal Transition , NADPH Oxidase 1/metabolism , Pleura/enzymology , Pleurisy/enzymology , Pneumonia, Pneumococcal/enzymology , Reactive Oxygen Species/metabolism , Streptococcus pneumoniae/pathogenicity , Animals , Cells, Cultured , Disease Models, Animal , Factor Xa/metabolism , Fibrosis , Host-Pathogen Interactions , Humans , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 1/deficiency , NADPH Oxidase 1/genetics , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , Pleura/microbiology , Pleura/pathology , Pleurisy/microbiology , Pleurisy/pathology , Pleurisy/physiopathology , Pneumonia, Pneumococcal/microbiology , Pneumonia, Pneumococcal/pathology , Signal Transduction , Thrombin/metabolism
6.
Am J Physiol Lung Cell Mol Physiol ; 320(5): L916-L925, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33655757

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a deadly condition characterized by progressive respiratory dysfunction. Exacerbations due to airway infections are believed to promote disease progression, and presence of Streptococcus in the lung microbiome has been associated with the progression of IPF and mortality. The aim of this study was to analyze the effect of lung fibrosis on susceptibility to pneumococcal pneumonia and bacteremia. The effects of subclinical (low dose) infection with Streptococcus pneumoniae were studied in a well characterized fos-related antigen-2 (Fra-2) transgenic (TG) mouse model of spontaneous, progressive pulmonary fibrosis. Forty-eight hours after transnasal infection with S. pneumoniae, bacterial load was assessed in lung tissue, bronchoalveolar lavage (BAL), blood, and spleen. Leukocyte subsets and cytokine levels were analyzed in BAL and blood. Lung compliance and arterial blood gases were assessed. In contrast to wildtype mice, low dose lung infection with S. pneumoniae in Fra-2 TG mice resulted in substantial pneumonia including weight loss, increased lung bacterial load, and bacteremia. BAL alveolar macrophages were reduced in Fra-2 TG mice compared to the corresponding WT mice. Proinflammatory cytokines and chemokines (IL-1ß, IL-6, TNF-α, and CXCL1) were elevated upon infection in BAL supernatant and plasma of Fra-2 TG mice. Lung compliance was decreased in Fra-2 TG mice following low dose infection with S. pneumoniae. Pulmonary fibrosis increases susceptibility to pneumococcal pneumonia and bacteremia possibly via impaired alveolar bacterial clearance.


Subject(s)
Fos-Related Antigen-2 , Macrophages, Alveolar , Pneumonia, Pneumococcal , Pulmonary Fibrosis , Streptococcus pneumoniae/metabolism , Animals , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Fos-Related Antigen-2/genetics , Fos-Related Antigen-2/metabolism , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/pathology , Mice , Mice, Transgenic , Pneumonia, Pneumococcal/genetics , Pneumonia, Pneumococcal/metabolism , Pneumonia, Pneumococcal/microbiology , Pneumonia, Pneumococcal/pathology , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/microbiology , Pulmonary Fibrosis/pathology
7.
Am J Physiol Lung Cell Mol Physiol ; 320(3): L377-L392, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33296268

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease (ILD) associated with high morbidity and mortality. Patients with ILD frequently develop an acute exacerbation of their disease, which may be triggered by viral and/or bacterial infections. Prostaglandin E2 (PGE2) is an eicosanoid released in a cyclooxygenase-2 (COX2)-dependent manner and is considered to contribute to regulation of lung fibrosis. However, its role in infection-induced exacerbation of lung fibrosis is poorly defined. We found significantly increased levels of PGE2 in lung tissue of patients with ILD. Increased levels of PGE2 were also found in lung tissue of mice with AdTGF-ß1-induced lung fibrosis and even more so in Streptococcus pneumoniae exacerbated lung fibrosis. Type II alveolar epithelial cells (AT II cells) and alveolar macrophages (AM) contributed to PGE2 release during exacerbating fibrosis. Application of parecoxib to inhibit PGE2 synthesis ameliorated lung fibrosis, whereas intratracheal application of PGE2 worsened lung fibrosis in mice. Both interventions had no effect on S. pneumoniae-exacerbated lung fibrosis. Together, we found that the COX2-PGE2 axis has dual roles in fibrosis that may offset each other: PGE2 helps resolve infection/attenuate inflammation in fibrosis exacerbation but accentuates TGF-ß/AT II cell-mediated fibrosis. These data support the efficacy of COX/PGE2 interventions in the setting of non-exacerbating lung fibrosis.


Subject(s)
Alveolar Epithelial Cells/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Pneumonia, Pneumococcal/metabolism , Pulmonary Fibrosis/metabolism , Signal Transduction , Streptococcus pneumoniae/metabolism , Alveolar Epithelial Cells/microbiology , Alveolar Epithelial Cells/pathology , Animals , Disease Models, Animal , Female , Isoxazoles/pharmacology , Mice , Pneumonia, Pneumococcal/pathology , Pulmonary Fibrosis/microbiology , Pulmonary Fibrosis/pathology , Transforming Growth Factor beta/metabolism
8.
BMC Infect Dis ; 21(1): 345, 2021 Apr 13.
Article in English | MEDLINE | ID: mdl-33849461

ABSTRACT

BACKGROUND: To guide decision-making on immunisation programmes for ageing adults in Europe, one of the aims of the Vaccines and InfecTious diseases in the Ageing popuLation (IMI2-VITAL) project is to assess the burden of disease (BoD) of (potentially) vaccine-preventable diseases ((P)VPD). We aimed to identify the available data sources to calculate the BoD of (P)VPD in participating VITAL countries and to pinpoint data gaps. Based on epidemiological criteria and vaccine availability, we prioritized (P) VPD caused by Extra-intestinal pathogenic Escherichia coli (ExPEC), norovirus, respiratory syncytial virus, Staphylococcus aureus, and pneumococcal pneumonia. METHODS: We conducted a survey on available data (e.g. incidence, mortality, disability-adjusted life years (DALY), quality-adjusted life years (QALY), sequelae, antimicrobial resistance (AMR), etc.) among national experts from European countries, and carried out five pathogen-specific literature reviews by searching MEDLINE for peer-reviewed publications published between 2009 and 2019. RESULTS: Morbidity and mortality data were generally available for all five diseases, while summary BoD estimates were mostly lacking. Available data were not always stratified by age and risk group, which is especially important when calculating BoD for ageing adults. AMR data were available in several countries for S. aureus and ExPEC. CONCLUSION: This study provides an exhaustive overview of the available data sources and data gaps for the estimation of BoD of five (P) VPD in ageing adults in the EU/EAA, which is useful to guide pathogen-specific BoD studies and contribute to calculation of (P)VPDs BoD.


Subject(s)
Cost of Illness , Vaccine-Preventable Diseases/economics , Aging , Caliciviridae Infections/economics , Caliciviridae Infections/epidemiology , Caliciviridae Infections/mortality , Caliciviridae Infections/pathology , Europe/epidemiology , Humans , Incidence , Pneumonia, Pneumococcal/economics , Pneumonia, Pneumococcal/epidemiology , Pneumonia, Pneumococcal/mortality , Pneumonia, Pneumococcal/pathology , Quality-Adjusted Life Years , Respiratory Syncytial Virus Infections/economics , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/mortality , Respiratory Syncytial Virus Infections/pathology , Surveys and Questionnaires , Vaccine-Preventable Diseases/epidemiology , Vaccine-Preventable Diseases/mortality , Vaccine-Preventable Diseases/pathology
9.
Int J Mol Sci ; 22(23)2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34884507

ABSTRACT

Streptococcus pneumoniae is an important causative organism of respiratory tract infections. Although periodontal bacteria have been shown to influence respiratory infections such as aspiration pneumonia, the synergistic effect of S. pneumoniae and Porphyromonas gingivalis, a periodontopathic bacterium, on pneumococcal infections is unclear. To investigate whether P. gingivalis accelerates pneumococcal infections, we tested the effects of inoculating P. gingivalis culture supernatant (PgSup) into S. pneumoniae-infected mice. Mice were intratracheally injected with S. pneumoniae and PgSup to induce pneumonia, and lung histopathological sections and the absolute number and frequency of neutrophils and macrophages in the lung were analyzed. Proinflammatory cytokine/chemokine expression was examined by qPCR and ELISA. Inflammatory cell infiltration was observed in S. pneumoniae-infected mice and S. pnemoniae and PgSup mixed-infected mice, and mixed-infected mice showed more pronounced inflammation in lung. The ratios of monocytes/macrophages and neutrophils were not significantly different between the lungs of S. pneumoniae-infected mice and those of mixed-infected mice. PgSup synergistically increased TNF-α expression/production and IL-17 production compared with S. pneumoniae infection alone. We demonstrated that PgSup enhanced inflammation in pneumonia caused by S. pneumoniae, suggesting that virulence factors produced by P. gingivalis are involved in the exacerbation of respiratory tract infections such as aspiration pneumonia.


Subject(s)
Bacteroidaceae Infections/complications , Inflammation/pathology , Lung/pathology , Neutrophil Infiltration/immunology , Pneumonia, Pneumococcal/pathology , Porphyromonas gingivalis/physiology , Streptococcus pneumoniae/physiology , Animals , Bacteroidaceae Infections/microbiology , Chemokines/metabolism , Cytokines/metabolism , Inflammation/etiology , Lung/immunology , Lung/metabolism , Lung/microbiology , Mice , Mice, Inbred C57BL , Pneumonia, Pneumococcal/epidemiology , Pneumonia, Pneumococcal/metabolism , Pneumonia, Pneumococcal/microbiology
10.
Infect Immun ; 89(1)2020 12 15.
Article in English | MEDLINE | ID: mdl-33020213

ABSTRACT

Streptococcus pneumoniae is a major causative bacterium of community-acquired pneumonia. Dendritic cell-associated C-type lectin-2 (dectin-2), one of the C-type lectin receptors (CLRs), was previously reported to play a pivotal role in host defense against pneumococcal infection through regulating phagocytosis by neutrophils while not being involved in neutrophil accumulation. In the present study, to elucidate the possible contribution of other CLRs to neutrophil accumulation, we examined the role of caspase recruitment domain-containing protein 9 (CARD9), a common adaptor molecule for signal transduction triggered by CLRs, in neutrophilic inflammatory response against pneumococcal infection. Wild-type (WT), CARD9 knockout (KO), and dectin-2 KO mice were infected intratracheally with pneumococcus, and the infected lungs were histopathologically analyzed to assess neutrophil accumulation at 24 h postinfection. Bronchoalveolar lavage fluids (BALFs) were collected at the same time point to count the neutrophils and assess the production of inflammatory cytokines and chemokines. Neutrophil accumulation was significantly decreased in CARD9 KO mice, but not in dectin-2 KO mice. Tumor necrosis factor alpha (TNF-α), keratinocyte-derived chemokine (KC), and macrophage inflammatory protein-2 (MIP-2) production in BALFs were also attenuated in CARD9 KO mice, but not in dectin-2 KO mice. Production of TNF-α and KC by alveolar macrophages stimulated with pneumococcal culture supernatants was significantly attenuated in CARD9 KO mice, but not in dectin-2 KO mice, compared to that in each group's respective control mice. In addition, pneumococcus-infected CARD9 KO mice showed larger bacterial burdens in the lungs than did WT mice. These data indicate that CARD9 is required for neutrophil migration after pneumococcal infection, as well as inflammatory cytokine and chemokine production by alveolar macrophages, and suggest that a CLR distinct from dectin-2 may be involved in this response.


Subject(s)
Candidiasis, Chronic Mucocutaneous/complications , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Neutrophils/immunology , Pneumonia, Pneumococcal/etiology , Streptococcus pneumoniae , Animals , Biopsy , Chemokines/metabolism , Cytokines/metabolism , Disease Susceptibility , Immunoglobulin G/immunology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice , Neutrophils/metabolism , Pneumonia, Pneumococcal/metabolism , Pneumonia, Pneumococcal/pathology
11.
Microb Pathog ; 144: 104126, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32173494

ABSTRACT

Streptococcus pneumoniae (pneumococcus) is touted to be the generally found pathogen in patients with respiratory issues and there is an epidemiologic linkage present between Respiratory syncytial virus (RSV). This study aim at investigating the interaction between RSV and two serotypes of S. pneumoniae using a distinct animal model and a well-established colonizing pneumococcal strain. Phase variants phenotype of each strain was determined under oblique light. Co infection model was developed using BALB/c mice housed in a BSL-2 facility. Coinfection experiments were performed and number of bacterial colonies was quantified and phase determination was evaluated. RSV was detected in sample through real-time quantitative PCR. Adherence assays were performed to determine adherence of Spn strains and its knock out ΔNanA to nasopharyngeal carcinoma (NPC) epithelial CNE3 cell line. The biofilm viability was determined and phase composition was counted using plate count. Neuraminidase activity was measured in fluorometircassessed using 2'-(4-methylumbelliferyl)-α-D-N-acetylneuraminic acid (MUAN) as substrate as described in earlier literature. The GraphPad Software version 5.01 i.e., GraphPad Prism was used to conduct the statistical analysis. The extent of bacterial colonization was increased significantly (p < 0.05), when the mice were co infected. Nasal epithelium remained intact in mock sample with features of a thick mucociliary border. A small percentage of pneumococci exhibit phase variation between opaque phase and transparent phase. The percentage adherent of both phase were not found to be varying significantly within serotype but it was seen that nonpathogenic type 27 was more adherent. Biofilm formation was selectively more for transparent phase from a mixed-phase inoculum. Adherence of both phase variant of S. pneumoniae to nasopharyngeal epithelial cells 2 h post infection expressed as the percentage of adherent bacteria relative to the inoculum. In absence of viral infection, the nasal colonization of the opaque and the transparent variant was increased many folds, which was a significant differences. The extent of nasal colonization by the ΔNanA mutant strain were significantly reduced post-bacterial infection for both type of wild-type (P < 0.05). The findings explore insights into the interactions occurring between S. pneumoniae and RSV during respiratory infections and pneumococcal acquisition, indicate that pneumococcal serotypes have different ability to cause infection as well as co infections and potentially follow an unappreciated mechanism. Much more research work is needed to further understand the minutiae of this interaction within co-infection process.


Subject(s)
Bacterial Adhesion/physiology , Bacterial Adhesion/radiation effects , Microbial Interactions/physiology , Pneumonia, Pneumococcal/pathology , Respiratory Syncytial Virus Infections/pathology , Animals , Biofilms/growth & development , Cell Line, Tumor , Coinfection/microbiology , Epithelial Cells/microbiology , Humans , Mice , Mice, Inbred BALB C , Nasal Mucosa/microbiology , Respiratory Syncytial Viruses/physiology , Streptococcus pneumoniae/physiology
12.
Microb Pathog ; 141: 103979, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31954822

ABSTRACT

PURPOSE: The aim of this study was to assess if long-term supplementation of omega-3 fatty acids or a diet rich in omega-6 fatty acids ameliorates disease severity in a murine model of pneumococcal pneumonia. We hypothesize that long-term dietary supplementation of omega-3 fatty acids will reduce inflammation, disease severity and improve survival compared to omega-6 fatty acids. METHODS: Mice receiving diets supplemented with Omega-3 or Omega-6 for two months were intranasally infected with Streptococcus pneumoniae. We analyzed survival, bacterial burden, histopathology and inflammatory biomarkers. RESULTS: Our results showed that Omega-3 supplementation had increased survival (p = 0.005), less bacteremia (p = 0.0001) and lower bacterial burden in the lungs (p = 0.0002) when compared to the Omega-6 supplementation. Overall, Omega-3 reduced lung pathology, in particular peribronchial inflammation and cell death. Analyses of lung homogenates showed the Omega-3 cohort had decreased levels of the inflammatory cytokine interleukin-6 and an increase in anti-inflammatory cytokine interleukin-10. CONCLUSIONS: Supplementation of mouse diets with Omega-3 fatty acids improved survival, bacterial invasion in the blood and lungs as well as decreased overall lung tissue inflammation and cell death when compared to the Omega-6 supplemented diets. Translation of these findings in humans may improve outcomes of patients at risk for pneumonia.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-6/administration & dosage , Pneumonia, Pneumococcal/microbiology , Streptococcus pneumoniae/drug effects , Animal Feed , Animals , Bacterial Load , Dietary Supplements , Disease Models, Animal , Disease Susceptibility , Lung/drug effects , Lung/microbiology , Lung/pathology , Mice , Mortality , Pneumonia, Pneumococcal/drug therapy , Pneumonia, Pneumococcal/mortality , Pneumonia, Pneumococcal/pathology , Treatment Outcome
13.
J Infect Dis ; 220(9): 1498-1502, 2019 09 26.
Article in English | MEDLINE | ID: mdl-31287867

ABSTRACT

Therapy to enhance host immune defenses may improve outcomes in serious infections, especially for antibiotic-resistant pathogens. Recombinant human plasma gelsolin (rhu-pGSN), a normally circulating protein, has beneficial effects in diverse preclinical models of inflammation and injury. We evaluated delayed therapy (24-48 hours after challenge) with rhu-pGSN in a mouse model of pneumococcal pneumonia. rhu-pGSN without antibiotics increased survival and reduced morbidity and weight loss after infection with either penicillin-susceptible or penicillin-resistant pneumococci (serotypes 3 and 14, respectively). rhu-pGSN improves outcomes in a highly lethal pneumococcal pneumonia model when given after a clinically relevant delay, even in the setting of antimicrobial resistance.


Subject(s)
Gelsolin/administration & dosage , Immunologic Factors/administration & dosage , Pneumonia, Pneumococcal/drug therapy , Animals , Disease Models, Animal , Male , Mice , Pneumonia, Pneumococcal/pathology , Recombinant Proteins/administration & dosage , Survival Analysis , Treatment Outcome , Weight Loss
14.
Am J Hum Genet ; 98(6): 1092-1100, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27236921

ABSTRACT

Bacteremia (bacterial bloodstream infection) is a major cause of illness and death in sub-Saharan Africa but little is known about the role of human genetics in susceptibility. We conducted a genome-wide association study of bacteremia susceptibility in more than 5,000 Kenyan children as part of the Wellcome Trust Case Control Consortium 2 (WTCCC2). Both the blood-culture-proven bacteremia case subjects and healthy infants as controls were recruited from Kilifi, on the east coast of Kenya. Streptococcus pneumoniae is the most common cause of bacteremia in Kilifi and was thus the focus of this study. We identified an association between polymorphisms in a long intergenic non-coding RNA (lincRNA) gene (AC011288.2) and pneumococcal bacteremia and replicated the results in the same population (p combined = 1.69 × 10(-9); OR = 2.47, 95% CI = 1.84-3.31). The susceptibility allele is African specific, derived rather than ancestral, and occurs at low frequency (2.7% in control subjects and 6.4% in case subjects). Our further studies showed AC011288.2 expression only in neutrophils, a cell type that is known to play a major role in pneumococcal clearance. Identification of this novel association will further focus research on the role of lincRNAs in human infectious disease.


Subject(s)
Bacteremia/genetics , Pneumonia, Pneumococcal/genetics , Polymorphism, Genetic/genetics , RNA, Long Noncoding/genetics , Streptococcus pneumoniae/genetics , Adolescent , Bacteremia/microbiology , Bacteremia/pathology , Case-Control Studies , Child , Child, Preschool , Genome-Wide Association Study , Humans , Infant , Infant, Newborn , Kenya/epidemiology , Pneumonia, Pneumococcal/microbiology , Pneumonia, Pneumococcal/pathology , Risk Factors
15.
Eur J Immunol ; 48(1): 66-79, 2018 01.
Article in English | MEDLINE | ID: mdl-28971472

ABSTRACT

Streptococcus (S.) pneumoniae is the most common cause of community-acquired pneumonia. The Nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, consisting of NLRP3, ASC (the adaptor apoptosis-associated speck-like protein containing a CARD) and caspase-1, has been implicated in protective immunity during pneumonia induced by high doses of S. pneumoniae serotype 2. Here we investigated the role of the NLRP3 inflammasome in the host response during lethal airway infection with a low dose of serotype 3 S. pneumoniae. Mice were euthanized at predefined endpoints for analysis or observed in survival studies. In additional studies, Tlr2-/- /Tlr4-/- mice and Myd88-/- mice incapable of Toll-like receptor signaling were studied. In stark contrast with existing literature, both Nlrp3-/- and Asc-/- mice showed a strongly improved host defense, as reflected by a markedly reduced mortality rate accompanied by diminished bacterial growth and dissemination. Host defense was unaltered in Tlr2-/- /Tlr4-/- mice and Myd88-/- mice. These results show that the NLRP3 inflammasome impairs host defense during lethal pneumonia caused by serotype 3 S. pneumoniae. Our findings challenge the current paradigm that proximal innate detection systems are indispensable for an adequate host immune response against bacteria.


Subject(s)
CARD Signaling Adaptor Proteins/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Pneumonia, Pneumococcal/immunology , Streptococcus pneumoniae/immunology , Animals , CARD Signaling Adaptor Proteins/genetics , Caspase 1/immunology , Community-Acquired Infections/immunology , Community-Acquired Infections/microbiology , Immunity, Innate/genetics , Immunity, Innate/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Pneumonia, Pneumococcal/pathology , Signal Transduction/immunology , Streptococcus pneumoniae/classification , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics
16.
J Antimicrob Chemother ; 74(8): 2385-2393, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31106377

ABSTRACT

BACKGROUND: The inflammatory response in pneumococcal infection is primarily driven by immunoreactive bacterial cell wall components [lipoteichoic acid (LTA)]. An acute release of these components occurs when pneumococcal infection is treated with ß-lactam antibiotics. OBJECTIVES: We hypothesized that non-lytic rifampicin compared with lytic ß-lactam antibiotic treatment would attenuate the inflammatory response in patients with pneumococcal pneumonia. METHODS: In the PRISTINE (Pneumonia treated with RIfampicin aTtenuates INflammation) trial, a randomized, therapeutic controlled, exploratory study in patients with community-acquired pneumococcal pneumonia, we looked at LTA release and inflammatory and clinical response during treatment with both rifampicin and ß-lactam compared with treatment with ß-lactam antibiotics only. The trial is registered in the Dutch trial registry, number NTR3751 (European Clinical Trials Database number 2012-003067-22). RESULTS: Forty-one patients with community-acquired pneumonia were included; 17 of them had pneumococcal pneumonia. LTA release, LTA-mediated inflammatory responses, clinical outcomes, inflammatory biomarkers and transcription profiles were not different between treatment groups. CONCLUSIONS: The PRISTINE study demonstrated the feasibility of adding rifampicin to ß-lactam antibiotics in the treatment of community-acquired pneumococcal pneumonia, but, despite solid in vitro and experimental animal research evidence, failed to demonstrate a difference in plasma LTA concentrations and subsequent inflammatory and clinical responses. Most likely, an inhibitory effect of human plasma contributes to the low immune response in these patients. In addition, LTA plasma concentration could be too low to mount a response via Toll-like receptor 2 in vitro, but may nonetheless have an effect in vivo.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Community-Acquired Infections/drug therapy , Inflammation/pathology , Pneumonia, Pneumococcal/drug therapy , Rifampin/therapeutic use , beta-Lactams/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Community-Acquired Infections/pathology , Female , Humans , Lipopolysaccharides/blood , Male , Middle Aged , Netherlands , Plasma/chemistry , Pneumonia, Pneumococcal/pathology , Teichoic Acids/blood , Treatment Outcome , Young Adult
17.
J Antimicrob Chemother ; 74(7): 1962-1970, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31049578

ABSTRACT

BACKGROUND: RBx 14255 is a fluoroketolide in pre-clinical evaluation with potent activity against MDR Gram-positive pathogens. OBJECTIVES: To investigate the efficacy of RBx 14255 against bacterial meningitis caused by Streptococcus pneumoniae, Neisseria meningitidis or Haemophilus influenzae in an experimental murine meningitis model. METHODS: In vitro activity of RBx 14255 was evaluated against clinical isolates of S. pneumoniae, N. meningitidis and H. influenzae. The in vivo efficacy of RBx 14255 was evaluated against bacterial meningitis, induced with S. pneumoniae 3579 erm(B), S. pneumoniae MA 80 erm(B), N. meningitidis 1852 and H. influenzae B1414 in a murine meningitis model. RESULTS: RBx 14255 showed strong in vitro bactericidal potential against S. pneumoniae, N. meningitidis and H. influenzae with MIC ranges of 0.004-0.1, 0.03-0.5 and 1-4 mg/L, respectively. In a murine meningitis model, a 50 mg/kg dose of RBx 14255, q12h, resulted in significant reduction of bacterial counts in the brain compared with the pretreatment control. The concentration of RBx 14255 in brain tissue correlated well with the efficacy in this mouse model. CONCLUSIONS: RBx 14255 showed superior bactericidal activity in time-kill assays in vitro and in vivo in an experimental murine meningitis model. RBx 14255 could be a promising candidate for future drug development against bacterial meningitis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Haemophilus influenzae/drug effects , Ketolides/pharmacology , Neisseria meningitidis/drug effects , Streptococcus pneumoniae/drug effects , Animals , Anti-Bacterial Agents/chemistry , Disease Models, Animal , Haemophilus Infections/drug therapy , Haemophilus Infections/microbiology , Ketolides/chemistry , Meningitis, Meningococcal/drug therapy , Meningitis, Meningococcal/microbiology , Meningitis, Meningococcal/pathology , Mice , Microbial Sensitivity Tests , Pneumonia, Pneumococcal/drug therapy , Pneumonia, Pneumococcal/microbiology , Pneumonia, Pneumococcal/pathology
18.
J Infect Dis ; 217(9): 1462-1471, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29415278

ABSTRACT

Streptococcus pneumoniae is the most common causative pathogen in community-acquired pneumonia. Protease-activated receptor 2 (PAR2) is expressed by different cell types in the lungs and can mediate inflammatory responses. We sought to determine the role of PAR2 during pneumococcal pneumonia. Pneumococcal pneumonia or sepsis was induced in wild-type and PAR2 knock-out (Par2-/-) mice by infection with viable S. pneumoniae. Par2-/- mice demonstrated improved host defense, a largely preserved lung barrier integrity, and reduced mortality during pneumococcal pneumonia. PAR2 deficiency did not influence bacterial growth after intravenous infection. Inhibition of the endogenous PAR2 activating proteases tissue factor/factor VIIa or tryptase did not impact on bacterial burdens during pneumonia. In a PAR2 reporter cell line it was demonstrated that S. pneumoniae-derived proteases are able to cleave PAR2. These results show that S. pneumoniae is able to cleave and exploit PAR2 to disseminate systemically from the airways.


Subject(s)
Pneumonia, Pneumococcal/microbiology , Receptor, PAR-2 , Streptococcus pneumoniae/physiology , Animals , Bacterial Load , Blood Coagulation , Gene Expression Regulation/drug effects , HEK293 Cells , Helminth Proteins/pharmacology , Humans , Inflammation , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia, Pneumococcal/pathology , Specific Pathogen-Free Organisms
19.
Am J Respir Cell Mol Biol ; 58(4): 440-448, 2018 04.
Article in English | MEDLINE | ID: mdl-29361238

ABSTRACT

Descriptive histopathology of mouse models of pneumonia is essential in assessing the outcome of infections, molecular manipulations, or therapies in the context of whole lungs. Quantitative comparisons between experimental groups, however, have been limited to laborious stereology or ill-defined scoring systems that depend on the subjectivity of a more or less experienced observer. Here, we introduce self-learning digital image analyses that allow us to transform optical information from whole mouse lung sections into statistically testable data. A pattern-recognition-based software and a nuclear count algorithm were adopted to quantify user-defined pathologies from whole slide scans of lungs infected with Streptococcus pneumoniae or influenza A virus compared with PBS-challenged lungs. The readout parameters "relative area affected" and "nuclear counts per area" are proposed as relevant criteria for the quantification of lesions from hematoxylin and eosin-stained sections, also allowing for the generation of a heat map of, for example, immune cell infiltrates with anatomical assignments across entire lung sections. Moreover, when combined with immunohistochemical labeling of marker proteins, both approaches are useful for the identification and counting of, for example, immune cell populations, as validated here by direct comparisons with flow cytometry data. The solutions can easily and flexibly be adjusted to specificities of different models or pathogens. Automated digital analyses of whole mouse lung sections may set a new standard for the user-defined, high-throughput comparative quantification of histological and immunohistochemical images. Still, our algorithms established here are only a start, and need to be tested in additional studies and other applications in the future.


Subject(s)
Algorithms , Cytological Techniques , Image Interpretation, Computer-Assisted/methods , Lung/pathology , Orthomyxoviridae Infections/pathology , Pneumonia, Pneumococcal/pathology , Pneumonia, Viral/pathology , Acute Disease , Animals , Automation, Laboratory , Disease Models, Animal , Influenza A virus/pathogenicity , Lung/microbiology , Lung/virology , Mice , Orthomyxoviridae Infections/virology , Pattern Recognition, Automated , Pneumonia, Pneumococcal/microbiology , Pneumonia, Viral/virology , Predictive Value of Tests , Software , Streptococcus pneumoniae/pathogenicity
20.
Am J Physiol Lung Cell Mol Physiol ; 315(1): L78-L86, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29565180

ABSTRACT

Leptin is a pleiotropic hormone produced by white adipose tissue that regulates appetite and many physiological functions, including the immune response to infection. Genetic leptin deficiency in humans and mice impairs host defenses against respiratory tract infections. Since leptin deficiency is associated with obesity and other metabolic abnormalities, we generated mice that lack the leptin receptor (LepRb) in cells of the myeloid linage (LysM-LepRb-KO) to evaluate its impact in lean metabolically normal mice in a murine model of pneumococcal pneumonia. We observed higher lung and spleen bacterial burdens in LysM-LepRb-KO mice following an intratracheal challenge with Streptococcus pneumoniae. Although numbers of leukocytes recovered from bronchoalveolar lavage fluid did not differ between groups, we did observe higher levels of pulmonary IL-13 and TNFα in LysM-LepRb-KO mice 48 h post infection. Phagocytosis and killing of ingested S. pneumoniae were also impaired in alveolar macrophages (AMs) from LysM-LepRb-KO mice in vitro and were associated with reduced LTB4 and enhanced PGE2 synthesis in vitro. Pretreatment of AMs with LTB4 and the cyclooxygenase inhibitor, indomethacin, restored phagocytosis but not bacterial killing in vitro. These results confirm our previous observations in leptin-deficient ( ob/ob) and fasted mice and demonstrate that decreased leptin action, as opposed to metabolic irregularities associated with obesity or starvation, is responsible for the defective host defense against pneumococcal pneumonia. They also provide novel targets for therapeutic intervention in humans with bacterial pneumonia.


Subject(s)
Lung/immunology , Macrophages/immunology , Phagocytosis , Pneumonia, Pneumococcal/immunology , Receptors, Leptin/immunology , Streptococcus pneumoniae/immunology , Animals , Interleukin-13/genetics , Interleukin-13/immunology , Lung/microbiology , Lung/pathology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Knockout , Pneumonia, Pneumococcal/genetics , Pneumonia, Pneumococcal/pathology , Receptors, Leptin/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL