Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 192
Filter
1.
Microbiol Spectr ; 12(5): e0390523, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38501823

ABSTRACT

Pseudomonas aeruginosa (P. aeruginosa) is a Gram-negative facultative anaerobe that has become an important cause of severe infections in humans, particularly in patients with cystic fibrosis. The development of efficacious methods or mendicants against P. aeruginosa is still needed. We previously reported that regenerating islet-derived family member 4 (Reg4) has bactericidal activity against Salmonella Typhimurium, a Gram-negative flagellated bacterium. We herein explore whether Reg4 has bactericidal activity against P. aeruginosa. In the P. aeruginosa PAO1-chronic infection model, Reg4 significantly inhibits the colonization of PAO1 in the lung and subsequently ameliorates pulmonary inflammation and fibrosis. Reg4 recombinant protein suppresses the growth motility and biofilm formation capability of PAO1 in vitro. Mechanistically, Reg4 not only exerts bactericidal action via direct binding to the P. aeruginosa cell wall but also enhances the phagocytosis of alveolar macrophages in the host. Taken together, our study demonstrates that Reg4 may provide protection against P. aeruginosa-induced pulmonary inflammation and fibrosis via its antibacterial activity.IMPORTANCEChronic lung infection with Pseudomonas aeruginosa is a leading cause of morbidity and mortality in patients with cystic fibrosis. Due to the antibiotic resistance of Pseudomonas aeruginosa, antimicrobial peptides appear to be a potential alternative to combat its infection. In this study, we report an antimicrobial peptide, regenerating islet-derived 4 (Reg4), that showed killing activity against clinical strains of Pseudomonas aeruginosa PAO1 and ameliorated PAO1-induced pulmonary inflammation and fibrosis. Experimental data also showed Reg4 directly bound to the bacterial cell membrane and enhanced the phagocytosis of host alveolar macrophages. Our presented study will be a helpful resource in searching for novel antimicrobial peptides that could have the potential to replace conventional antibiotics.


Subject(s)
Anti-Bacterial Agents , Pancreatitis-Associated Proteins , Pseudomonas Infections , Pseudomonas aeruginosa , Pseudomonas aeruginosa/drug effects , Animals , Pseudomonas Infections/microbiology , Pseudomonas Infections/drug therapy , Mice , Pancreatitis-Associated Proteins/metabolism , Pancreatitis-Associated Proteins/genetics , Anti-Bacterial Agents/pharmacology , Humans , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/immunology , Biofilms/drug effects , Biofilms/growth & development , Mice, Inbred C57BL , Pneumonia/microbiology , Antimicrobial Peptides/pharmacology , Phagocytosis/drug effects , Lung/microbiology , Lung/pathology , Cystic Fibrosis/microbiology , Cystic Fibrosis/complications , Pulmonary Fibrosis/microbiology , Disease Models, Animal
3.
Biomed Res Int ; 2021: 9963700, 2021.
Article in English | MEDLINE | ID: mdl-34604389

ABSTRACT

The study is aimed at observing the influence of microribonucleic acid- (miRNA-) 30a-50p on the pulmonary fibrosis in mice with Streptococcus pneumoniae infection through the regulation of autophagy by Beclin-1. Specific pathogen-free mice were instilled with Streptococcus pneumoniae through the trachea to establish the pulmonary fibrosis model. Then, they were divided into the miRNA-30a-50p mimics group (mimics group, n = 10) and miRNA-30a-5p inhibitors group (inhibitors group, n = 10), with the control group (n = 10) also set. Pulmonary tissue wet weight/dry weight (W/D) was detected. The content of tumor necrosis factor-α (TNF-α), interleukin- (IL-) 6, and myeloperoxidase (MPO) was determined using enzyme-linked immunosorbent assay (ELISA). Besides, the changes in the pulmonary function index dynamic lung compliance (Cdyn), plateau pressure (Pplat), and peak airway pressure (Ppeak) were monitored, and the gene and protein expression levels were measured via quantitative PCR (qPCR) and Western blotting. The expression level of miRNA-30a-5p was substantially raised in the mimics group (p < 0.05), but extremely low in the inhibitors group (p < 0.05). The mimics group had obviously raised levels of serum aminotransferase (AST), glutamic-pyruvic transaminase (GPT), alkaline phosphatase (ALP), and pulmonary tissue W/D (p < 0.05). Additionally, the expression levels of TNF-α, IL-6, and MPO were notably elevated in the mimics group, while their expression levels showed the opposite conditions in the inhibitors group (p < 0.05). According to the HE staining results, the inhibitors group had arranged orderly cells, while the mimics group exhibited lung injury, pulmonary edema, severe inflammatory response, and alveolar congestion. In the inhibitors group, Cdyn was remarkably elevated, but Pplat and Ppeak declined considerably (p < 0.05). Besides, the inhibitors group exhibited elevated messenger RNA (mRNA) levels of Beclin-1 and LC3, lowered mRNA levels of α-SMA and p62, a raised protein level of Beclin-1, and a markedly decreased protein level of p62 (p < 0.05). Silencing miRNA-30a-5p expression can promote the expression of Beclin-1 to accelerate the occurrence of autophagy, thereby treating pulmonary fibrosis in mice with Streptococcus pneumoniae infection.


Subject(s)
Autophagy/genetics , Beclin-1/metabolism , MicroRNAs/metabolism , Pneumococcal Infections/genetics , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/microbiology , Actins/metabolism , Alkaline Phosphatase/blood , Animals , Aspartate Aminotransferases/blood , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Gene Expression Regulation , Inflammation Mediators/metabolism , Lung/pathology , Lung/physiopathology , Mice , MicroRNAs/genetics , Microtubule-Associated Proteins/metabolism , Organ Size , Respiratory Function Tests , Sequestosome-1 Protein/metabolism , Transaminases/blood
4.
Am J Pathol ; 191(5): 838-856, 2021 05.
Article in English | MEDLINE | ID: mdl-33705752

ABSTRACT

Growing evidence shows that the lungs are an unavoidable target organ of diabetic complications. However, the pathologic mechanisms of diabetic lung injury are still controversial. This study demonstrated the dysbiosis of the gut and lung microbiome, pulmonary alveolar wall thickening, and fibrotic change in streptozotocin-induced diabetic mice and antibiotic-induced gut dysbiosis mice compared with controls. In both animal models, the NF-κB signaling pathway was activated in the lungs. Enhanced pulmonary alveolar well thickening and fibrotic change appeared in the lungs of transgenic mice expressing a constitutively active NF-κB mutant compared with wild type. When lincomycin hydrochloride-induced gut dysbiosis was ameliorated by fecal microbiota transplant, enhanced inflammatory response in the intestine and pulmonary fibrotic change in the lungs were significantly decreased compared with lincomycin hydrochloride-treated mice. Furthermore, the application of fecal microbiota transplant and baicalin could also redress the microbial dysbiosis of the gut and lungs in streptozotocin-induced diabetic mice. Taken together, these data suggest that multiple as yet undefined factors related to microbial dysbiosis of gut and lungs cause pulmonary fibrogenesis associated with diabetes mellitus through an NF-κB signaling pathway.


Subject(s)
Diabetes Mellitus, Experimental/complications , Dysbiosis/complications , Microbiota , NF-kappa B/metabolism , Pulmonary Fibrosis/microbiology , Signal Transduction , Animals , Anti-Infective Agents/administration & dosage , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/pathology , Disease Models, Animal , Dysbiosis/chemically induced , Dysbiosis/pathology , Dysbiosis/therapy , Fecal Microbiota Transplantation , Flavonoids/administration & dosage , Gastrointestinal Microbiome , Intestines/microbiology , Intestines/pathology , Lincomycin/adverse effects , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/therapy , Streptozocin/adverse effects
5.
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
6.
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
7.
Biomed Pharmacother ; 132: 110826, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33068929

ABSTRACT

OBJECTIVE: Radiation pneumonia and fibrosis are major clinical complications of radiotherapy for thoracic tumor patients, and may significantly reduce survival and quality of life. At present, no safe and effective radiation protection measures have been approved for clinical use. Phycocyanin, a protein responsible for photosynthesis from Spirulina, has been shown to have a variety of biological activities and to be beneficial for a variety of diseases, including pulmonary fibrosis. However, the preventive and protective effects of phycocyanin on radiation-induced pulmonary fibrosis have not been studied. DESIGN: X-ray single dose irradiation was used on the chest of mice to prepare a mouse model of pulmonary fibrosis, from which the effect of phycocyanin on pulmonary histopathologic change, pulmonary fibrosis, the microbiota in lung and gut, LPS, TNF-α, and IL-6 at different time after irradiation were evaluated. RESULTS: Phycocyanin alleviated the radiation-induced lung injury and reduced the level of inflammatory factors. Thorax irradiation led to the disorder in microbiota of the lung and gut. The variation trend of the diversity of the two tissues was opposite, but that of the microbiota composition was similar. The phycocyanin intervention regulated the composition of the lung and gut microbiota, transformed them into normal state, and reduced the level of LPS, which significantly reduced the abundance of inflammation-related bacteria, and increased the abundance of probiotics that produce short-chain fatty acids. CONCLUSION: Phycocyanin could regulate the radiation-induced disorder in lung and gut microbiota of mice, and reduce the radiation-induced lung inflammation and fibrosis.


Subject(s)
Gastrointestinal Microbiome/drug effects , Phycocyanin/pharmacology , Pulmonary Fibrosis/prevention & control , Radiation Pneumonitis/prevention & control , Animals , Gastrointestinal Microbiome/radiation effects , Inflammation/drug therapy , Inflammation/etiology , Inflammation/microbiology , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Phycocyanin/isolation & purification , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/microbiology , Radiation Injuries, Experimental/microbiology , Radiation Injuries, Experimental/prevention & control , Radiation Pneumonitis/microbiology , Spirulina/chemistry
8.
Rev Mal Respir ; 37(7): 561-571, 2020 Sep.
Article in French | MEDLINE | ID: mdl-32684338

ABSTRACT

INTRODUCTION: Common major pathogens like Pseudomonas aeruginosa are identified in the airways of patients with cystic fibrosis (CF) and non-CF bronchiectasis. However, other opportunistic bacterial pathogens like Achromobacter xylosoxidans complex, Stenotrophomonas maltophilia and non-tuberculous mycobacteria are currently emerging in CF and are also reported in non-CF bronchiectasis. BACKGROUND: The emergence of opportunistic bacterial pathogens has been recognized in CF through annual national reports of sputum microbiology data. Despite common factors driving the emergence of bacteria identified in CF and non-CF bronchiectasis patients, bronchiectasis registries have been created more recently and no longitudinal analysis of recorded microbiological data is currently available in the literature, thereby preventing the recognition of emerging bacteria in patients with non-CF bronchiectasis. OUTLOOK: A longitudinal follow-up of microbiological data is still needed in non-CF bronchiectasis to identify emerging opportunistic bacterial pathogens. Homogeneity in practice of sputum microbiological examination is also required to allow comparative analysis of data in CF and non-CF bronchiectasis. CONCLUSION: Bacterial pathogens recognized as emerging in CF have to be more carefully monitored in non-CF bronchiectasis in view of their association with deterioration of the lung disease.


Subject(s)
Bronchiectasis/microbiology , Cystic Fibrosis/microbiology , Microbiology/trends , Pulmonary Fibrosis/microbiology , Respiratory Tract Infections/microbiology , Bronchiectasis/complications , Bronchiectasis/epidemiology , Bronchiectasis/therapy , Communicable Diseases, Emerging/epidemiology , Communicable Diseases, Emerging/microbiology , Communicable Diseases, Emerging/therapy , Cystic Fibrosis/complications , Cystic Fibrosis/epidemiology , Cystic Fibrosis/therapy , Humans , Microbiological Techniques/statistics & numerical data , Microbiological Techniques/trends , Monitoring, Physiologic/methods , Monitoring, Physiologic/standards , Monitoring, Physiologic/trends , Opportunistic Infections/epidemiology , Opportunistic Infections/microbiology , Opportunistic Infections/therapy , Pulmonary Fibrosis/complications , Pulmonary Fibrosis/epidemiology , Pulmonary Fibrosis/therapy , Respiratory Tract Infections/complications , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/therapy , Sputum/microbiology
9.
Microbes Infect ; 22(3): 137-143, 2020 04.
Article in English | MEDLINE | ID: mdl-31770592

ABSTRACT

Paracoccidioidomycosis (PCM) is a systemic mycosis caused by thermally dimorphic fungi of the genus Paracoccidioides that affects predominantly 30-60-year-old male rural workers. The main clinical forms of the disease are acute/subacute, chronic (CF); almost all CF patients develop pulmonary fibrosis, and they also exhibit emphysema due to smoke. An important cytokine in this context, IL-1ß, different from the others, is produced by an intracellular multimolecular complex called inflammasome that is activated by pathogens and/or host signs of damage. Inflammasome has been recognized for its contribution to chronic inflammatory diseases, from that, we hypothesized that this activation could be involved in paracoccidioidomycosis, contributing to chronic inflammation. While inflammasome activation has been demonstrated in experimental models of Paracoccidioides brasiliensis infection, no information is available in patients, leading us to investigate the participation of NLRP3-inflammasome machinery in CF/PCM patients from a Brazilian endemic area. Our findings showed increased priming in mRNA levels of NLRP3 inflammasome genes by monocytes of PCM patients in vitro than healthy controls. Similar intracellular protein expression of NLRP3, CASP-1, ASC, and IL-1ß were also observed in freshly isolated monocytes of PCM patients and smoker controls. Increased expression of NLRP3 and ASC was observed in monocytes from PCM patients under hypoxia in comparison with smoker controls. For the first time, we showed that primed monocytes of CF-PCM patients were associated with enhanced expression of components of NLRP3-inflammasome due to smoke. Also, hypoxemia boosted this machinery. These findings reinforce the systemic low-grade inflammation activation observed in PCM during and after treatment.


Subject(s)
Monocytes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Paracoccidioidomycosis/immunology , Smoking , Cell Hypoxia , Humans , Invasive Fungal Infections/immunology , Invasive Fungal Infections/microbiology , Lung Diseases, Fungal/microbiology , Monocytes/microbiology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Paracoccidioides , Paracoccidioidomycosis/microbiology , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/microbiology
10.
Am J Physiol Lung Cell Mol Physiol ; 318(2): L407-L418, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31644311

ABSTRACT

During the newborn period, intestinal commensal bacteria influence pulmonary mucosal immunology via the gut-lung axis. Epidemiological studies have linked perinatal antibiotic exposure in human newborns to an increased risk for bronchopulmonary dysplasia, but whether this effect is mediated by the gut-lung axis is unknown. To explore antibiotic disruption of the newborn gut-lung axis, we studied how perinatal maternal antibiotic exposure influenced lung injury in a hyperoxia-based mouse model of bronchopulmonary dysplasia. We report that disruption of intestinal commensal colonization during the perinatal period promotes a more severe bronchopulmonary dysplasia phenotype characterized by increased mortality and pulmonary fibrosis. Mechanistically, metagenomic shifts were associated with decreased IL-22 expression in bronchoalveolar lavage and were independent of hyperoxia-induced inflammasome activation. Collectively, these results demonstrate a previously unrecognized influence of the gut-lung axis during the development of neonatal lung injury, which could be leveraged to ameliorate the most severe and persistent pulmonary complication of preterm birth.


Subject(s)
Anti-Bacterial Agents/adverse effects , Bronchopulmonary Dysplasia/complications , Lung Injury/chemically induced , Maternal Exposure , Prenatal Exposure Delayed Effects/pathology , Airway Resistance/drug effects , Animals , Animals, Newborn , Bronchoalveolar Lavage Fluid , Bronchopulmonary Dysplasia/physiopathology , Cytokines/metabolism , Female , Granulocytes/metabolism , Hyperoxia/complications , Hyperoxia/physiopathology , Inflammasomes/metabolism , Leukocyte Common Antigens/metabolism , Lung/pathology , Lung Injury/microbiology , Lung Injury/physiopathology , Mice, Inbred C57BL , Oxygen/metabolism , Phenotype , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Pulmonary Fibrosis/complications , Pulmonary Fibrosis/microbiology , Survival Analysis , Vascular Remodeling/drug effects
11.
Am J Respir Cell Mol Biol ; 61(2): 198-208, 2019 08.
Article in English | MEDLINE | ID: mdl-30741559

ABSTRACT

We established a murine model of multiwall carbon nanotube (MWCNT)-elicited chronic granulomatous disease that bears similarities to human sarcoidosis pathology, including alveolar macrophage deficiency of peroxisome proliferator-activated receptor γ (PPARγ). Because lymphocyte reactivity to mycobacterial antigens has been reported in sarcoidosis, we hypothesized that addition of mycobacterial ESAT-6 (early secreted antigenic target protein 6) to MWCNT might exacerbate pulmonary granulomatous pathology. MWCNTs with or without ESAT-6 peptide 14 were instilled by the oropharyngeal route into macrophage-specific PPARγ-knockout (KO) or wild-type mice. Control animals received PBS or ESAT-6. Lung tissues, BAL cells, and BAL fluid were evaluated 60 days after instillation. PPARγ-KO mice receiving MWCNT + ESAT-6 had increased granulomas and significantly elevated fibrosis (trichrome staining) compared with wild-type mice or PPARγ-KO mice that received only MWCNT. Immunostaining of lung tissues revealed elevated fibronectin and Siglec F expression on CD11c+ infiltrating alveolar macrophages in the presence of MWCNT + ESAT-6 compared with MWCNT alone. Analyses of BAL fluid proteins indicated increased levels of transforming growth factor (TGF)-ß and the TGF-ß pathway mediator IL-13 in PPARγ-KO mice that received MWCNT + ESAT-6 compared with wild-type or PPARγ-KO mice that received MWCNT. Similarly, mRNA levels of matrix metalloproteinase 9, another requisite factor for TGF-ß production, was elevated in PPARγ-KO mice by MWCNT + ESAT-6. Analysis of ESAT-6 in lung tissues by mass spectrometry revealed ESAT-6 retention in lung tissues of PPARγ-KO but not wild-type mice. These data indicate that PPARγ deficiency promotes pulmonary ESAT-6 retention, exacerbates macrophage responses to MWCNT + ESAT-6, and intensifies pulmonary fibrosis. The present findings suggest that the model may facilitate understanding of the effects of environmental factors on sarcoidosis-associated pulmonary fibrosis.


Subject(s)
Antigens, Bacterial/pharmacology , Bacterial Proteins/pharmacology , Macrophages, Alveolar/metabolism , PPAR gamma/deficiency , Pulmonary Fibrosis/microbiology , Sarcoidosis, Pulmonary/microbiology , Animals , Bronchoalveolar Lavage , Bronchoalveolar Lavage Fluid , CD11 Antigens/metabolism , Disease Models, Animal , Fibronectins/metabolism , Fibrosis/metabolism , Inflammation , Lung/pathology , Macrophages/metabolism , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Nanotubes, Carbon/chemistry , PPAR gamma/genetics , Pulmonary Fibrosis/genetics , Sarcoidosis, Pulmonary/pathology
12.
Article in English | MEDLINE | ID: mdl-30348661

ABSTRACT

Paracoccidioidomycosis (PCM), caused by Paracoccidioides, is a systemic mycosis with granulomatous character and a restricted therapeutic arsenal. The aim of this work was to search for new alternatives to treat largely neglected tropical mycosis, such as PCM. In this context, the enzymes of the shikimate pathway constitute excellent drug targets for conferring selective toxicity because this pathway is absent in humans but essential for the fungus. In this work, we have used a homology model of the chorismate synthase (EC 4.2.3.5) from Paracoccidioides brasiliensis (PbCS) and performed a combination of virtual screening and molecular dynamics testing to identify new potential inhibitors. The best hit, CP1, successfully adhered to pharmacological criteria (adsorption, distribution, metabolism, excretion, and toxicity) and was therefore used in in vitro experiments. Here we demonstrate that CP1 binds with a dissociation constant of 64 ± 1 µM to recombinant chorismate synthase from P. brasiliensis and inhibits enzymatic activity, with a 50% inhibitory concentration (IC50) of 47 ± 5 µM. As expected, CP1 showed no toxicity in three cell lines. On the other hand, CP1 reduced the fungal burden in lungs from treated mice, similar to itraconazole. In addition, histopathological analysis showed that animals treated with CP1 displayed less lung tissue infiltration, fewer yeast cells, and large areas with preserved architecture. Therefore, CP1 was able to control PCM in mice with a lower inflammatory response and is thus a promising candidate and lead structure for the development of drugs useful in PCM treatment.


Subject(s)
Antifungal Agents/pharmacology , Drug Discovery/methods , Paracoccidioides/drug effects , Paracoccidioidomycosis/drug therapy , Phosphorus-Oxygen Lyases/antagonists & inhibitors , Quinolines/pharmacology , Amino Acid Sequence , Animals , Cell Line, Tumor , Disease Models, Animal , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Itraconazole/pharmacology , Male , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Molecular Dynamics Simulation , Paracoccidioides/classification , Paracoccidioides/isolation & purification , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/microbiology , Sequence Analysis, Protein
13.
J Cell Biochem ; 120(6): 10137-10144, 2019 06.
Article in English | MEDLINE | ID: mdl-30548648

ABSTRACT

Mycoplasma pneumoniae (MP) can infect both the upper and lower respiratory tracts. Polydatin (PD), a traditional Chinese medicine, is known to have anti-inflammation and antifibrosis properties. However, the protective effects of PD against MP pneumonia (MPP) remain unclear. So, the aim of this study was to describe the therapeutic effects and underlying mechanisms of PD against MPP. BALB/c mice were assigned to three groups: a normal control group, MP infection group, or PD-treated MP infection group. BEAS-2B cells transfected with or without NACHT domain-, leucine-rich repeat-, and pyd-containing protein 3 (NLRP3) were used to confirm the protective mechanisms of PD. Immunohistochemical analysis, Western blot analysis, enzyme-linked immunosorbent assay, and flow cytometry were used in this study. The results showed that PD treatment suppressed MP-induced lung injury in mice by suppressing the expression of inflammatory factors and inhibiting the development of pulmonary fibrosis. Meanwhile, PD treatment inhibited activation of the NLRP3 inflammasome and nuclear factor κB (NF-κB) pathway. Overexpression of NLRP3 reversed the protective effect of PD against MP-induced injury of BEAS-2B cells. Taken together, these results indicate that PD treatment suppressed the inflammatory response and the development of pulmonary fibrosis by inhibiting the NLRP3 inflammasome and NF-κB pathway after MP infection.


Subject(s)
Glucosides/pharmacology , Inflammasomes/drug effects , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pneumonia, Mycoplasma/prevention & control , Pneumonia/prevention & control , Pulmonary Fibrosis/prevention & control , Stilbenes/pharmacology , Animals , Cell Line , Drugs, Chinese Herbal/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Inflammasomes/metabolism , Lung/cytology , Mice, Inbred BALB C , Pneumonia/metabolism , Pneumonia/microbiology , Pneumonia, Mycoplasma/metabolism , Pneumonia, Mycoplasma/microbiology , Protective Agents/pharmacology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/microbiology , Signal Transduction/drug effects
14.
PLoS One ; 12(11): e0187540, 2017.
Article in English | MEDLINE | ID: mdl-29121069

ABSTRACT

The lungs of cystic fibrosis (CF) patients are chronically colonized by a polymicrobial biofilm community, leading to difficult-to-treat infections. To combat these infections, CF patients are commonly treated with a variety of antibiotics. Understanding the dynamics of polymicrobial community composition in response to antibiotic therapy is essential in the search for novel therapies. Culture-dependent quantification of individual bacteria from defined multispecies biofilms is frequently carried out by plating on selective media. However, the influence of the selective agents in these media on quantitative recovery before or after antibiotic treatment is often unknown. In the present study we developed selective media for six bacterial species that are frequently co-isolated from the CF lung, i.e. Pseudomonas aeruginosa, Staphylococcus aureus, Streptococcus anginosus, Achromobacter xylosoxidans, Rothia mucilaginosa, and Gemella haemolysans. We show that certain supplementations to selective media strongly influence quantitative recovery of (un)treated biofilms. Hence, the developed media were optimized for selectivity and quantitative recovery before or after treatment with antibiotics of four major classes, i.e. ceftazidime, ciprofloxacin, colistin, or tobramycin. Finally, in a proof of concept experiment the novel selective media were applied to determine the community composition of multispecies biofilms before and after treatment with tobramycin.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria , Bacterial Physiological Phenomena/drug effects , Biofilms , Culture Media/chemistry , Pulmonary Fibrosis/microbiology , Bacteria/growth & development , Bacteria/isolation & purification , Biofilms/drug effects , Biofilms/growth & development , Humans
15.
Yale J Biol Med ; 90(2): 219-227, 2017 06.
Article in English | MEDLINE | ID: mdl-28656009

ABSTRACT

Pulmonary fibrosis is a form of lung disease that develops due to aberrant wound-healing following repeated alveoli injury in genetically susceptible individuals, resulting in chronic inflammation, excess deposition of the extracellular matrix components, mainly collagen, and scarring of lung tissue. In addition to irradiation, environmental agents such occupational inhalants, and chemotherapeutic agents, microbial agents also play a role in the etiology of the disease. While viruses have received the most attention, emerging evidence suggest that bacteria and fungi also play a part in the etiology of pulmonary fibrosis. Furthermore, successful use of antibiotics, antiviral and antifungal drugs in several studies to attenuate fibrosis progression is also an indication of microbial involvement in the pathogenesis of the disease and could be a promising therapeutic modality for treating pulmonary fibrosis initiated or exacerbated by infectious agents.


Subject(s)
Pulmonary Fibrosis/etiology , Animals , Anti-Infective Agents/therapeutic use , Bacterial Infections/complications , Disease Models, Animal , Humans , Mice , Mycoses/complications , Pulmonary Fibrosis/microbiology , Pulmonary Fibrosis/prevention & control , Vaccination , Virus Diseases/complications
16.
Am J Respir Cell Mol Biol ; 55(6): 759-766, 2016 12.
Article in English | MEDLINE | ID: mdl-27603223

ABSTRACT

Transforming growth factor-ß (TGFB) regulates cell proliferation, differentiation, apoptosis, and matrix homeostasis and is intimately involved in fibrosis. TGFB expression is increased in fibrotic lung diseases, such as idiopathic pulmonary fibrosis, and in chronic inflammatory conditions, such as chronic obstructive pulmonary disease and asthma. In addition to exhibiting profibrotic activities, the protein exhibits profound immune-suppressive actions involving both innate and adaptive responses, but often this aspect of TGFB biology is overlooked. Recent investigations have demonstrated that TGFB causes wide-ranging immune suppression, including blunting of pivotal early innate IFN responses. These activities permit severe virus infections, often followed by secondary bacterial infections, which may last longer, with augmented inflammation, scarring, fibrosis, and loss of lung function. Strategies to oppose TGFB actions or to enhance IFN responses may help ameliorate the detrimental consequences of infection in patients with diseases characterized by TGFB overexpression, inflammation, and fibrosis.


Subject(s)
Immunity, Innate , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/metabolism , Transforming Growth Factor beta/metabolism , Animals , Humans , Lung/metabolism , Lung/microbiology , Lung/pathology , Lung/virology , Models, Biological , Pulmonary Fibrosis/microbiology , Pulmonary Fibrosis/virology , Signal Transduction
17.
Int J Tuberc Lung Dis ; 20(5): 582-7, 2016 May.
Article in English | MEDLINE | ID: mdl-27084809

ABSTRACT

SETTING: Tertiary referral center, National Institutes of Health (NIH), USA. OBJECTIVE: To estimate the mortality rate and its correlates among persons with pulmonary non-tuberculous mycobacteria (PNTM) disease. DESIGN: A retrospective review of 106 patients who were treated at the NIH Clinical Center and met American Thoracic Society/Infectious Diseases Society of America criteria for PNTM. Eligible patients were aged ⩾18 years and did not have cystic fibrosis or human immunodeficiency virus (HIV) infection. RESULTS: Of 106 patients followed for a median of 4.9 years, 27 (25%) died during follow-up, for a mortality rate of 4.2 per 100 person-years. The population was predominantly female (88%) and White (88%), with infrequent comorbidities. Fibrocavitary disease (adjusted hazard ratio [aHR] 3.3, 95% confidence interval [CI] 1.3-8.3) and pulmonary hypertension (aHR 2.1, 95%CI 0.9-5.1) were associated with a significantly elevated risk of mortality in survival analysis. CONCLUSIONS: PNTM remains a serious public health concern, with a consistently elevated mortality rate across multiple populations. Significant risk factors for death include fibrocavitary disease and pulmonary hypertension. Further research is needed to more specifically identify clinical and microbiologic factors that jointly influence disease outcome.


Subject(s)
Lung/microbiology , Mycobacterium Infections, Nontuberculous/mortality , Nontuberculous Mycobacteria/isolation & purification , Respiratory Tract Infections/mortality , Female , Humans , Hypertension, Pulmonary/microbiology , Hypertension, Pulmonary/mortality , Kaplan-Meier Estimate , Lung/diagnostic imaging , Male , Middle Aged , Mycobacterium Infections, Nontuberculous/diagnostic imaging , Mycobacterium Infections, Nontuberculous/microbiology , National Institutes of Health (U.S.) , Nontuberculous Mycobacteria/classification , Proportional Hazards Models , Pulmonary Fibrosis/microbiology , Pulmonary Fibrosis/mortality , Respiratory Tract Infections/diagnostic imaging , Respiratory Tract Infections/microbiology , Retrospective Studies , Risk Factors , Tertiary Care Centers , Time Factors , Tomography, X-Ray Computed , United States/epidemiology
18.
Respir Med ; 109(12): 1509-15, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26507434

ABSTRACT

BACKGROUND: Chronic pulmonary aspergillosis (CPA) is an occasional complication of allergic bronchopulmonaryaspergillosis (ABPA) but the transition is poorly understood. METHODS: All patients referred to the UK's National Aspergillosis Centre with CPA between May 2009 and June 2012 were screened with serum total IgE and anti-Aspergillus IgE for a dual diagnosis of ABPA and CPA. Those patients suspected of having both conditions were re-evaluated and their imaging reviewed. RESULTS: Of 407 referred patients, 42 screened positive and 22 were confirmed as having both ABPA and CPA. Asthma was present from early childhood in 19 (86%), the median interval between ABPA and onset of CPA was 7.5 years; one patient developed ABPA and CPA simultaneously. Aspergillus IgG levels varied from 23 to 771 mg/L, median 82 mg/L. All 22 patients had bronchiectasis. In patients with ABPA, CT typically demonstrated varicose or cystic bronchiectasis primarily affecting segmental and proximal subsegmental upper lobe bronchi. Other findings included mucoid impaction and centrilobular nodules. Radiological changes associated with CPA included pleural thickening which was often bilateral and accentuated by adjacent hypertrophied extrapleural fat, upper lobe volume loss, thick walled apical cavities, some of which contained aspergillomas, and cavitating pulmonary nodules. CPA secondary to ABPA has more subtle radiological appearances than when due to other underlying diseases. CONCLUSIONS: CPA may complicate ABPA and have distinct radiology features, in addition to bronchiectasis. A novel biomarker is required to anticipate this serious complication, as current serology is not specific enough.


Subject(s)
Asthma/complications , Pulmonary Aspergillosis/complications , Adolescent , Adult , Aged , Aspergillosis, Allergic Bronchopulmonary/complications , Aspergillosis, Allergic Bronchopulmonary/pathology , Asthma/pathology , Child , Child, Preschool , Chronic Disease , Disease Progression , Humans , Infant , Infant, Newborn , Middle Aged , Pulmonary Aspergillosis/pathology , Pulmonary Fibrosis/microbiology , Pulmonary Fibrosis/pathology , Retrospective Studies , Young Adult
19.
Thorax ; 70(7): 636-46, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25964315

ABSTRACT

RATIONALE: Respiratory tract infections are common in patients suffering from pulmonary fibrosis. The interplay between bacterial infection and fibrosis is characterised poorly. OBJECTIVES: To assess the effect of Gram-positive bacterial infection on fibrosis exacerbation in mice. METHODS: Fibrosis progression in response to Streptococcus pneumoniae was examined in two different mouse models of pulmonary fibrosis. MEASUREMENTS AND MAIN RESULTS: We demonstrate that wild-type mice exposed to adenoviral vector delivery of active transforming growth factor-ß1 (TGFß1) or diphteria toxin (DT) treatment of transgenic mice expressing the DT receptor (DTR) under control of the surfactant protein C (SPC) promoter (SPC-DTR) to induce pulmonary fibrosis developed progressive fibrosis following infection with Spn, without exhibiting impaired lung protective immunity against Spn. Antibiotic treatment abolished infection-induced fibrosis progression. The cytotoxin pneumolysin (Ply) of Spn caused this phenomenon in a TLR4-independent manner, as Spn lacking Ply (SpnΔply) failed to trigger progressive fibrogenesis, whereas purified recombinant Ply did. Progressive fibrogenesis was also observed in AdTGFß1-exposed Ply-challenged TLR4 KO mice. Increased apoptotic cell death of alveolar epithelial cells along with an attenuated intrapulmonary release of antifibrogenic prostaglandin E2 was found to underlie progressive fibrogenesis in Ply-challenged AdTGFß1-exposed mice. Importantly, vaccination of mice with the non-cytotoxic Ply derivative B (PdB) substantially attenuated Ply-induced progression of lung fibrosis in AdTGFß1-exposed mice. CONCLUSIONS: Our data unravel a novel mechanism by which infection with Spn through Ply release induces progression of established lung fibrosis, which can be attenuated by protein-based vaccination of mice.


Subject(s)
Pneumonia, Pneumococcal/complications , Pulmonary Fibrosis/microbiology , Streptolysins/physiology , Animals , Anti-Bacterial Agents/therapeutic use , Apoptosis/drug effects , Bacterial Proteins/pharmacology , Bacterial Proteins/physiology , Bronchoalveolar Lavage Fluid/immunology , Diphtheria Toxin , Disease Models, Animal , Disease Progression , Epithelial Cells/drug effects , Female , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pneumococcal Vaccines , Pneumonia, Pneumococcal/drug therapy , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/metabolism , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/prevention & control , Streptolysins/deficiency , Streptolysins/pharmacology , Transforming Growth Factor beta1/metabolism
20.
Mycopathologia ; 179(1-2): 31-43, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25487973

ABSTRACT

Paracoccidioidomycosis (PCM) is a systemic mycosis in which the host response to the infectious agent typically consists of a chronic granulomatous inflammatory process. This condition causes lesions that impair lung function and lead to chronic pulmonary insufficiency resulting from fibrosis development, which is a sequel and disabling feature of the disease. The rPb27 protein has been studied for prophylactic and therapeutic treatment against PCM. Previous studies from our laboratory have shown a protective effect of rPb27 against PCM. However, these studies have not determined whether rPb27 immunization prevents lung fibrosis. We therefore conducted this study to investigate fibrosis resulting from infection by Paracoccidioides brasiliensis in the lungs of animals immunized with rPb27. Animals were immunized with rPb27 and subsequently infected with a virulent strain of P. brasiliensis. Fungal load was evaluated by counting colony-forming units, and Masson's trichrome staining was performed to evaluate fibrosis at 30 and 90 days post-infection. The levels of CCR7, active caspase 3, collagen and cytokines were analyzed. At the two time intervals mentioned, the rPb27 group showed lower levels of fibrosis on histology and reduced levels of collagen and the chemokine receptor CCR7 in the lungs. CCR7 was detected at higher levels in the control groups that developed very high levels of pulmonary fibrosis. Additionally, the immunized groups showed high levels of active caspase 3, IFN-γ, TGF-ß and IL-10 in the early phase of P. brasiliensis infection. Immunization with Pb27, in addition to its protective effect, was shown to prevent pulmonary fibrosis.


Subject(s)
Fungal Proteins/immunology , Paracoccidioides/immunology , Paracoccidioidomycosis/immunology , Pulmonary Fibrosis/prevention & control , Animals , Antifungal Agents/therapeutic use , Antigens, Fungal/immunology , Caspase 3/metabolism , Collagen/metabolism , Fluconazole/therapeutic use , Fungal Proteins/administration & dosage , Fungal Vaccines/immunology , Immunization , Inflammation/immunology , Inflammation/prevention & control , Interferon-gamma/metabolism , Interleukin-10/metabolism , Lung/microbiology , Lung/pathology , Male , Mice , Mice, Inbred BALB C , Paracoccidioidomycosis/microbiology , Paracoccidioidomycosis/prevention & control , Propionibacterium acnes/immunology , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/microbiology , Receptors, CCR7/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...