Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Respir Crit Care Med ; 209(12): 1453-1462, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38324627

RESUMEN

Rationale: Pseudomonas aeruginosa is the major bacterial pathogen colonizing the airways of adult patients with cystic fibrosis (CF) and causes chronic infections that persist despite antibiotic therapy. Intracellular bacteria may represent an unrecognized reservoir of bacteria that evade the immune system and antibiotic therapy. Although the ability of P. aeruginosa to invade and survive within epithelial cells has been described in vitro in different epithelial cell models, evidence of this intracellular lifestyle in human lung tissues is currently lacking. Objectives: To detect and characterize intracellular P. aeruginosa in CF airway epithelium from human lung explant tissues. Methods: We sampled lung explant tissues from patients with CF undergoing lung transplantation and non-CF lung donor control tissue. We analyzed lung tissue sections for the presence of intracellular P. aeruginosa using quantitative culture and microscopy, in parallel to histopathology and airway morphometry. Measurements and Main Results: P. aeruginosa was isolated from the lungs of seven patients with CF undergoing lung transplantation. Microscopic assessment revealed the presence of intracellular P. aeruginosa within airway epithelial cells in three of the seven patients analyzed at a varying but low frequency. We observed those events occurring in lung regions with high bacterial burden. Conclusions: This is the first study describing the presence of intracellular P. aeruginosa in CF lung tissues. Although intracellular P. aeruginosa in airway epithelial cells is likely relatively rare, our findings highlight the plausible occurrence of this intracellular bacterial reservoir in chronic CF infections.


Asunto(s)
Fibrosis Quística , Trasplante de Pulmón , Pulmón , Infecciones por Pseudomonas , Pseudomonas aeruginosa , Mucosa Respiratoria , Humanos , Fibrosis Quística/microbiología , Fibrosis Quística/complicaciones , Femenino , Masculino , Adulto , Mucosa Respiratoria/microbiología , Mucosa Respiratoria/patología , Infecciones por Pseudomonas/microbiología , Pulmón/microbiología , Pulmón/patología , Adulto Joven , Células Epiteliales/microbiología
2.
PLoS Pathog ; 17(3): e1009375, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33690714

RESUMEN

Pseudomonas aeruginosa causes chronic airway infections, a major determinant of lung inflammation and damage in cystic fibrosis (CF). Loss-of-function lasR mutants commonly arise during chronic CF infections, are associated with accelerated lung function decline in CF patients and induce exaggerated neutrophilic inflammation in model systems. In this study, we investigated how lasR mutants modulate airway epithelial membrane bound ICAM-1 (mICAM-1), a surface adhesion molecule, and determined its impact on neutrophilic inflammation in vitro and in vivo. We demonstrated that LasR-deficient strains induce increased mICAM-1 levels in airway epithelial cells compared to wild-type strains, an effect attributable to the loss of mICAM-1 degradation by LasR-regulated proteases and associated with enhanced neutrophil adhesion. In a subacute airway infection model, we also observed that lasR mutant-infected mice displayed greater airway epithelial ICAM-1 expression and increased neutrophilic pulmonary inflammation. Our findings provide new insights into the intricate interplay between lasR mutants, LasR-regulated proteases and airway epithelial ICAM-1 expression, and reveal a new mechanism involved in the exaggerated inflammatory response induced by lasR mutants.


Asunto(s)
Fibrosis Quística/complicaciones , Neumonía/microbiología , Pseudomonas aeruginosa/patogenicidad , Sistema Respiratorio/parasitología , Animales , Proteínas Bacterianas/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Regulación Bacteriana de la Expresión Génica/fisiología , Humanos , Ratones , Neumonía/complicaciones , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa/metabolismo , Sistema Respiratorio/metabolismo , Transactivadores/genética
3.
Clin Proteomics ; 20(1): 44, 2023 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-37875801

RESUMEN

The quest for understanding and managing the long-term effects of COVID-19, often referred to as Long COVID or post-COVID-19 condition (PCC), remains an active research area. Recent findings highlighted angiopoietin-1 (ANG-1) and p-selectin (P-SEL) as potential diagnostic markers, but validation is essential, given the inconsistency in COVID-19 biomarker studies. Leveraging the biobanque québécoise de la COVID-19 (BQC19) biobank, we analyzed the data of 249 participants. Both ANG-1 and P-SEL levels were significantly higher in patients with PCC participants compared with control subjects at 3 months using the Mann-Whitney U test. We managed to reproduce and validate the findings, emphasizing the importance of collaborative biobanking efforts in enhancing the reproducibility and credibility of Long COVID research outcomes.

4.
PLoS Comput Biol ; 17(3): e1008810, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33684134

RESUMEN

Abnormal coagulation and an increased risk of thrombosis are features of severe COVID-19, with parallels proposed with hemophagocytic lymphohistiocytosis (HLH), a life-threating condition associated with hyperinflammation. The presence of HLH was described in severely ill patients during the H1N1 influenza epidemic, presenting with pulmonary vascular thrombosis. We tested the hypothesis that genes causing primary HLH regulate pathways linking pulmonary thromboembolism to the presence of SARS-CoV-2 using novel network-informed computational algorithms. This approach led to the identification of Neutrophils Extracellular Traps (NETs) as plausible mediators of vascular thrombosis in severe COVID-19 in children and adults. Taken together, the network-informed analysis led us to propose the following model: the release of NETs in response to inflammatory signals acting in concert with SARS-CoV-2 damage the endothelium and direct platelet-activation promoting abnormal coagulation leading to serious complications of COVID-19. The underlying hypothesis is that genetic and/or environmental conditions that favor the release of NETs may predispose individuals to thrombotic complications of COVID-19 due to an increase risk of abnormal coagulation. This would be a common pathogenic mechanism in conditions including autoimmune/infectious diseases, hematologic and metabolic disorders.


Asunto(s)
COVID-19/complicaciones , COVID-19/genética , Trampas Extracelulares/genética , Linfohistiocitosis Hemofagocítica/complicaciones , Linfohistiocitosis Hemofagocítica/genética , Modelos Biológicos , SARS-CoV-2/genética , Trombosis/etiología , Trombosis/genética , Algoritmos , Degranulación de la Célula/genética , Biología Computacional , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Predisposición Genética a la Enfermedad , Humanos , Pandemias , Mapas de Interacción de Proteínas , Embolia Pulmonar/etiología , Embolia Pulmonar/genética , Proteínas Virales/genética
5.
J Immunol ; 204(8): 2285-2294, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32169849

RESUMEN

Neutrophils promote tumor growth and metastasis at multiple stages of cancer progression. One mechanism through which this occurs is via release of neutrophil extracellular traps (NETs). We have previously shown that NETs trap tumor cells in both the liver and the lung, increasing their adhesion and metastasis following postoperative complications. Multiple studies have since shown that NETs play a role in tumor progression and metastasis. NETs are composed of nuclear DNA-derived web-like structures decorated with neutrophil-derived proteins. However, it is unknown which, if any, of these NET-affiliated proteins is responsible for inducing the metastatic phenotype. In this study, we identify the NET-associated carcinoembryonic Ag cell adhesion molecule 1 (CEACAM1) as an essential element for this interaction. Indeed, blocking CEACAM1 on NETs, or knocking it out in a murine model, leads to a significant decrease in colon carcinoma cell adhesion, migration and metastasis. Thus, this work identifies NET-associated CEACAM1 as a putative therapeutic target to prevent the metastatic progression of colon carcinoma.


Asunto(s)
Antígenos CD/metabolismo , Antígeno Carcinoembrionario/metabolismo , Moléculas de Adhesión Celular/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Trampas Extracelulares/inmunología , Trampas Extracelulares/metabolismo , Neutrófilos/inmunología , Células A549 , Animales , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Células HT29 , Humanos , Ratones , Neutrófilos/patología
6.
Biochemistry ; 60(50): 3829-3840, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34845903

RESUMEN

Catalytic promiscuity is the coincidental ability to catalyze nonbiological reactions in the same active site as the native biological reaction. Several lines of evidence show that catalytic promiscuity plays a role in the evolution of new enzyme functions. Thus, studying catalytic promiscuity can help identify structural features that predispose an enzyme to evolve new functions. This study identifies a potentially preadaptive residue in a promiscuous N-succinylamino acid racemase/o-succinylbenzoate synthase (NSAR/OSBS) enzyme from Amycolatopsis sp. T-1-60. This enzyme belongs to a branch of the OSBS family which includes many catalytically promiscuous NSAR/OSBS enzymes. R266 is conserved in all members of the NSAR/OSBS subfamily. However, the homologous position is usually hydrophobic in other OSBS subfamilies, whose enzymes lack NSAR activity. The second-shell amino acid R266 is close to the catalytic acid/base K263, but it does not contact the substrate, suggesting that R266 could affect the catalytic mechanism. Mutating R266 to glutamine in Amycolatopsis NSAR/OSBS profoundly reduces NSAR activity but moderately reduces OSBS activity. This is due to a 1000-fold decrease in the rate of proton exchange between the substrate and the general acid/base catalyst K263. This mutation is less deleterious for the OSBS reaction because K263 forms a cation-π interaction with the OSBS substrate and/or the intermediate, rather than acting as a general acid/base catalyst. Together, the data explain how R266 contributes to NSAR reaction specificity and was likely an essential preadaptation for the evolution of NSAR activity.


Asunto(s)
Isomerasas de Aminoácido/química , Isomerasas de Aminoácido/metabolismo , Liasas de Carbono-Carbono/química , Liasas de Carbono-Carbono/metabolismo , Isomerasas de Aminoácido/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Amycolatopsis/enzimología , Amycolatopsis/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biocatálisis , Liasas de Carbono-Carbono/genética , Dominio Catalítico/genética , Secuencia Conservada , Cristalografía por Rayos X , Estabilidad de Enzimas/genética , Evolución Molecular , Cinética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
7.
Anal Bioanal Chem ; 413(20): 5135-5146, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34173039

RESUMEN

Drug safety assessment in the early phases of drug discovery is critical to facilitate the rapid development of novel therapeutics. Recently, teleost zebrafish (Danio rerio) has emerged as a promising vertebrate model for the assessment of drug safety. Zebrafish is a convenient model because of its small size, high fecundity, embryo transparency, and ex utero development. In this study, we developed a matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) method applied to zebrafish larvae to investigate safety and metabolism of sahaquine (Sq), an anticancer agent inhibiting histone deacetylase 6. This technique improves on prior studies using liquid chromatography-mass spectrometry (LC-MS) by adding analysis of the drug spatial distribution. Using this method, it was determined that Sq dissolved in fish water (1-2000 µM) did not reach the larval body and was mainly distributed throughout the yolk. High Sq concentration (800 µM) administered intravenously allowed the compound to reach the larval body but did not induce phenotypic abnormalities. Sq was metabolized into its glucuronidated form within 24 h and was excreted within 72 h. MALDI MSI was instrumental in showing that Sq-glucuronide was mainly formed in the gut and slightly in yolk syncytial layer, and provided valuable insights into xenobiotics elimination in zebrafish larvae. This study indicates that Sq has a good safety profile and merits further investigations in other disease models. In addition, the optimized MALDI MSI protocol provided here can be widely applied to study distribution and metabolic fate of other structurally related molecules.


Asunto(s)
Espectrometría de Masas/métodos , Animales , Línea Celular Tumoral , Embrión no Mamífero/efectos de los fármacos , Humanos , Larva/efectos de los fármacos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Pez Cebra
8.
J Allergy Clin Immunol ; 143(2): 591-603.e3, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29935218

RESUMEN

BACKGROUND: The effect of Staphylococcus aureus on nasal epithelial repair has never been assessed in patients with chronic rhinosinusitis with nasal polyps (CRSwNP). OBJECTIVE: This study aimed to determine whether (1) nasal epithelial cell cultures from patients with CRSwNP and control subjects repair differently; (2) S aureus exoproducts compromise nasal epithelial repair; (3) S aureus alters lamellipodial dynamics; and (4) deleterious effects could be counteracted by the Rho-associated coiled-coil kinase inhibitor Y-27632. METHODS: Primary nasal epithelial cells (pNECs) collected during surgeries were cultured and injured under 3 conditions: (1) basal conditions, (2) exposed to S aureus exoproducts, and (3) exposed to S aureus exoproducts and Y-27632. Epithelial repair, lamellipodial dynamics, and cytoskeletal organization were assessed. RESULTS: Under basal conditions, pNEC cultures from patients with CRSwNP presented significantly lower repair rates and reduced lamellipodial protrusion length and velocity than those from control subjects. S aureus exoproducts significantly decreased repair rates and protrusion dynamics in both control subjects and patients with CRSwNP; however, the effect of S aureus on cell protrusions was more sustained over time in patients with CRSwNP. Under basal conditions, immunofluorescence assays showed significantly reduced percentages of cells with lamellipodia at the wound edge in patients with CRSwNP compared with control subjects. S aureus altered cell polarity and decreased the percentage of cells with lamellipodia in both groups. Finally, Y-27632 prevented the deleterious effects of S aureus exoproducts on CRSwNP repair rates, as well as on lamellipodial dynamics and formation. CONCLUSIONS: S aureus exoproducts significantly alter epithelial repair and lamellipodial dynamics on pNECs, and this impairment was more pronounced in patients with CRSwNP. Importantly, Y-27632 restored epithelial repair and lamellipodial dynamics in the presence of S aureus exoproducts.


Asunto(s)
Pólipos Nasales/inmunología , Senos Paranasales/patología , Mucosa Respiratoria/fisiología , Rinitis/inmunología , Sinusitis/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/fisiología , Adulto , Anciano , Amidas/farmacología , Células Cultivadas , Enfermedad Crónica , Citoesqueleto/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Senos Paranasales/microbiología , Piridinas/farmacología , Mucosa Respiratoria/patología , Cicatrización de Heridas , Quinasas Asociadas a rho/metabolismo
10.
Am J Physiol Lung Cell Mol Physiol ; 314(4): L635-L641, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29351441

RESUMEN

Cystic fibrosis patients exhibit chronic Pseudomonas aeruginosa respiratory infections and sustained proinflammatory state favoring lung tissue damage and remodeling, ultimately leading to respiratory failure. Loss of cystic fibrosis transmembrane conductance regulator (CFTR) function is associated with MAPK hyperactivation and increased cytokines expression, such as interleukin-8 [chemoattractant chemokine (C-X-C motif) ligand 8 (CXCL8)]. Recently, new therapeutic strategies directly targeting the basic CFTR defect have been developed, and ORKAMBI (Vx-809/Vx-770 combination) is the only Food and Drug Administration-approved treatment for CF patients homozygous for the F508del mutation. Here we aimed to determine the effect of the Vx-809/Vx-770 combination on the induction of the inflammatory response by fully differentiated primary bronchial epithelial cell cultures from CF patients carrying F508del mutations, following exposure to P. aeruginosa exoproducts. Our data unveiled that CFTR functional rescue with Vx-809/Vx-770 drastically reduces CXCL8 (as well as CXCL1 and CXCL2) transcripts and p38 MAPK phosphorylation in response to P. aeruginosa exposure through a CFTR-dependent mechanism. These results suggest that ORKAMBI has anti-inflammatory properties that could decrease lung inflammation and contribute to the observed beneficial impact of this treatment in CF patients.


Asunto(s)
Aminofenoles/uso terapéutico , Aminopiridinas/uso terapéutico , Benzodioxoles/uso terapéutico , Bronquios/inmunología , Fibrosis Quística/tratamiento farmacológico , Células Epiteliales/inmunología , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/aislamiento & purificación , Quinolonas/uso terapéutico , Bronquios/efectos de los fármacos , Bronquios/patología , Células Cultivadas , Agonistas de los Canales de Cloruro/uso terapéutico , Fibrosis Quística/inmunología , Fibrosis Quística/microbiología , Fibrosis Quística/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Humanos , Interleucina-8/metabolismo , Mutación , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/patología
11.
Sensors (Basel) ; 18(4)2018 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-29587396

RESUMEN

In this paper, we present a new mobile wireless communication platform for real-time monitoring of an individual's breathing rate. The platform takes the form of a wearable stretching T-shirt featuring a sensor and a detection base station. The sensor is formed by a spiral-shaped antenna made from a multi-material fiber connected to a compact transmitter. Based on the resonance frequency of the antenna at approximately 2.4 GHz, the breathing sensor relies on its Bluetooth transmitter. The contactless and non-invasive sensor is designed without compromising the user's comfort. The sensing mechanism of the system is based on the detection of the signal amplitude transmitted wirelessly by the sensor, which is found to be sensitive to strain. We demonstrate the capability of the platform to detect the breathing rates of four male volunteers who are not in movement. The breathing pattern is obtained through the received signal strength indicator (RSSI) which is filtered and analyzed with home-made algorithms in the portable system. Numerical simulations of human breath are performed to support the experimental detection, and both results are in a good agreement. Slow, fast, regular, irregular, and shallow breathing types are successfully recorded within a frequency interval of 0.16-1.2 Hz, leading to a breathing rate varying from 10 to 72 breaths per minute.

12.
Int J Cancer ; 140(10): 2321-2330, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28177522

RESUMEN

Despite advances in cancer treatment, metastasis remains today the main cause of cancer death. Local control through complete surgical resection of the primary tumor continues to be a key principle in cancer treatment. However, surgical interventions themselves lead to adverse oncologic outcomes and are associated with significantly increased rates of metastasis. Neutrophils through release of neutrophil extracellular traps (NETs) in response to infections were shown to be able to capture circulating cancer cells, and in doing so, support the development of metastatic disease. To be able to intervene on this process, understanding the exact molecular nature of these mechanisms is crucial. We therefore hypothesize and demonstrate that ß1-integrin is an important factor mediating the interactions between circulating tumor cells and NETs. We show that ß1-integrin expression on both cancer cells and NETs is important for the adhesion of circulating tumor cells to NETs both in vitro and in vivo. Using a murine model of intra-abdominal sepsis to mimic the postoperative inflammatory environment, we show that ß1-integrin expression is upregulated in the context of inflammation in vivo. Ultimately, we show that this increased early cancer cell adhesion to NETs in vivo and this effect is abrogated when mice are administered DNAse 1. Our data therefore sheds light on the first molecular mechanism by which NETs can trap circulating tumor cells (CTCs), broadening our understanding of this process.


Asunto(s)
Trampas Extracelulares/metabolismo , Inflamación/patología , Integrina beta1/metabolismo , Neoplasias Pulmonares/patología , Células Neoplásicas Circulantes/patología , Neutrófilos/patología , Animales , Western Blotting , Adhesión Celular , Proliferación Celular , Modelos Animales de Enfermedad , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Inflamación/metabolismo , Integrina beta1/química , Integrina beta1/genética , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Células Neoplásicas Circulantes/metabolismo , Infiltración Neutrófila , ARN Interferente Pequeño/genética , Células Tumorales Cultivadas
13.
Int J Cancer ; 141(3): 561-571, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28401532

RESUMEN

Surgical resection of early stage nonsmall cell lung cancer (NSCLC) is necessary for cure. However, rates of postoperative bacterial pneumonias remain high and may confer an increased risk for metastasis. Toll-like receptors (TLRs) mediate the inflammatory cascade by recognizing microbial products at the surface of numerous cell types in the lung; however, little is known about how host TLRs influence NSCLC metastasis. TLR2 recognizes gram-positive bacterial cell wall components activating innate immunity. We demonstrate that lower respiratory tract infection with Streptococcus pneumonia augments the formation of murine H59 NSCLC liver metastases in C57BL/6 mice through host TLR2 activation. Infected mice demonstrate increased H59 and human A549 NSCLC adhesion to hepatic sinusoids in vivo compared with noninfected controls, a response that is significantly diminished in TLR2 knock-out mice. Intra-tracheal injection of purified TLR2 ligand lipoteichoic acid into mice similarly augments in vivo adhesion of H59 cells to hepatic sinusoids. Additionally, H59 and A549 NSCLC cells incubated with bronchoepithelial conditioned media show increased cell adhesion to extracellular matrix components in vitro and hepatic sinusoids in vivo in a manner that is dependent on bronchoepithelial TLR2 activation and interleukin-6 secretion. TLR2 is therefore a potential therapeutic target for gram-positive pneumonia-driven NSCLC metastasis.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/patología , Neumonía/complicaciones , Streptococcus pneumoniae/patogenicidad , Receptor Toll-Like 2/metabolismo , Animales , Apoptosis , Carcinoma de Pulmón de Células no Pequeñas/etiología , Adhesión Celular , Proliferación Celular , Humanos , Lipopolisacáridos/farmacología , Neoplasias Hepáticas/etiología , Neoplasias Pulmonares/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía/microbiología , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Biochem Biophys Res Commun ; 483(2): 829-833, 2017 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-28062182

RESUMEN

Chronic bacterial infections in cystic fibrosis lung disease are often characterized by Pseudomonas aeruginosa biofilms that are regulated by bacterial intercellular signals termed quorum sensing (QS), such as N-(3-oxododecanoyl)-l-homoserine lactone (3OC12-HSL). This study reports that biofilm-derived exoproducts decrease the transcriptional activity of the anti-oxidant response element in bronchial epithelial cells. In a live co-culture assay of BEAS-2B cells and P. aeruginosa biofilm, the QS molecule 3OC12-HSL was an important but not sole contributor to the inhibition of basal NRF2 luciferase reporter activity. Moreover, biofilm-derived exoproducts and 3OC12-HSL decrease the expression of endogenous antioxidant response element-regulated genes hemeoxygenase-1 (HO-1) and NAD(P)H Quinone Dehydrogenase-1 (NQO-1) while they increase IL-8 expression. As previously reported, IL-8 expression is partially dependent on p38 MAPK activity, but the inhibitory effect of biofilm QS molecules on HO-1 and NQO-1 expression occurs independently of this protein kinase. Finally, the transfection of CFTRdelF508 but not its wild type counterpart decreases basal, planktonic PsaDM and sulforaphane-driven NRF2 luciferase reporter activity in BEAS-2B cells. Therefore, the presence of quorum sensing molecules derived from bacterial biofilms lowers the transcriptional activity of the anti-oxidant response element, which may contribute to the establishment of chronic bacterial infections, especially in the presence of mutated CFTR. Increasing NRF2 activity may thus be a promising strategy to potentiate anti-biofilm activity in cystic fibrosis lung disease.


Asunto(s)
Elementos de Respuesta Antioxidante , Factor 2 Relacionado con NF-E2/metabolismo , Pseudomonas aeruginosa/patogenicidad , Percepción de Quorum/fisiología , 4-Butirolactona/análogos & derivados , 4-Butirolactona/metabolismo , Biopelículas , Línea Celular , Fibrosis Quística/microbiología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Regulación de la Expresión Génica , Hemo-Oxigenasa 1/genética , Homoserina/análogos & derivados , Homoserina/metabolismo , Humanos , Interleucina-8/genética , Pulmón/metabolismo , Pulmón/microbiología , Pulmón/patología , NAD(P)H Deshidrogenasa (Quinona)/genética , Factor 2 Relacionado con NF-E2/genética , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa/metabolismo
15.
Biochem Biophys Res Commun ; 482(4): 645-650, 2017 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-27865842

RESUMEN

Niemann-Pick disease (NPD) type B is a rare autosomal recessive disease characterized by variable levels of impairment in sphingomyelin phosphodiesterase 1 (SMPD1) activity. Lung involvement is the most important prognostic factor in NPD-B, with recurrent respiratory infections starting in infancy being the major cause of morbidity and mortality. We hypothesized that decreased SMPD1 activity impaired airway epithelium host defense response. SMPD1 activity was reduced using inducible shRNA. Surprisingly, decreasing SMPD1 activity by 50%, resulted in increased neutrophil recruitment, both at baseline and in response to bacterial stimulation. This correlated with elevated levels of cytokine mRNA shown to contribute to neutrophil recruitment in unstimulated (e.g. IL-8 and GRO-α) and infected cells (e.g. IL-8, GRO-α, GM-CSF and CCL20). Instead of preventing the host defence responses, decreased SMPD1 activity results in an inflammatory response even in the absence of infection. Moreover, decreasing SMPD1 activity resulted in a pro-oxidative shift. Accordingly, expression of an inactive mutant, SMPD1[L225P] but not the WT enzyme increased activation of the antioxidant transcription factor NRF2. Therefore, decreasing SMPD1 activity by 50% in airway epithelial cells, the equivalent of the loss of one allele, results in the accumulation of oxidants that activates NRF2 and a concomitant increased cytokine production as well as neutrophil recruitment. This can result in a chronic inflammatory state that impairs host defence similar to scenarios observe in other chronic inflammatory lung disease such as Chronic Obstructive Pulmonary Disease or Cystic Fibrosis.


Asunto(s)
Citocinas/inmunología , Factor 2 Relacionado con NF-E2/inmunología , Infiltración Neutrófila , Enfermedad de Niemann-Pick Tipo B/inmunología , Mucosa Respiratoria/inmunología , Esfingomielina Fosfodiesterasa/inmunología , Bronquios/citología , Bronquios/inmunología , Bronquios/patología , Línea Celular , Humanos , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Especies Reactivas de Oxígeno/inmunología , Mucosa Respiratoria/citología , Mucosa Respiratoria/patología
16.
RNA ; 21(2): 262-78, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25525152

RESUMEN

The nuclear exosome targeting complex (NEXT) directs a major 3'-5' exonuclease, the RNA exosome, for degradation of nuclear noncoding (nc) RNAs. We identified the RNA-binding component of the NEXT complex, RBM7, as a substrate of p38(MAPK)/MK2-mediated phosphorylation at residue S136. As a result of this phosphorylation, RBM7 displays a strongly decreased RNA-binding capacity, while inhibition of p38(MAPK) or mutation of S136A in RBM7 increases its RNA association. Interestingly, promoter-upstream transcripts (PROMPTs), such as proRBM39, proEXT1, proDNAJB4, accumulated upon stress stimulation in a p38(MAPK)/MK2-dependent manner, a process inhibited by overexpression of RBM7(S136A). While there are no stress-dependent changes in RNA-polymerase II (RNAPII) occupation of PROMPT regions representing unchanged transcription, stability of PROMPTs is increased. Hence, we propose that phosphorylation of RBM7 by the p38(MAPK)/MK2 axis increases nuclear ncRNA stability by blocking their RBM7-binding and subsequent RNA exosome targeting to allow stress-dependent modulations of the noncoding transcriptome.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Portadoras/metabolismo , Núcleo Celular/metabolismo , Exosomas , Células HEK293 , Células HeLa , Humanos , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Fosforilación , Unión Proteica , Estabilidad del ARN , ARN no Traducido/genética , ARN no Traducido/metabolismo , Estrés Fisiológico
17.
FASEB J ; 30(9): 3011-25, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27178322

RESUMEN

Chronic Pseudomonas aeruginosa lung infections are associated with progressive epithelial damage and lung function decline. In addition to its role in tissue injury, the persistent presence of P. aeruginosa-secreted products may also affect epithelial repair ability, raising the need for new antivirulence therapies. The purpose of our study was to better understand the outcomes of P. aeruginosa exoproducts exposure on airway epithelial repair processes to identify a strategy to counteract their deleterious effect. We found that P. aeruginosa exoproducts significantly decreased wound healing, migration, and proliferation rates, and impaired the ability of directional migration of primary non-cystic fibrosis (CF) human airway epithelial cells. Impact of exoproducts was inhibited after mutations in P. aeruginosa genes that encoded for the quorum-sensing (QS) transcriptional regulator, LasR, and the elastase, LasB, whereas impact was restored by LasB induction in ΔlasR mutants. P. aeruginosa purified elastase also induced a significant decrease in non-CF epithelial repair, whereas protease inhibition with phosphoramidon prevented the effect of P. aeruginosa exoproducts. Furthermore, treatment of P. aeruginosa cultures with 4-hydroxy-2,5-dimethyl-3(2H)-furanone, a QS inhibitor, abrogated the negative impact of P. aeruginosa exoproducts on airway epithelial repair. Finally, we confirmed our findings in human airway epithelial cells from patients with CF, a disease featuring P. aeruginosa chronic respiratory infection. These data demonstrate that secreted proteases under the control of the LasR QS system impair airway epithelial repair and that QS inhibitors could be of benefit to counteract the deleterious effect of P. aeruginosa in infected patients.-Ruffin, M., Bilodeau, C., Maillé, É., LaFayette, S. L., McKay, G. A., Trinh, N. T. N., Beaudoin, T., Desrosiers, M.-Y., Rousseau, S., Nguyen, D., Brochiero, E. Quorum-sensing inhibition abrogates the deleterious impact of Pseudomonas aeruginosa on airway epithelial repair.


Asunto(s)
Proteínas Bacterianas/metabolismo , Células Epiteliales/microbiología , Pseudomonas aeruginosa/fisiología , Movimiento Celular , Proliferación Celular , Células Cultivadas , Regulación Bacteriana de la Expresión Génica/fisiología , Humanos , Mutación , Mucosa Respiratoria/citología , Sistema Respiratorio
18.
Chemphyschem ; 18(21): 2999-3006, 2017 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-28759147

RESUMEN

The Hansen solubility parameter approach is revisited by implementing the thermodynamics of dissolution and mixing. Hansen's pragmatic approach has earned its spurs in predicting solvents for polymer solutions, but for molecular solutes improvements are needed. By going into the details of entropy and enthalpy, several corrections are suggested that make the methodology thermodynamically sound without losing its ease of use. The most important corrections include accounting for the solvent molecules' size, the destruction of the solid's crystal structure, and the specificity of hydrogen-bonding interactions, as well as opportunities to predict the solubility at extrapolated temperatures. Testing the original and the improved methods on a large industrial dataset including solvent blends, fit qualities improved from 0.89 to 0.97 and the percentage of correct predictions rose from 54 % to 78 %. Full Matlab scripts are included in the Supporting Information, allowing readers to implement these improvements on their own datasets.

19.
Proc Natl Acad Sci U S A ; 111(20): E2110-9, 2014 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-24799673

RESUMEN

To subvert host immunity, influenza A virus (IAV) induces early apoptosis in innate immune cells by disrupting mitochondria membrane potential via its polymerase basic protein 1-frame 2 (PB1-F2) accessory protein. Whether immune cells have mechanisms to counteract PB1-F2-mediated apoptosis is currently unknown. Herein, we define that the host mitochondrial protein nucleotide-binding oligomerization domain-like receptor (NLR)X1 binds to viral protein PB1-F2, preventing IAV-induced macrophage apoptosis and promoting both macrophage survival and type I IFN signaling. We initially observed that Nlrx1-deficient mice infected with IAV exhibited increased pulmonary viral replication, as well as enhanced inflammatory-associated pulmonary dysfunction and morbidity. Analysis of the lungs of IAV-infected mice revealed markedly enhanced leukocyte recruitment but impaired production of type I IFN in Nlrx1(-/-) mice. Impaired type I IFN production and enhanced viral replication was recapitulated in Nlrx1(-/-) macrophages and was associated with increased mitochondrial mediated apoptosis. Through gain- and loss-of-function strategies for protein interaction, we identified that NLRX1 directly bound PB1-F2 in the mitochondria of macrophages. Using a recombinant virus lacking PB1-F2, we confirmed that deletion of PB1-F2 abrogated NLRX1-dependent macrophage type I IFN production and apoptosis. Thus, our results demonstrate that NLRX1 acts as a mitochondrial sentinel protecting macrophages from PB1-F2-induced apoptosis and preserving their antiviral function. We further propose that NLRX1 is critical for macrophage immunity against IAV infection by sensing the extent of viral replication and maintaining a protective balance between antiviral immunity and excessive inflammation within the lungs.


Asunto(s)
Apoptosis , Virus de la Influenza A/inmunología , Macrófagos/inmunología , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Virales/metabolismo , Animales , Línea Celular Tumoral , Humanos , Inflamación , Virus de la Influenza A/fisiología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Mitocondriales/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Replicación Viral
20.
J Immunol ; 193(3): 1416-26, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24981451

RESUMEN

Airway smooth muscle cell (ASMC) migration is an important mechanism postulated to play a role in airway remodeling in asthma. CXCL1 chemokine has been linked to tissue growth and metastasis. In this study, we present a detailed examination of the inhibitory effect of CXCL1 on human primary ASMC migration and the role of the decoy receptor, Duffy AgR for chemokines (DARC), in this inhibition. Western blots and pathway inhibitors showed that this phenomenon was mediated by activation of the ERK-1/2 MAPK pathway, but not p38 MAPK or PI3K, suggesting a biased selection in the signaling mechanism. Despite being known as a nonsignaling receptor, small interference RNA knockdown of DARC showed that ERK-1/2 MAPK activation was significantly dependent on DARC functionality, which, in turn, was dependent on the presence of heat shock protein 90 subunit α. Interestingly, DARC- or heat shock protein 90 subunit α-deficient ASMCs responded to CXCL1 stimulation by enhancing p38 MAPK activation and ASMC migration through the CXCR2 receptor. In conclusion, we demonstrated DARC's ability to facilitate CXCL1 inhibition of ASMC migration through modulation of the ERK-1/2 MAPK-signaling pathway.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/inmunología , Inhibición de Migración Celular/inmunología , Quimiocina CXCL1/fisiología , Sistema del Grupo Sanguíneo Duffy/fisiología , Receptores de Superficie Celular/fisiología , Receptores de Interleucina-8B/fisiología , Biomarcadores/metabolismo , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/fisiología , Sistema del Grupo Sanguíneo Duffy/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/inmunología , Cultivo Primario de Células , Receptores de Superficie Celular/metabolismo , Receptores de Interleucina-8B/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA