Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 22(3): 322-335, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33531712

RESUMEN

Immune system dysfunction is paramount in coronavirus disease 2019 (COVID-19) severity and fatality rate. Mucosal-associated invariant T (MAIT) cells are innate-like T cells involved in mucosal immunity and protection against viral infections. Here, we studied the immune cell landscape, with emphasis on MAIT cells, in cohorts totaling 208 patients with various stages of disease. MAIT cell frequency is strongly reduced in blood. They display a strong activated and cytotoxic phenotype that is more pronounced in lungs. Blood MAIT cell alterations positively correlate with the activation of other innate cells, proinflammatory cytokines, notably interleukin (IL)-18, and with the severity and mortality of severe acute respiratory syndrome coronavirus 2 infection. We also identified a monocyte/macrophage interferon (IFN)-α-IL-18 cytokine shift and the ability of infected macrophages to induce the cytotoxicity of MAIT cells in an MR1-dependent manner. Together, our results suggest that altered MAIT cell functions due to IFN-α-IL-18 imbalance contribute to disease severity, and their therapeutic manipulation may prevent deleterious inflammation in COVID-19 aggravation.


Asunto(s)
COVID-19/inmunología , Interferón-alfa/inmunología , Interleucina-18/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Células T Invariantes Asociadas a Mucosa/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Lavado Broncoalveolar , Estudios de Casos y Controles , Chlorocebus aethiops , Estudios de Cohortes , Femenino , Francia , Humanos , Inmunofenotipificación , Interleucina-10/inmunología , Interleucina-15/inmunología , Interleucina-1beta/inmunología , Interleucina-6/inmunología , Interleucina-8/inmunología , Masculino , Persona de Mediana Edad , RNA-Seq , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Análisis de la Célula Individual , Células Vero , Adulto Joven
2.
EMBO J ; 41(12): e108306, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35506364

RESUMEN

Influenza virus infection causes considerable morbidity and mortality, but current therapies have limited efficacy. We hypothesized that investigating the metabolic signaling during infection may help to design innovative antiviral approaches. Using bronchoalveolar lavages of infected mice, we here demonstrate that influenza virus induces a major reprogramming of lung metabolism. We focused on mitochondria-derived succinate that accumulated both in the respiratory fluids of virus-challenged mice and of patients with influenza pneumonia. Notably, succinate displays a potent antiviral activity in vitro as it inhibits the multiplication of influenza A/H1N1 and A/H3N2 strains and strongly decreases virus-triggered metabolic perturbations and inflammatory responses. Moreover, mice receiving succinate intranasally showed reduced viral loads in lungs and increased survival compared to control animals. The antiviral mechanism involves a succinate-dependent posttranslational modification, that is, succinylation, of the viral nucleoprotein at the highly conserved K87 residue. Succinylation of viral nucleoprotein altered its electrostatic interactions with viral RNA and further impaired the trafficking of viral ribonucleoprotein complexes. The finding that succinate efficiently disrupts the influenza replication cycle opens up new avenues for improved treatment of influenza pneumonia.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Gripe Humana , Infecciones por Orthomyxoviridae , Neumonía , Animales , Antivirales/farmacología , Humanos , Subtipo H3N2 del Virus de la Influenza A/metabolismo , Ratones , Proteínas de la Nucleocápside , Nucleoproteínas/metabolismo , Ácido Succínico/metabolismo , Ácido Succínico/farmacología , Ácido Succínico/uso terapéutico , Replicación Viral
3.
Antimicrob Agents Chemother ; 68(5): e0136123, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38526073

RESUMEN

The increasing prevalence of multidrug-resistant Pseudomonas aeruginosa (PA) is a significant concern for chronic respiratory disease exacerbations. Host-directed drugs, such as flagellin, an agonist of toll-like receptor 5 (TLR5), have emerged as a promising solution. In this study, we evaluated the prophylactic intranasal administration of flagellin against a multidrug-resistant strain of PA (PAMDR) in mice and assessed the possible synergy with the antibiotic gentamicin (GNT). The results indicated that flagellin treatment before infection decreased bacterial load in the lungs, likely due to an increase in neutrophil recruitment, and reduced signs of inflammation, including proinflammatory cytokines. The combination of flagellin and GNT showed a synergistic effect, decreasing even more the bacterial load and increasing mice survival rates, in comparison to mice pre-treated only with flagellin. These findings suggest that preventive nasal administration of flagellin could restore the effect of GNT against MDR strains of PA, paving the way for the use of flagellin in vulnerable patients with chronic respiratory diseases.


Asunto(s)
Administración Intranasal , Antibacterianos , Farmacorresistencia Bacteriana Múltiple , Flagelina , Gentamicinas , Infecciones por Pseudomonas , Pseudomonas aeruginosa , Pseudomonas aeruginosa/efectos de los fármacos , Gentamicinas/farmacología , Animales , Flagelina/farmacología , Ratones , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/microbiología , Antibacterianos/farmacología , Femenino , Pulmón/microbiología , Pulmón/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Receptor Toll-Like 5/agonistas , Carga Bacteriana/efectos de los fármacos , Sinergismo Farmacológico
4.
PLoS Pathog ; 17(12): e1010106, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34969061

RESUMEN

The development of safe and effective vaccines in a record time after the emergence of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a remarkable achievement, partly based on the experience gained from multiple viral outbreaks in the past decades. However, the Coronavirus Disease 2019 (COVID-19) crisis also revealed weaknesses in the global pandemic response and large gaps that remain in our knowledge of the biology of coronaviruses (CoVs) and influenza viruses, the 2 major respiratory viruses with pandemic potential. Here, we review current knowns and unknowns of influenza viruses and CoVs, and we highlight common research challenges they pose in 3 areas: the mechanisms of viral emergence and adaptation to humans, the physiological and molecular determinants of disease severity, and the development of control strategies. We outline multidisciplinary approaches and technological innovations that need to be harnessed in order to improve preparedeness to the next pandemic.


Asunto(s)
COVID-19/virología , Gripe Humana/virología , Orthomyxoviridae/fisiología , SARS-CoV-2/fisiología , Animales , Antivirales , COVID-19/terapia , COVID-19/transmisión , Desarrollo de Medicamentos , Evolución Molecular , Humanos , Gripe Humana/terapia , Gripe Humana/transmisión , Orthomyxoviridae/inmunología , SARS-CoV-2/inmunología , Selección Genética , Carga Viral , Vacunas Virales
5.
Int J Mol Sci ; 24(3)2023 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-36769174

RESUMEN

Pseudomonas aeruginosa is a major hospital-associated pathogen that can cause severe infections, most notably in patients with cystic fibrosis (CF) or those hospitalized in intensive care units. Given its remarkable ability to resist antibiotics, P. aeruginosa eradication has grown more challenging. Therefore, there is an urgent need to discover and develop new strategies that can counteract P. aeruginosa-resistant strains. Here, we evaluated the efficacy of poly-L-lysine (pLK) in combination with commonly used antibiotics as an alternative treatment option against P. aeruginosa. First, we demonstrated by scanning electron microscopy that pLK alters the integrity of the surface membrane of P. aeruginosa. We also showed using a fluorometry test that this results in an enhanced permeability of the bacteria membrane. Based on these data, we further evaluated the effect of the combinations of pLK with imipenem, ceftazidime, or aztreonam using the broth microdilution method in vitro. We found synergies in terms of bactericidal effects against either sensitive or resistant P. aeruginosa strains, with a reduction in bacterial growth (up to 5-log10 compared to the control). Similarly, these synergistic and bactericidal effects were confirmed ex vivo using a 3D model of human primary bronchial epithelial cells maintained in an air-liquid interface. In conclusion, pLK could be an innovative antipseudomonal molecule, opening its application as an adjuvant antibiotherapy against drug-resistant P. aeruginosa strains.


Asunto(s)
Infecciones por Pseudomonas , Pseudomonas aeruginosa , Humanos , Polilisina/farmacología , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/microbiología , Pruebas de Sensibilidad Microbiana , Antibacterianos/farmacología
6.
FASEB J ; 35(10): e21838, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34582061

RESUMEN

Inflammation, oxidative stress, and protease/protease inhibitor imbalance with excessive production of proteases are factors associated with pathogenesis of the chronic obstructive pulmonary disease (COPD). In this study, we report that kallikrein-related peptidase 5 (KLK5) is a crucial protease involved in extracellular matrix (ECM) remodeling and bronchial epithelial repair after injury. First, we showed that KLK5 degrades the basal layer formed by culture of primary bronchial epithelial cells from COPD or non-COPD patients. Also, exogenous KLK5 acted differently on BEAS-2B cells already engaged in epithelial-to-mesenchymal transition (EMT) or on 16HBE 14o- cells harboring epithelial characteristics. Indeed, by inducing EMT, KLK5 reduced BEAS-2B cell adherence to the ECM. This effect, neutralized by tissue factor pathway inhibitor 2, a kunitz-type serine protease inhibitor, was due to a direct proteolytic activity of KLK5 on E-cadherin, ß-catenin, fibronectin, and α5ß1 integrin. Thus, KLK5 may strengthen EMT mechanisms and promote the migration of cells by activating the mitogen-activated protein kinase signaling pathway required for this function. In contrast, knockdown of endogenous KLK5 in 16HBE14o- cells, accelerated wound healing repair after injury, and exogenous KLK5 addition delayed the closure repair. These data suggest that among proteases, KLK5 could play a critical role in airway remodeling events associated with COPD during exposure of the pulmonary epithelium to inhaled irritants or smoking and the inflammation process.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Bronquios/patología , Células Epiteliales/patología , Transición Epitelial-Mesenquimal , Calicreínas/metabolismo , Neoplasias Pulmonares/patología , Enfermedad Pulmonar Obstructiva Crónica/patología , Anciano , Antígenos CD/genética , Antígenos CD/metabolismo , Bronquios/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Células Epiteliales/metabolismo , Femenino , Humanos , Calicreínas/genética , Neoplasias Pulmonares/metabolismo , Masculino , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Transducción de Señal
7.
Am J Respir Cell Mol Biol ; 65(4): 378-389, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34102087

RESUMEN

Excessive lung inflammation and airway epithelial damage are hallmarks of human inflammatory lung diseases, such as cystic fibrosis (CF). Enhancement of innate immunity provides protection against pathogens while reducing lung-damaging inflammation. However, the mechanisms underlying innate immunity-mediated protection in the lung remain mysterious, in part because of the lack of appropriate animal models for these human diseases. TLR5 (Toll-like receptor 5) stimulation by its specific ligand, the bacterial protein flagellin, has been proposed to enhance protection against several respiratory infectious diseases, although other cellular events, such as calcium signaling, may also control the intensity of the innate immune response. Here, we investigated the molecular events prompted by stimulation with flagellin and its role in regulating innate immunity in the lung of the pig, which is anatomically and genetically more similar to humans than rodent models. We found that flagellin treatment modulated NF-κB signaling and intracellular calcium homeostasis in airway epithelial cells. Flagellin pretreatment reduced the NF-κB nuclear translocation and the expression of proinflammatory cytokines to a second flagellin stimulus as well as to Pseudomonas aeruginosa infection. Moreover, in vivo administration of flagellin decreased the severity of P. aeruginosa-induced pneumonia. Then we confirmed these beneficial effects of flagellin in a pathological model of CF by using ex vivo precision-cut lung slices from a CF pigz model. These results provide evidence that flagellin treatment contributes to a better regulation of the inflammatory response in inflammatory lung diseases such as CF.


Asunto(s)
Flagelina/farmacología , Inflamación/tratamiento farmacológico , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/patogenicidad , Animales , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Flagelina/inmunología , Flagelina/metabolismo , Inmunidad Innata/efectos de los fármacos , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Transducción de Señal/efectos de los fármacos , Porcinos
8.
Am J Physiol Lung Cell Mol Physiol ; 316(6): L1127-L1140, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30908937

RESUMEN

Host cell proteases are involved in influenza pathogenesis. We examined the role of tissue kallikrein 1 (KLK1) by comparing wild-type (WT) and KLK1-deficient mice infected with influenza H3N2 virus. The levels of KLK1 in lung tissue and in bronchoalveolar lavage (BAL) fluid increased substantially during infection. KLK1 did not promote virus infectivity despite its trypsin-like activity, but it did decrease the initial virus load. We examined two cell types involved in the early control of pathogen infections, alveolar macrophages (AMs) and natural killer (NK) cells to learn more about the antiviral action of KLK1. Inactivating the Klk1 gene or treating WT mice with an anti-KLK1 monoclonal antibody to remove KLK1 activity accelerated the initial virus-induced apoptotic depletion of AMs. Intranasal instillation of deficient mice with recombinant KLK1 (rKLK1) reversed the phenotype. The levels of granulocyte-macrophage colony-stimulating factor in infected BAL fluid were significantly lower in KLK1-deficient mice than in WT mice. Treating lung epithelial cells with rKLK1 increased secretion of this factor known to enhance AM resistance to pathogen-induced apoptosis. The recruitment of NK cells to the air spaces peaked 3 days after infection in WT mice but not in KLK1-deficient mice, as did increases in several NK-attracting chemokines (CCL2, CCL3, CCL5, and CXCL10) in BAL. Chronic obstructive pulmonary disease (COPD) patients are highly susceptible to viral infection, and we observed that the KLK1 mRNA levels decreased with increasing COPD severity. Our findings indicate that KLK1 intervenes early in the antiviral defense modulating the severity of influenza infection. Decreased KLK1 expression in COPD patients could contribute to the worsening of influenza.


Asunto(s)
Apoptosis/fisiología , Macrófagos Alveolares/patología , Infecciones por Orthomyxoviridae/patología , Enfermedad Pulmonar Obstructiva Crónica/patología , Calicreínas de Tejido/metabolismo , Células A549 , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/virología , Animales , Línea Celular , Quimiocina CCL2/metabolismo , Quimiocina CCL3/metabolismo , Quimiocina CCL5/metabolismo , Quimiocina CXCL10/metabolismo , Perros , Factor Estimulante de Colonias de Granulocitos y Macrófagos/análisis , Humanos , Subtipo H3N2 del Virus de la Influenza A , Células Asesinas Naturales/inmunología , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/inmunología , Enfermedad Pulmonar Obstructiva Crónica/virología , Mucosa Respiratoria/metabolismo , Calicreínas de Tejido/antagonistas & inhibidores , Calicreínas de Tejido/genética
9.
Cytokine ; 113: 470-474, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30377053

RESUMEN

Interleukin (IL)-22 plays a critical role in regulating the maintenance of the mucosal barrier. As airway epithelial regeneration is abnormal in cystic fibrosis (CF), we investigated IL-22 integrity in CF. We first demonstrated, using Il-22-/- mice, that IL-22 is important to prevent lung damage induced by the CF pathogen Pseudomonas aeruginosa. Next, IL-22 receptor was found normally expressed at the airway epithelial surfaces of CF patients. In wound-healing assays, IL-22-treated CF cultures had higher wound-closure rate than controls, suggesting that IL-22 signaling per se could be functional in a CF context. However, persistence of neutrophil-derived serine-proteases is a major feature of CF airways. Remarkably, IL-22 was found altered in this protease-rich inflammatory microenvironment; the serine protease-3 being the most prone to fully degrade IL-22. Consequently, we suspect an acquired deficiency of the IL-22 pathway in the lungs of CF patients due to IL-22 cleavage by the surrounding neutrophil serine-proteases.


Asunto(s)
Interleucinas/inmunología , Pulmón/inmunología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Mucosa Respiratoria/inmunología , Adolescente , Adulto , Anciano , Animales , Niño , Fibrosis Quística , Femenino , Humanos , Interleucinas/genética , Pulmón/microbiología , Pulmón/patología , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Infecciones por Pseudomonas/genética , Infecciones por Pseudomonas/patología , Mucosa Respiratoria/microbiología , Mucosa Respiratoria/patología , Interleucina-22
10.
Artículo en Inglés | MEDLINE | ID: mdl-30104272

RESUMEN

The endotracheal tube (ETT) is an essential interface between the patient and ventilator in mechanically ventilated patients. However, a microbial biofilm is formed gradually on this tube and is associated with the development of ventilator-associated pneumonia. The bacteria present in the biofilm are more resistant to antibiotics, and current medical practices do not make it possible to eliminate. Pseudomonas aeruginosa is one of the leading pathogens that cause biofilm infections and ventilator-associated pneumonia. Poly-l-lysine (pLK) is a cationic polypeptide possessing antibacterial properties and mucolytic activity by compacting DNA. Here, we explored the antibiofilm activity of pLK to treat P. aeruginosa biofilms on ETTs while taking into consideration the necessary constraints for clinical translation in our experimental designs. First, we showed that pLK eradicates a P. aeruginosa biofilm formed in vitro on 96-well microplates. We further demonstrated that pLK alters bacterial membrane integrity, as revealed by scanning electron microscopy, and eventually eradicates biofilm formed either by reference or clinical strains of P. aeruginosa biofilms generated in vitro on ETTs. Second, we collected the ETT from patients with P. aeruginosa ventilator-associated pneumonia. We observed that a single dose of pLK is able to immediately disrupt the biofilm structure and kills more than 90% of bacteria present in the biofilm. Additionally, we did not observe any lung tolerance issue when the pLK solution was instilled into the ETT of ventilated pigs, an animal model particularly relevant to mimic invasive mechanical ventilation in humans. In conclusion, pLK appears as an innovative antibiofilm molecule, which could be applied in the ETT of mechanically ventilated patients.


Asunto(s)
Biopelículas/efectos de los fármacos , Intubación Intratraqueal/efectos adversos , Polilisina/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Respiración Artificial/efectos adversos , Animales , Antibacterianos/farmacología , Contaminación de Equipos , Humanos , Microscopía Electrónica de Rastreo/métodos , Neumonía Asociada al Ventilador/tratamiento farmacológico , Porcinos
11.
J Virol ; 91(16)2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28615200

RESUMEN

Hemagglutinin (HA) of influenza virus must be activated by proteolysis before the virus can become infectious. Previous studies indicated that HA cleavage is driven by membrane-bound or extracellular serine proteases in the respiratory tract. However, there is still uncertainty as to which proteases are critical for activating HAs of seasonal influenza A viruses (IAVs) in humans. This study focuses on human KLK1 and KLK5, 2 of the 15 serine proteases known as the kallikrein-related peptidases (KLKs). We find that their mRNA expression in primary human bronchial cells is stimulated by IAV infection. Both enzymes cleaved recombinant HA from several strains of the H1 and/or H3 virus subtype in vitro, but only KLK5 promoted the infectivity of A/Puerto Rico/8/34 (H1N1) and A/Scotland/20/74 (H3N2) virions in MDCK cells. We assessed the ability of treated viruses to initiate influenza in mice. The nasal instillation of only the KLK5-treated virus resulted in weight loss and lethal outcomes. The secretion of this protease in the human lower respiratory tract is enhanced during influenza. Moreover, we show that pretreatment of airway secretions with a KLK5-selective inhibitor significantly reduced the activation of influenza A/Scotland/20/74 virions, providing further evidence of its importance. Differently, increased KLK1 secretion appeared to be associated with the recruitment of inflammatory cells in human airways regardless of the origin of inflammation. Thus, our findings point to the involvement of KLK5 in the proteolytic activation and spread of seasonal influenza viruses in humans.IMPORTANCE Influenza A viruses (IAVs) cause acute infection of the respiratory tract that affects millions of people during seasonal outbreaks every year. Cleavage of the hemagglutinin precursor by host proteases is a critical step in the life cycle of these viruses. Consequently, host proteases that activate HA can be considered promising targets for the development of new antivirals. However, the specific proteases that activate seasonal influenza viruses, especially H3N2 viruses, in the human respiratory tract have remain undefined despite many years of work. Here we demonstrate that the secreted, extracellular protease KLK5 (kallikrein-related peptidase 5) is efficient in promoting the infectivity of H3N2 IAV in vitro and in vivo Furthermore, we found that its secretion was selectively enhanced in the human lower respiratory tract during a seasonal outbreak dominated by an H3N2 virus. Collectively, our data support the clinical relevance of this protease in human influenza pathogenesis.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Interacciones Huésped-Patógeno , Subtipo H3N2 del Virus de la Influenza A/crecimiento & desarrollo , Calicreínas/metabolismo , Animales , Peso Corporal , Células Cultivadas , Modelos Animales de Enfermedad , Células Epiteliales/virología , Humanos , Subtipo H1N1 del Virus de la Influenza A/crecimiento & desarrollo , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Proteolisis , Análisis de Supervivencia
12.
J Biol Chem ; 291(16): 8486-99, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26884336

RESUMEN

The cysteine protease cathepsin C (CatC) activates granule-associated proinflammatory serine proteases in hematopoietic precursor cells. Its early inhibition in the bone marrow is regarded as a new therapeutic strategy for treating proteolysis-driven chronic inflammatory diseases, but its complete inhibition is elusive in vivo Controlling the activity of CatC may be achieved by directly inhibiting its activity with a specific inhibitor or/and by preventing its maturation. We have investigated immunochemically and kinetically the occurrence of CatC and its proform in human hematopoietic precursor cells and in differentiated mature immune cells in lung secretions. The maturation of proCatC obeys a multistep mechanism that can be entirely managed by CatS in neutrophilic precursor cells. CatS inhibition by a cell-permeable inhibitor abrogated the release of the heavy and light chains from proCatC and blocked ∼80% of CatC activity. Under these conditions the activity of neutrophil serine proteases, however, was not abolished in precursor cell cultures. In patients with neutrophilic lung inflammation, mature CatC is found in large amounts in sputa. It is secreted by activated neutrophils as confirmed through lipopolysaccharide administration in a nonhuman primate model. CatS inhibitors currently in clinical trials are expected to decrease the activity of neutrophilic CatC without affecting those of elastase-like serine proteases.


Asunto(s)
Catepsina C/metabolismo , Pulmón/enzimología , Neutrófilos/enzimología , Neumonía/enzimología , Animales , Catepsina C/genética , Modelos Animales de Enfermedad , Células HL-60 , Humanos , Pulmón/patología , Macaca fascicularis , Ratones , Ratones Endogámicos BALB C , Neutrófilos/patología , Neumonía/inducido químicamente , Neumonía/patología , Ratas Sprague-Dawley , Esputo/metabolismo
13.
Eur Respir J ; 50(4)2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-29025886

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory disease of the airways caused mainly by cigarette smoke exposure. COPD progression is marked by exacerbations of the disease, often associated with infections. Recent data show the involvement in COPD pathophysiology of interleukin (IL)-17 and IL-22, two cytokines that are important in the control of lung inflammation and infection. During the initiation and progression of the disease, increased IL-17 secretion causes neutrophil recruitment, leading to chronic inflammation, airways obstruction and emphysema. In the established phase of COPD, a defective IL-22 response facilitates pathogen-associated infections and disease exacerbations. Altered production of these cytokines involves a complex network of immune cells and dysfunction of antigen-presenting cells. In this review, we describe current knowledge on the involvement of IL-17 and IL-22 in COPD pathophysiology at steady state and during exacerbations, and discuss implications for COPD management and future therapeutic approaches.


Asunto(s)
Interleucina-17/inmunología , Interleucinas/inmunología , Enfermedad Pulmonar Obstructiva Crónica , Progresión de la Enfermedad , Descubrimiento de Drogas , Humanos , Inmunidad/efectos de los fármacos , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Interleucina-22
14.
J Infect Dis ; 214(2): 237-47, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27034344

RESUMEN

BACKGROUND: The pathogenesis of influenza A virus (IAV) infections is a multifactorial process that includes the replication capacity of the virus and a harmful inflammatory response to infection. Formyl peptide receptor 2 (FPR2) emerges as a central receptor in inflammatory processes controlling resolution of acute inflammation. Its role in virus pathogenesis has not been investigated yet. METHODS: We used pharmacologic approaches to investigate the role of FPR2 during IAV infection in vitro and in vivo. RESULTS: In vitro, FPR2 expressed on A549 cells was activated by IAV, which harbors its ligand, annexin A1, in its envelope. FPR2 activation by IAV promoted viral replication through an extracellular-regulated kinase (ERK)-dependent pathway. In vivo, activating FPR2 by administering the agonist WKYMVm-NH2 decreased survival and increased viral replication and inflammation after IAV infection. This effect was abolished by treating the mice with U0126, a specific ERK pathway inhibitor, showing that, in vivo, the deleterious role of FPR2 also occurs through an ERK-dependent pathway. In contrast, administration of the FPR2 antagonist WRW4 protected mice from lethal IAV infections. CONCLUSIONS: These data show that viral replication and IAV pathogenesis depend on FPR2 signaling and suggest that FPR2 may be a promising novel strategy to treat influenza.


Asunto(s)
Interacciones Huésped-Patógeno , Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Células Epiteliales/fisiología , Células Epiteliales/virología , Humanos , Virus de la Influenza A/fisiología , Ratones Endogámicos C57BL , Virulencia , Replicación Viral
15.
Am J Physiol Lung Cell Mol Physiol ; 310(7): L689-99, 2016 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-26747784

RESUMEN

Influenza A viruses (IAV) trigger contagious acute respiratory diseases. A better understanding of the molecular mechanisms of IAV pathogenesis and host immune responses is required for the development of more efficient treatments of severe influenza. Calpains are intracellular proteases that participate in diverse cellular responses, including inflammation. Here, we used in vitro and in vivo approaches to investigate the role of calpain signaling in IAV pathogenesis. Calpain expression and activity were found altered in IAV-infected bronchial epithelial cells. With the use of small-interfering RNA (siRNA) gene silencing, specific synthetic inhibitors of calpains, and mice overexpressing calpastatin, we found that calpain inhibition dampens IAV replication and IAV-triggered secretion of proinflammatory mediators and leukocyte infiltration. Remarkably, calpain inhibition has a protective impact in IAV infection, since it significantly reduced mortality of mice challenged not only by seasonal H3N2- but also by hypervirulent H5N1 IAV strains. Hence, our study suggests that calpains are promising therapeutic targets for treating IAV acute pneumonia.


Asunto(s)
Calpaína/fisiología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Gripe Humana/enzimología , Animales , Citocinas/metabolismo , Perros , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Viral de la Expresión Génica , Humanos , Gripe Humana/inmunología , Gripe Humana/virología , Células de Riñón Canino Madin Darby , Ratones Endogámicos C57BL , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal , Replicación Viral
16.
Am J Physiol Lung Cell Mol Physiol ; 311(3): L664-75, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27496898

RESUMEN

Cystic fibrosis (CF) is an inherited disease associated with chronic severe lung inflammation, leading to premature death. To develop innovative anti-inflammatory treatments, we need to characterize new cellular and molecular components contributing to the mechanisms of lung inflammation. Here, we focused on the potential role of "transient receptor potential vanilloid-4" (TRPV4), a nonselective calcium channel. We used both in vitro and in vivo approaches to demonstrate that TRPV4 expressed in airway epithelial cells triggers the secretion of major proinflammatory mediators such as chemokines and biologically active lipids, as well as a neutrophil recruitment in lung tissues. We characterized the contribution of cytosolic phospholipase A2, MAPKs, and NF-κB in TRPV4-dependent signaling. We also showed that 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids, i.e., four natural lipid-based TRPV4 agonists, are present in expectorations of CF patients. Also, TRPV4-induced calcium mobilization and inflammatory responses were enhanced in cystic fibrosis transmembrane conductance regulator-deficient cellular and animal models, suggesting that TRPV4 is a promising target for the development of new anti-inflammatory treatments for diseases such as CF.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Fibrosis Quística/metabolismo , Canales Catiónicos TRPV/fisiología , Células A549 , Animales , Señalización del Calcio , Fibrosis Quística/inmunología , Fibrosis Quística/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Femenino , Humanos , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas Sprague-Dawley
19.
Eur Respir J ; 46(3): 771-82, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26250498

RESUMEN

Chronic obstructive pulmonary disease (COPD) is punctuated by episodes of infection-driven acute exacerbations. Despite the life-threatening nature of these exacerbations, the underlying mechanisms remain unclear, although a high number of neutrophils in the lungs of COPD patients is known to correlate with poor prognosis. Interleukin (IL)-22 is a cytokine that plays a pivotal role in lung antimicrobial defence and tissue protection. We hypothesised that neutrophils secrete proteases that may have adverse effects in COPD, by altering the IL-22 receptor (IL-22R)-dependent signalling.Using in vitro and in vivo approaches as well as reverse transcriptase quantitative PCR, flow cytometry and/or Western blotting techniques, we first showed that pathogens such as the influenza virus promote IL-22R expression in human bronchial epithelial cells, whereas Pseudomonas aeruginosa, bacterial lipopolysaccharide or cigarette smoke do not. Most importantly, neutrophil proteases cleave IL-22R and impair IL-22-dependent immune signalling and expression of antimicrobial effectors such as ß-defensin-2. This proteolysis resulted in the release of a soluble fragment of IL-22R, which was detectable both in cellular and animal models as well as in sputa from COPD patients with acute exacerbations.Hence, our study reveals an unsuspected regulation by the proteolytic action of neutrophil enzymes of IL-22-dependent lung host response. This process probably enhances pathogen replication, and ultimately COPD exacerbations.


Asunto(s)
Células Epiteliales/enzimología , Inmunidad Innata/efectos de los fármacos , Neutrófilos/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/microbiología , Receptores de Interleucina/metabolismo , Animales , Biomarcadores/metabolismo , Western Blotting , Células Cultivadas , Modelos Animales de Enfermedad , Células Epiteliales/microbiología , Humanos , Inmunidad Innata/fisiología , Ratones , Neutrófilos/efectos de los fármacos , Péptido Hidrolasas/metabolismo , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/patogenicidad , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Interleucina/inmunología , Muestreo , Sensibilidad y Especificidad , Fumar/efectos adversos , Estadísticas no Paramétricas , beta-Defensinas/farmacología
20.
J Infect Dis ; 210(2): 214-23, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24493823

RESUMEN

Influenza A virus triggers a contagious respiratory disease that can cause considerable morbidity and mortality. Using an in vitro approach, we previously demonstrated that the pattern recognition receptor retinoic acid-inducible gene I (RIG-I) plays a key role in influenza A virus-mediated immune response. However, the importance of RIG-I signaling in vivo has not been thoroughly examined, because of the lack of an appropriate mouse models. To circumvent this issue, we generated a new transgenic mouse overexpressing LGP2 (hereafter, "LGP2 TG mice"), a major regulator of the RIG-I signaling pathway. The time course of several parameters was compared in infected wild-type and LGP2 TG mice. We found that LGP2 TG mice displayed significantly reduced inflammatory mediators and a lower leukocyte infiltration into the bronchoalveolar airspace. More importantly, LGP2 TG mice had a significant survival advantage. Hence, our in vivo study reveals that LGP2 is a major downregulator of the influenza A virus-triggered detrimental inflammatory response.


Asunto(s)
Interacciones Huésped-Patógeno , Virus de la Influenza A/fisiología , ARN Helicasas/metabolismo , Animales , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Mediadores de Inflamación/análisis , Leucocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Transducción de Señal , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA