Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Molecules ; 25(23)2020 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-33255656

RESUMEN

Staphylococcus aureus (S. aureus)-induced acute lung injury (ALI) is a serious disease that has a high risk of death among infants and teenagers. Acetylharpagide, a natural compound of Ajuga decumbens Thunb. (family Labiatae), has been found to have anti-tumor, anti-inflammatory and anti-viral effects. This study investigates the therapeutic effects of acetylharpagide on S. aureus-induced ALI in mice. Here, we found that acetylharpagide alleviated S. aureus-induced lung pathological morphology damage, protected the pulmonary blood-gas barrier and improved the survival of S. aureus-infected mice. Furthermore, S. aureus-induced myeloperoxidase (MPO) activity of lung homogenate and pro-inflammatory factors in bronchoalveolar lavage (BAL) fluid were suppressed by acetylharpagide. Mechanically, acetylharpagide inhibited the interaction between polyubiquitinated receptor interacting protein 1 (RIP1) and NF-κB essential modulator (NEMO), thereby suppressing NF-κB activity. In summary, these results show that acetylharpagide protects mice from S. aureus-induced ALI by suppressing the NF-κB signaling pathway. Acetylharpagide is expected to become a potential treatment for S. aureus-induced ALI.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/metabolismo , FN-kappa B/metabolismo , Extractos Vegetales/farmacología , Transducción de Señal/efectos de los fármacos , Infecciones Estafilocócicas/complicaciones , Staphylococcus aureus , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/patología , Animales , Biopsia , Barrera Alveolocapilar/efectos de los fármacos , Barrera Alveolocapilar/metabolismo , Barrera Alveolocapilar/patología , Citocinas/metabolismo , Histocitoquímica , Mediadores de Inflamación/metabolismo , Lamiaceae/química , Ratones , Estructura Molecular , Extractos Vegetales/química , Células RAW 264.7
2.
Am J Physiol Lung Cell Mol Physiol ; 316(1): L255-L268, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30382767

RESUMEN

Irrespective of its diverse etiologies, acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) leads to increased permeability of the alveolar-capillary barrier, which in turn promotes edema formation and respiratory failure. We investigated the mechanism of ALI/ARDS lung hyperpermeability triggered by pulmonary exposure of mice to the highly toxic plant-derived toxin ricin. One prominent hallmark of ricin-mediated pulmonary intoxication is the rapid and massive influx of neutrophils to the lungs, where they contribute to the developing inflammation yet may also cause tissue damage, thereby promoting ricin-mediated morbidity. Here we show that pulmonary exposure of mice to ricin results in the rapid diminution of the junction proteins VE-cadherin, claudin 5, and connexin 43, belonging, respectively, to the adherens, tight, and gap junction protein families. Depletion of neutrophils in ricin-intoxicated mice attenuated the damage caused to these junction proteins, alleviated pulmonary edema, and significantly postponed the time to death of the intoxicated mice. Inhibition of matrix metalloproteinase (MMP) activity recapitulated the response to neutrophil depletion observed in ricin-intoxicated mice and was associated with decreased insult to the junction proteins and alveolar-capillary barrier. However, neutrophil-mediated MMP activity was not the sole mechanism responsible for pulmonary hyperpermeability, as exemplified by the ricin-mediated disruption of claudin 18, via a neutrophil-independent mechanism involving tyrosine phosphorylation. This in-depth study of the early stage mechanisms governing pulmonary tissue integrity during ALI/ARDS is expected to facilitate the tailoring of novel therapeutic approaches for the treatment of these diseases.


Asunto(s)
Antígenos CD/metabolismo , Barrera Alveolocapilar/metabolismo , Cadherinas/metabolismo , Claudina-5/metabolismo , Conexina 43/metabolismo , Uniones Intercelulares/metabolismo , Síndrome de Dificultad Respiratoria/metabolismo , Ricina/toxicidad , Animales , Barrera Alveolocapilar/patología , Claudinas/metabolismo , Colagenasas/metabolismo , Modelos Animales de Enfermedad , Humanos , Inflamación , Uniones Intercelulares/patología , Ratones , Síndrome de Dificultad Respiratoria/inducido químicamente , Síndrome de Dificultad Respiratoria/patología
3.
Am J Physiol Lung Cell Mol Physiol ; 316(1): L94-L104, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30358437

RESUMEN

Nicotine is a highly addictive principal component of both tobacco and electronic cigarette that is readily absorbed in blood. Nicotine-containing electronic cigarettes are promoted as a safe alternative to cigarette smoking. However, the isolated effects of inhaled nicotine are largely unknown. Here we report a novel rat model of aerosolized nicotine with a particle size (~1 µm) in the respirable diameter range. Acute nicotine inhalation caused increased pulmonary edema and lung injury as measured by enhanced bronchoalveolar lavage fluid protein, IgM, lung wet-to-dry weight ratio, and high-mobility group box 1 (HMGB1) protein and decreased lung E-cadherin protein. Immunohistochemical analysis revealed congested blood vessels and increased neutrophil infiltration. Lung myeloperoxidase mRNA and protein increased in the nicotine-exposed rats. Complete blood counts also showed an increase in neutrophils, white blood cells, eosinophils, and basophils. Arterial blood gas measurements showed an increase in lactate. Lungs of nicotine-inhaling animals revealed increased mRNA levels of IL-1A and CXCL1. There was also an increase in IL-1α protein. In in vitro air-liquid interface cultures of airway epithelial cells, there was a dose dependent increase in HMGB1 release with nicotine treatment. Air-liquid cultures exposed to nicotine also resulted in a dose-dependent loss of barrier as measured by transepithelial electrical resistance and a decrease in E-cadherin expression. Nicotine also caused a dose-dependent increase in epithelial cell death and an increase in caspase-3/7 activities. These results show that the nicotine content of electronic cigarettes may have adverse pulmonary and systemic effects.


Asunto(s)
Barrera Alveolocapilar , Nicotina/efectos adversos , Vapeo , Aerosoles , Animales , Barrera Alveolocapilar/lesiones , Barrera Alveolocapilar/metabolismo , Barrera Alveolocapilar/patología , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Quimiocina CXCL1/sangre , Proteína HMGB1/metabolismo , Inmunoglobulina M/sangre , Interleucina-1alfa/sangre , Recuento de Leucocitos , Masculino , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/metabolismo , Neutrófilos/patología , Nicotina/farmacología , Tamaño de la Partícula , Edema Pulmonar/sangre , Edema Pulmonar/inducido químicamente , Edema Pulmonar/patología , Ratas , Ratas Sprague-Dawley , Vapeo/efectos adversos , Vapeo/sangre , Vapeo/patología
4.
Am J Physiol Lung Cell Mol Physiol ; 315(1): L66-L77, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29597831

RESUMEN

Compromised pulmonary endothelial cell (PEC) barrier function characterizes acute respiratory distress syndrome (ARDS), a cause of substantial morbidity and mortality. Survival from ARDS is greater in children compared with adults. Whether developmental differences intrinsic to PEC barrier function contribute to this survival advantage remains unknown. To test the hypothesis that PEC barrier function is more well-preserved in neonatal lungs compared with adult lungs in response to inflammation, we induced lung injury in neonatal and adult mice with systemic lipopolysaccharide (LPS). We assessed PEC barrier function in vivo and in vitro, evaluated changes in the expression of focal adhesion kinase 1 (FAK1) and phosphorylation in response to LPS, and determined the effect of FAK silencing and overexpression on PEC barrier function. We found that LPS induced a greater increase in lung permeability and PEC barrier disruption in the adult mice, despite similar degrees of inflammation and apoptosis. Although baseline expression was similar, LPS increased FAK1 expression in neonatal PEC but increased FAK1 phosphorylation and decreased FAK1 expression in adult PEC. Pharmacologic inhibition of FAK1 accentuated LPS-induced barrier disruption most in adult PEC. Finally, in response to LPS, FAK silencing markedly impaired neonatal PEC barrier function, whereas FAK overexpression preserved adult PEC barrier function. Thus, developmental differences in FAK expression during inflammatory injury serve to preserve neonatal pulmonary endothelial barrier function compared with that of adults and suggest that intrinsic differences in the immature versus pulmonary endothelium, especially relative to FAK1 phosphorylation, may contribute to the improved outcomes of children with ARDS.


Asunto(s)
Apoptosis , Barrera Alveolocapilar/enzimología , Células Endoteliales/enzimología , Endotelio/enzimología , Quinasa 1 de Adhesión Focal/metabolismo , Transducción de Señal , Animales , Barrera Alveolocapilar/crecimiento & desarrollo , Barrera Alveolocapilar/patología , Células Endoteliales/patología , Endotelio/patología , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Inflamación/inducido químicamente , Inflamación/enzimología , Inflamación/patología , Lipopolisacáridos/toxicidad , Ratones
5.
Am J Physiol Lung Cell Mol Physiol ; 314(5): L808-L821, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29368549

RESUMEN

Acid (HCl) aspiration during anesthesia may lead to acute lung injury. There is no effective therapy. We hypothesized that HCl instilled intratracheally in C57BL/6 mice results in the formation of low-molecular weight hyaluronan (L-HA), which activates RhoA and Rho kinase (ROCK), causing airway hyperresponsiveness (AHR) and increased permeability. Furthermore, instillation of high-molecular weight hyaluronan (H-HA; Yabro) will reverse lung injury. We instilled HCl in C57BL/6 wild-type (WT), myeloperoxidase gene-deficient (MPO-/-) mice, and CD44 gene-deficient (CD44-/-) mice. WT mice were also instilled intranasally with H-HA (Yabro) at 1 and 23 h post-HCl. All measurements were performed at 1, 5, or 24 h post-HCl. Instillation of HCl in WT but not in CD44-/- resulted in increased inflammation, AHR, lung injury, and L-HA in the bronchoalveolar lavage fluid (BALF) 24 h post-HCl; L-HA levels and lung injury were significantly lower in HCl-instilled MPO-/- mice. Isolated perfused lungs of HCl instilled WT but not of CD44-/- mice had elevated values of the filtration coefficient ( Kf). Addition of L-HA on the apical surface of human primary bronchial epithelial cell monolayer decreased barrier resistance ( RT). H-HA significantly mitigated inflammation, AHR, and pulmonary vascular leakage at 24 h after HCl instillation and mitigated the increase of Kf and RT, as well as ROCK2 phosphorylation. Increased H- and L-HA levels were found in the BALF of mechanically ventilated patients but not in healthy volunteers. HCl instillation-induced lung injury is mediated by the L-HA-CD44-RhoA-ROCK2 signaling pathway, and H-HA is a potential novel therapeutic agent for acid aspiration-induced lung injury.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Barrera Alveolocapilar/efectos de los fármacos , Receptores de Hialuranos/fisiología , Ácido Hialurónico/farmacología , Ácido Clorhídrico/toxicidad , Peroxidasa/fisiología , Neumonía/tratamiento farmacológico , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Animales , Barrera Alveolocapilar/metabolismo , Barrera Alveolocapilar/patología , Líquido del Lavado Bronquioalveolar/química , Células Cultivadas , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía/inducido químicamente , Neumonía/metabolismo , Neumonía/patología , Intercambio Gaseoso Pulmonar , Viscosuplementos/farmacología
6.
J Anat ; 232(2): 283-295, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29193065

RESUMEN

More frequent utilization of non-heart-beating donor (NHBD) organs for lung transplantation has the potential to relieve the shortage of donor organs. In particular with respect to uncontrolled NHBD, concerns exist regarding the risk of ischaemia/reperfusion (IR) injury-related graft damage or dysfunction. Due to their immunomodulating and tissue-remodelling properties, bone-marrow-derived mesenchymal stem cells (MSCs) have been suspected of playing a beneficial role regarding short- and long-term survival and function of the allograft. Thus, MSC administration might represent a promising pretreatment strategy for NHBD organs. To study the initial effects of warm ischaemia and MSC application, a large animal lung transplantation model was generated, and the structural organ composition of the transplanted lungs was analysed stereologically with particular respect to the blood-gas barrier and the surfactant system. In this study, porcine lungs (n = 5/group) were analysed. Group 1 was the sham-operated control group. In pigs of groups 2-4, cardiac arrest was induced, followed by a period of 3 h of ventilated ischaemia at room temperature. In groups 3 and 4, 50 × 106 MSCs were administered intravascularly via the pulmonary artery and endobronchially, respectively, during the last 10 min of ischaemia. The left lungs were transplanted, followed by a reperfusion period of 4 h. Then, lungs were perfusion-fixed and processed for light and electron microscopy. Samples were analysed stereologically for IR injury-related structural parameters, including volume densities and absolute volumes of parenchyma components, alveolar septum components, intra-alveolar oedema, and the intracellular and intra-alveolar surfactant pool. Additionally, the volume-weighted mean volume of lamellar bodies (lbs) and their profile size distribution were determined. Three hours of ventilated warm ischaemia was tolerated without eliciting histological or ultrastructural signs of IR injury, as revealed by qualitative and quantitative assessment. However, warm ischaemia influenced the surfactant system. The volume-weighted mean volume of lbs was reduced significantly (P = 0.024) in groups subjected to ischaemia (group medians of groups 2-4: 0.180-0.373 µm³) compared with the sham control group (median 0.814 µm³). This was due to a lower number of large lb profiles (size classes 5-15). In contrast, the intra-alveolar surfactant system was not altered significantly. No significant differences were encountered comparing ischaemia alone (group 2) or ischaemia plus application of MSCs (groups 3 and 4) in this short-term model.


Asunto(s)
Barrera Alveolocapilar/patología , Trasplante de Pulmón/métodos , Pulmón/patología , Trasplante de Células Madre Mesenquimatosas/métodos , Surfactantes Pulmonares , Animales , Modelos Animales de Enfermedad , Paro Cardíaco , Daño por Reperfusión/patología , Porcinos , Isquemia Tibia
7.
Am J Physiol Lung Cell Mol Physiol ; 312(1): L1-L12, 2017 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-27815259

RESUMEN

Acute lung injury (ALI) is characterized by hypoxemia, enhanced permeability of the air-blood barrier, and pulmonary edema. Particularly in the elderly, ALI is associated with increased morbidity and mortality. The reasons for this, however, are poorly understood. We hypothesized that age-related changes in pulmonary structure, function, and inflammation lead to a worse prognosis in ALI. ALI was induced in young (10 wk old) and old (18 mo old) male C57BL/6 mice by intranasal application of 2.5 mg lipopolysaccharide (LPS)/kg body wt or saline (control mice). After 24 h, lung function was assessed, and lungs were either processed for stereological or inflammatory analysis, such as bronchoalveolar lavage fluid (BALF) cytometry and qPCR. Both young and old mice developed severe signs of ALI, including alveolar and septal edema and enhanced inflammatory BALF cells. However, the pathology of ALI was more pronounced in old compared with young mice with nearly sixfold higher BALF protein concentration, twice the number of neutrophils, and significantly higher expression of neutrophil chemokine Cxcl1, adhesion molecule Icam-1, and metalloprotease-9, whereas the expression of tight junction protein occludin significantly decreased. The old LPS mice had thicker alveolar septa attributable to higher volumes of interstitial cells and extracellular matrix. Tissue resistance and elastance reflected observed changes at the ultrastructural level in the lung parenchyma in ALI of young and old mice. In summary, the pathology of ALI with advanced age in mice is characterized by a greater neutrophilic inflammation, leakier air-blood barrier, and altered lung function, which is in line with findings in elderly patients.


Asunto(s)
Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/fisiopatología , Envejecimiento/patología , Barrera Alveolocapilar/patología , Pulmón/patología , Pulmón/fisiopatología , Neumonía/patología , Neumonía/fisiopatología , Lesión Pulmonar Aguda/complicaciones , Lesión Pulmonar Aguda/genética , Animales , Barrera Alveolocapilar/ultraestructura , Peso Corporal , Líquido del Lavado Bronquioalveolar/citología , Recuento de Células , Colágeno/metabolismo , Progresión de la Enfermedad , Elastina/metabolismo , Matriz Extracelular/metabolismo , Citometría de Flujo , Regulación de la Expresión Génica , Lipopolisacáridos , Masculino , Ratones Endogámicos C57BL , Neumonía/complicaciones , Neumonía/genética , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Pruebas de Función Respiratoria
8.
Sud Med Ekspert ; 60(4): 9-11, 2017.
Artículo en Ruso | MEDLINE | ID: mdl-28766520

RESUMEN

The acute respiratory viral infections (ARVI) are ranked among the most widespread diseases affecting the children in the early infancy. They account for 60 to 85.4% of all infections recorded in the young children. AIM: The objective of the present study was to elucidate the peculiar features of the accumulation of the effector cells of the local immunity system and intercellular interplay in the broncho-vascular barrier of the breast-fed infants presenting with various ARVIs. MATERIAL AND METHODS: We undertook the analysis of 32 cases of infections caused by influenza A and B viruses and of the adenovirus infection verified by the immunofluorescence assay. The group of comparison was comprised of 10 children presenting with congenial heart disease in the absence of the signs of inflammatory processes in the lungs. The tissue samples were harvested from the upper lobe of the lung at the level of the lobe bronchus and a terminal bronchiola. The materials for the histological study were prepared using the Romanovsky method. The effector cells were examined in the lamina propria of bronchial mucosa and the submucous layer. The morphometric analysis included direct counting the number of lymphocytes, mast cells, macrophages, plasmocytes, eosinophils, and neutrophils with the subsequent recalculation of the data thus obtained per unit volume of the connective tissue. The results of the morphometric analysis were subjected to the statistical treatment. RESULTS: The study has demonstrated that the young children suffering from a viral infection, regardless of the type of the causative factor, experience a change in the total number and the ratio of the effector cells at all the levels of the broncho-vascular barrier.


Asunto(s)
Barrera Alveolocapilar , Cardiopatías Congénitas , Sistema Inmunológico/patología , Infecciones del Sistema Respiratorio , Virosis , Enfermedad Aguda , Barrera Alveolocapilar/inmunología , Barrera Alveolocapilar/patología , Comunicación Celular/inmunología , Femenino , Técnica del Anticuerpo Fluorescente/métodos , Medicina Legal/métodos , Cardiopatías Congénitas/inmunología , Cardiopatías Congénitas/patología , Humanos , Lactante , Masculino , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/patología , Infecciones del Sistema Respiratorio/virología , Virosis/inmunología , Virosis/patología
9.
Respir Res ; 17(1): 71, 2016 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-27301375

RESUMEN

BACKGROUND: Gastric contents aspiration in humans is a risk factor for severe respiratory failure with elevated mortality. Although aspiration-induced local lung inflammation has been studied in animal models, little is known about extrapulmonary effects of aspiration. We investigated whether a single orotracheal instillation of whole gastric fluid elicits a liver acute phase response and if this response contributes to enrich the alveolar spaces with proteins having antiprotease activity. METHODS: In anesthetized Sprague-Dawley rats receiving whole gastric fluid, we studied at different times after instillation (4 h -7 days): changes in blood cytokines and acute phase proteins (fibrinogen and the antiproteases alpha1-antitrypsin and alpha2-macroglobulin) as well as liver mRNA expression of the two antiproteases. The impact of the systemic changes on lung antiprotease defense was evaluated by measuring levels and bioactivity of antiproteases in broncho-alveolar lavage fluid (BALF). Markers of alveolar-capillary barrier derangement were also studied. Non-parametric ANOVA (Kruskall-Wallis) and linear regression analysis were used. RESULTS: Severe peribronchiolar injury involving edema, intra-alveolar proteinaceous debris, hemorrhage and PMNn cell infiltration was seen in the first 24 h and later resolved. Despite a large increase in several lung cytokines, only IL-6 was found elevated in blood, preceding increased liver expression and blood concentration of both antiproteases. These changes, with an acute phase response profile, were significantly larger for alpha2-macroglobulin (40-fold increment in expression with 12-fold elevation in blood protein concentration) than for alpha1-antitrypsin (2-3 fold increment in expression with 0.5-fold elevation in blood protein concentration). Both the increment in capillary-alveolar antiprotease concentration gradient due to increased antiprotease liver synthesis and a timely-associated derangement of the alveolar-capillary barrier induced by aspiration, contributed a 58-fold and a 190-fold increase in BALF alpha1-antitrypsin and alpha2-macroglobulin levels respectively (p < 0.001). CONCLUSIONS: Gastric contents-induced acute lung injury elicits a liver acute phase response characterized by increased mRNA expression of antiproteases and elevation of blood antiprotease concentrations. Hepatic changes act in concert with derangement of the alveolar capillary barrier to enrich alveolar spaces with antiproteases. These findings may have significant implications decreasing protease burden, limiting injury in this and other models of acute lung injury and likely, in recurrent aspiration.


Asunto(s)
Lesión Pulmonar Aguda/enzimología , Reacción de Fase Aguda/enzimología , Hígado/metabolismo , alfa 2-Macroglobulinas Asociadas al Embarazo/biosíntesis , Alveolos Pulmonares/enzimología , Aspiración Respiratoria de Contenidos Gástricos/complicaciones , alfa 1-Antitripsina/biosíntesis , Lesión Pulmonar Aguda/sangre , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/patología , Reacción de Fase Aguda/sangre , Reacción de Fase Aguda/etiología , Reacción de Fase Aguda/patología , Animales , Barrera Alveolocapilar/enzimología , Barrera Alveolocapilar/patología , Modelos Animales de Enfermedad , Inducción Enzimática , Mediadores de Inflamación/sangre , Interleucina-6/sangre , Masculino , alfa 2-Macroglobulinas Asociadas al Embarazo/genética , Alveolos Pulmonares/patología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas Sprague-Dawley , Factores de Tiempo , alfa 1-Antitripsina/sangre , alfa 1-Antitripsina/genética
10.
J Immunol ; 193(5): 2469-82, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25063875

RESUMEN

A disintegrin and a metalloproteinase domain (ADAM) 9 is known to be expressed by monocytes and macrophages. In this study, we report that ADAM9 is also a product of human and murine polymorphonuclear neutrophils (PMNs). ADAM9 is not synthesized de novo by circulating PMNs. Rather, ADAM9 protein is stored in the gelatinase and specific granules and the secretory vesicles of human PMNs. Unstimulated PMNs express minimal quantities of surface ADAM9, but activation of PMNs with degranulating agonists rapidly (within 15 min) increases PMN surface ADAM9 levels. Human PMNs produce small quantities of soluble forms of ADAM9. Surprisingly, ADAM9 degrades several extracellular matrix (ECM) proteins, including fibronectin, entactin, laminin, and insoluble elastin, as potently as matrix metalloproteinase-9. However, ADAM9 does not degrade types I, III, or IV collagen or denatured collagens in vitro. To determine whether Adam9 regulates PMN recruitment or ECM protein turnover during inflammatory responses, we compared wild-type and Adam9(-/-) mice in bacterial LPS- and bleomycin-mediated acute lung injury (ALI). Adam9 lung levels increase 10-fold during LPS-mediated ALI in wild-type mice (due to increases in leukocyte-derived Adam9), but Adam9 does not regulate lung PMN (or macrophage) counts during ALI. Adam9 increases mortality, promotes lung injury, reduces lung compliance, and increases degradation of lung elastin during LPS- and/or bleomycin-mediated ALI. Adam9 does not regulate collagen accumulation in the bleomycin-treated lung. Thus, ADAM9 is expressed in an inducible fashion on PMN surfaces where it degrades some ECM proteins, and it promotes alveolar-capillary barrier injury during ALI in mice.


Asunto(s)
Proteínas ADAM/inmunología , Lesión Pulmonar Aguda/inmunología , Matriz Extracelular/inmunología , Proteínas de la Membrana/inmunología , Neutrófilos/inmunología , Proteolisis , Proteínas ADAM/genética , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Animales , Antibióticos Antineoplásicos/efectos adversos , Antibióticos Antineoplásicos/farmacología , Bleomicina/efectos adversos , Bleomicina/farmacología , Barrera Alveolocapilar/inmunología , Barrera Alveolocapilar/patología , Colágeno/genética , Colágeno/inmunología , Elastina/genética , Elastina/inmunología , Matriz Extracelular/genética , Humanos , Lipopolisacáridos/toxicidad , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Neutrófilos/patología
11.
Part Fibre Toxicol ; 13: 20, 2016 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-27108236

RESUMEN

Aerosol generation and characterization are critical components in the assessment of the inhalation hazards of engineered nanomaterials (NMs). An extensive review was conducted on aerosol generation and exposure apparatus as part of an international expert workshop convened to discuss the design of an in vitro testing strategy to assess pulmonary toxicity following exposure to aerosolized particles. More specifically, this workshop focused on the design of an in vitro method to predict the development of pulmonary fibrosis in humans following exposure to multi-walled carbon nanotubes (MWCNTs). Aerosol generators, for dry or liquid particle suspension aerosolization, and exposure chambers, including both commercially available systems and those developed by independent researchers, were evaluated. Additionally, characterization methods that can be used and the time points at which characterization can be conducted in order to interpret in vitro exposure results were assessed. Summarized below is the information presented and discussed regarding the relevance of various aerosol generation and characterization techniques specific to aerosolized MWCNTs exposed to cells cultured at the air-liquid interface (ALI). The generation of MWCNT aerosols relevant to human exposures and their characterization throughout exposure in an ALI system is critical for extrapolation of in vitro results to toxicological outcomes in humans.


Asunto(s)
Lesión Pulmonar Aguda/inducido químicamente , Barrera Alveolocapilar/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Nanotubos de Carbono/toxicidad , Pruebas de Toxicidad/métodos , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Aerosoles , Alternativas a las Pruebas en Animales , Animales , Barrera Alveolocapilar/metabolismo , Barrera Alveolocapilar/patología , Técnicas de Cultivo de Célula , Células Cultivadas , Congresos como Asunto , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Exposición por Inhalación , Nanotubos de Carbono/química , Tamaño de la Partícula , Medición de Riesgo
12.
J Biol Chem ; 289(48): 33355-63, 2014 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-25315770

RESUMEN

Pulmonary damages of oxygen toxicity include vascular leakage and pulmonary edema. We have previously reported that hyperoxia increases the formation of NO and peroxynitrite in lung endothelial cells via increased interaction of endothelial nitric oxide (eNOS) with ß-actin. A peptide (P326TAT) with amino acid sequence corresponding to the actin binding region of eNOS residues 326-333 has been shown to reduce the hyperoxia-induced formation of NO and peroxynitrite in lung endothelial cells. In the present study, we found that exposure of pulmonary artery endothelial cells to hyperoxia (95% oxygen and 5% CO2) for 48 h resulted in disruption of monolayer barrier integrity in two phases, and apoptosis occurred in the second phase. NOS inhibitor N(G)-nitro-L-arginine methyl ester attenuated the endothelial barrier disruption in both phases. Peroxynitrite scavenger uric acid did not affect the first phase but ameliorated the second phase of endothelial barrier disruption and apoptosis. P326TAT inhibited hyperoxia-induced disruption of monolayer barrier integrity in two phases and apoptosis in the second phase. More importantly, injection of P326TAT attenuated vascular leakage, pulmonary edema, and endothelial apoptosis in the lungs of mice exposed to hyperoxia. P326TAT also significantly reduced the increase in eNOS-ß-actin association and protein tyrosine nitration. Together, these results indicate that peptide P326TAT ameliorates barrier dysfunction of hyperoxic lung endothelial monolayer and attenuates eNOS-ß-actin association, peroxynitrite formation, endothelial apoptosis, and pulmonary edema in lungs of hyperoxic mice. P326TAT can be a novel therapeutic agent to treat or prevent acute lung injury in oxygen toxicity.


Asunto(s)
Apoptosis/efectos de los fármacos , Barrera Alveolocapilar/metabolismo , Endotelio Vascular/metabolismo , Péptidos/farmacología , Ácido Peroxinitroso/metabolismo , Edema Pulmonar , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/prevención & control , Animales , Barrera Alveolocapilar/patología , Bovinos , Células Cultivadas , Endotelio Vascular/patología , Hiperoxia/tratamiento farmacológico , Hiperoxia/metabolismo , Hiperoxia/patología , Ratones , Óxido Nítrico Sintasa de Tipo III/metabolismo , Edema Pulmonar/tratamiento farmacológico , Edema Pulmonar/metabolismo , Edema Pulmonar/patología
13.
Am J Physiol Lung Cell Mol Physiol ; 309(9): L983-94, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26361873

RESUMEN

Inflammatory mediators released in acute lung injury (ALI) trigger the disruption of interendothelial junctions, leading to loss of vascular barrier function, protein-rich pulmonary edema, and severe hypoxemia. Genetic signatures that predict patient recovery or disease progression are poorly defined, but recent genetic screening of ALI patients has identified an association between lung inflammatory disease and a single nucleotide polymorphism (SNP) in the gene for the actin-binding and barrier-regulatory protein cortactin. This study investigated the impact of this disease-linked cortactin variant on wound healing processes that may contribute to endothelial barrier restoration. A microfabricated platform was used to quantify wound healing in terms of gap closure speed, lamellipodia dynamics, and cell velocity. Overexpression of wild-type cortactin in endothelial cells (ECs) improved directional cell motility and enhanced lamellipodial protrusion length, resulting in enhanced gap closure rates. By contrast, the cortactin SNP impaired wound closure and cell locomotion, consistent with the observed reduction in lamellipodial protrusion length and persistence. Overexpression of the cortactin SNP in lung ECs mitigated the barrier-enhancing activity of sphingosine 1-phosphate. These findings suggest that this common cortactin variant may functionally contribute to ALI predisposition by impeding endothelial wound healing.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Cortactina/metabolismo , Células Endoteliales/metabolismo , Polimorfismo de Nucleótido Simple , Seudópodos/metabolismo , Cicatrización de Heridas , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Animales , Barrera Alveolocapilar/metabolismo , Barrera Alveolocapilar/patología , Bovinos , Células Cultivadas , Cortactina/genética , Células Endoteliales/patología , Humanos , Lisofosfolípidos/metabolismo , Seudópodos/genética , Esfingosina/análogos & derivados , Esfingosina/metabolismo
14.
PLoS Pathog ; 9(11): e1003727, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24244159

RESUMEN

Streptococcus pneumoniae infection is a leading cause of bacterial pneumonia, sepsis and meningitis and is associated with high morbidity and mortality. Type I interferon (IFN-I), whose contribution to antiviral and intracellular bacterial immunity is well established, is also elicited during pneumococcal infection, yet its functional significance is not well defined. Here, we show that IFN-I plays an important role in the host defense against pneumococci by counteracting the transmigration of bacteria from the lung to the blood. Mice that lack the type I interferon receptor (Ifnar1 (-/-)) or mice that were treated with a neutralizing antibody against the type I interferon receptor, exhibited enhanced development of bacteremia following intranasal pneumococcal infection, while maintaining comparable bacterial numbers in the lung. In turn, treatment of mice with IFNß or IFN-I-inducing synthetic double stranded RNA (poly(I:C)), dramatically reduced the development of bacteremia following intranasal infection with S. pneumoniae. IFNß treatment led to upregulation of tight junction proteins and downregulation of the pneumococcal uptake receptor, platelet activating factor receptor (PAF receptor). In accordance with these findings, IFN-I reduced pneumococcal cell invasion and transmigration across epithelial and endothelial layers, and Ifnar1 (-/-) mice showed overall enhanced lung permeability. As such, our data identify IFN-I as an important component of the host immune defense that regulates two possible mechanisms involved in pneumococcal invasion, i.e. PAF receptor-mediated transcytosis and tight junction-dependent pericellular migration, ultimately limiting progression from a site-restricted lung infection to invasive, lethal disease.


Asunto(s)
Barrera Alveolocapilar/metabolismo , Barrera Hematoencefálica/metabolismo , Interferón Tipo I/metabolismo , Neumonía Neumocócica/metabolismo , Streptococcus pneumoniae/patogenicidad , Animales , Bacteriemia/genética , Bacteriemia/metabolismo , Barrera Alveolocapilar/microbiología , Barrera Alveolocapilar/patología , Barrera Hematoencefálica/microbiología , Barrera Hematoencefálica/patología , Interferón Tipo I/genética , Ratones , Ratones Noqueados , Neumonía Neumocócica/genética , Neumonía Neumocócica/patología , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Streptococcus pneumoniae/metabolismo
15.
Respir Res ; 16: 30, 2015 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-25851441

RESUMEN

BACKGROUND: The cadmium (Cd) present in air pollutants and cigarette smoke has the potential of causing multiple adverse health outcomes involving damage to pulmonary and cardiovascular tissue. Injury to pulmonary epithelium may include alterations in tight junction (TJ) integrity, resulting in impaired epithelial barrier function and enhanced penetration of chemicals and biomolecules. Herein, we investigated mechanisms involved in the disruption of TJ integrity by Cd exposure using an in vitro human air-liquid-interface (ALI) airway tissue model derived from normal primary human bronchial epithelial cells. METHODS: ALI cultures were exposed to noncytotoxic doses of CdCl2 basolaterally and TJ integrity was measured by Trans-Epithelial Electrical Resistance (TEER) and immunofluorescence staining with TJ markers. PCR array analysis was used to identify genes involved with TJ collapse. To explore the involvement of kinase signaling pathways, cultures were treated with CdCl2 in the presence of kinase inhibitors specific for cellular Src or Protein Kinase C (PKC). RESULTS: Noncytotoxic doses of CdCl2 resulted in the collapse of barrier function, as demonstrated by TEER measurements and Zonula occludens-1 (ZO-1) and occludin staining. CdCl2 exposure altered the expression of several groups of genes encoding proteins involved in TJ homeostasis. In particular, down-regulation of select junction-interacting proteins suggested that a possible mechanism for Cd toxicity involves disruption of the peripheral junctional complexes implicated in connecting membrane-bound TJ components to the actin cytoskeleton. Inhibition of kinase signaling using inhibitors specific for cellular Src or PKC preserved the integrity of TJs, possibly by preventing occludin tyrosine hyperphosphorylation, rather than reversing the down-regulation of the junction-interacting proteins. CONCLUSIONS: Our findings indicate that acute doses of Cd likely disrupt TJ integrity in human ALI airway cultures both through occludin hyperphosphorylation via kinase activation and by direct disruption of the junction-interacting complex.


Asunto(s)
Barrera Alveolocapilar/efectos de los fármacos , Bronquios/efectos de los fármacos , Cloruro de Cadmio/toxicidad , Células Epiteliales/efectos de los fármacos , Uniones Estrechas/efectos de los fármacos , Barrera Alveolocapilar/metabolismo , Barrera Alveolocapilar/patología , Bronquios/metabolismo , Bronquios/patología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Impedancia Eléctrica , Células Epiteliales/metabolismo , Células Epiteliales/patología , Regulación de la Expresión Génica , Humanos , Ocludina/genética , Ocludina/metabolismo , Fosforilación , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Uniones Estrechas/metabolismo , Uniones Estrechas/patología , Factores de Tiempo , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
16.
J Biol Chem ; 288(52): 37343-54, 2013 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-24265314

RESUMEN

At the interface between host and external environment, the airway epithelium serves as a major protective barrier. In the present study we show that protein kinase D (PKD) plays an important role in the formation and integrity of the airway epithelial barrier. Either inhibition of PKD activity or silencing of PKD increased transepithelial electrical resistance (TEER), resulting in a tighter epithelial barrier. Among the three PKD isoforms, PKD3 knockdown was the most efficient one to increase TEER in polarized airway epithelial monolayers. In contrast, overexpression of PKD3 wild type, but not PKD3 kinase-inactive mutant, disrupted the formation of apical intercellular junctions and their reassembly, impaired the development of TEER, and increased paracellular permeability to sodium fluorescein in airway epithelial monolayers. We further found that overexpression of PKD, in particular PKD3, markedly suppressed the mRNA and protein levels of claudin-1 but had only minor effects on the expression of other tight junctional proteins (claudin-3, claudin-4, claudin-5, occludin, and ZO-1) and adherent junctional proteins (E-cadherin and ß-catenin). Immunofluorescence study revealed that claudin-1 level was markedly reduced and almost disappeared from intercellular contacts in PKD3-overexpressed epithelial monolayers and that claudin-4 was also restricted from intercellular contacts and tended to accumulate in the cell cytosolic compartments. Last, we found that claudin-1 knockdown prevented TEER elevation by PKD inhibition or silencing in airway epithelial monolayers. These novel findings indicate that PKD negatively regulates human airway epithelial barrier formation and integrity through down-regulation of claudin-1, which is a key component of tight junctions.


Asunto(s)
Barrera Alveolocapilar/metabolismo , Claudina-1/biosíntesis , Regulación hacia Abajo , Proteína Quinasa C/metabolismo , Mucosa Respiratoria/metabolismo , Uniones Estrechas/metabolismo , Barrera Alveolocapilar/patología , Células Cultivadas , Claudina-1/genética , Impedancia Eléctrica , Células Epiteliales/metabolismo , Células Epiteliales/patología , Técnicas de Silenciamiento del Gen , Humanos , Permeabilidad , Proteína Quinasa C/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Mucosa Respiratoria/patología , Uniones Estrechas/genética , Uniones Estrechas/patología
17.
Am J Physiol Lung Cell Mol Physiol ; 307(12): L924-35, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25381026

RESUMEN

Endothelial barrier function is an essential and tightly regulated process that ensures proper compartmentalization of the vascular and interstitial space, while allowing for the diffusive exchange of small molecules and the controlled trafficking of macromolecules and immune cells. Failure to control endothelial barrier integrity results in excessive leakage of fluid and proteins from the vasculature that can rapidly become fatal in scenarios such as sepsis or the acute respiratory distress syndrome. Here, we highlight recent advances in our understanding on the regulation of endothelial permeability, with a specific focus on the endothelial glycocalyx and endothelial scaffolds, regulatory intracellular signaling cascades, as well as triggers and mediators that either disrupt or enhance endothelial barrier integrity, and provide our perspective as to areas of seeming controversy and knowledge gaps, respectively.


Asunto(s)
Barrera Alveolocapilar/metabolismo , Permeabilidad Capilar , Endotelio Vascular/metabolismo , Glicocálix/metabolismo , Síndrome de Dificultad Respiratoria/metabolismo , Animales , Barrera Alveolocapilar/patología , Endotelio Vascular/patología , Glicocálix/patología , Síndrome de Dificultad Respiratoria/patología , Sepsis/metabolismo , Sepsis/patología
18.
Am J Physiol Lung Cell Mol Physiol ; 307(11): L817-21, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25281637

RESUMEN

Transient receptor potential (TRP) channels are emerging as important players and drug targets in respiratory disease. Amongst the vanilloid-type TRP channels (which includes the six members of the TRPV family), target diseases include cough, asthma, cancer, and more recently, pulmonary edema associated with acute respiratory distress syndrome. Here, we critically evaluate a recent report that addresses TRPV4 as a candidate target for the management of acute lung injury that develops as a consequence of aspiration of gastric contents, or acute chlorine gas exposure. By use of two new TRPV4 inhibitors (GSK2220691 or GSK2337429A) and a trpv4(-/-) mouse strain, TRPV4 was implicated as a key mediator of pulmonary inflammation after direct chemical insult. Additionally, applied therapeutically, TRPV4 inhibitors exhibited vasculoprotective effects after chlorine gas exposure, inhibiting vascular leakage, and improving blood oxygenation. These observations underscore TRPV4 channels as candidate therapeutic targets in the management of lung injury, with the added need to balance these against the potential drawbacks of TRPV4 inhibition, such as the danger of limiting the immune response in settings of pathogen-provoked injury.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Barrera Alveolocapilar/patología , Neumonía por Aspiración/tratamiento farmacológico , Canales Catiónicos TRPV/antagonistas & inhibidores , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/etiología , Animales , Ácidos Araquidónicos/farmacología , Calcio/metabolismo , Cannabinoides/farmacología , Cloro/toxicidad , Endocannabinoides/farmacología , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/trasplante , Ratones , Ratones Noqueados , Músculo Liso Vascular/metabolismo , Neumonía por Aspiración/etiología , Neumonía por Aspiración/metabolismo , Alcamidas Poliinsaturadas/farmacología , Arteria Pulmonar/metabolismo , Aspiración Respiratoria de Contenidos Gástricos/complicaciones , Canales Catiónicos TRPV/genética
19.
Eur J Immunol ; 43(12): 3125-37, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24165907

RESUMEN

Chronic asthma is an inflammatory disease of the airway wall that leads to bronchial smooth muscle hyperreactivity and airway obstruction, caused by inflammation, goblet cell metaplasia, and airway wall remodeling. In response to allergen presentation by airway DCs, T-helper lymphocytes of the adaptive immune system control many aspects of the disease through secretion of IL-4, IL-5, IL-13, IL-17, and IL-22, and these are counterbalanced by cytokines produced by Treg cells. Many cells of the innate immune system such as mast cells, basophils, neutrophils, eosinophils, and innate lymphoid cells also play an important role in disease pathogenesis. Barrier epithelial cells are being ever more implicated in disease pathogenesis than previously thought, as these cells have in recent years been shown to sense exposure to allergens via pattern recognition receptors and to activate conventional and inflammatory-type DCs and other innate immune cells through the secretion of thymic stromal lymphopoietin, granulocyte-macrophage colony stimulating factor, IL-1, IL-33, and IL-25. Understanding this cytokine crosstalk between barrier epithelial cells, DCs, and immune cells provides important insights into the mechanisms of allergic sensitization and asthma progression as discussed in this review.


Asunto(s)
Alérgenos/inmunología , Presentación de Antígeno , Asma/inmunología , Barrera Alveolocapilar/inmunología , Células Dendríticas/inmunología , Células Caliciformes/inmunología , Animales , Asma/patología , Barrera Alveolocapilar/patología , Citocinas/inmunología , Células Dendríticas/patología , Células Epiteliales/inmunología , Células Epiteliales/patología , Células Caliciformes/patología , Humanos , Inflamación/inmunología , Inflamación/patología , Mastocitos/inmunología , Mastocitos/patología , Metaplasia , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología
20.
Am J Respir Cell Mol Biol ; 48(4): 477-88, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23306835

RESUMEN

Earlier studies indicated a role for reactive oxygen species (ROS) in host defense against Pseudomonas aeruginosa infection. However, the role of nicotinamide adenine dinucleotide phosphate-reduced (NADPH) oxidase (NOX) proteins and the mechanism of activation for NADPH oxidase in P. aeruginosa infection are not well-defined. Here, we investigated the role of NOX2 and NOX4 proteins in P. aeruginosa infection, ROS generation, and endothelial barrier function in murine lungs and in human lung microvascular endothelial cells (HLMVECs). Airway instillation of P. aeruginosa strain 103 (PA103) significantly increased ROS concentrations in bronchial alveolar lavage (BAL) fluid, along with the expression of NOX2 and NOX4, but not NOX1 and NOX3, in lung tissue. In addition, PA103-infected HLMVECs revealed elevated concentrations of ROS, NOX2, and NOX4. In murine lungs and HLMVECs, PA103 induced the NF-κB pathway, and its inhibition blocked PA103-dependent NOX2 and NOX4 expression. Barrier function analysis showed that heat-killed PA103 induced endothelial permeability in a dose-dependent manner, which was attenuated by treatment with small interfering (si)RNA specific for NOX4, but not NOX2. Furthermore, the knockdown of NOX4, but not NOX2, with siRNA reduced PA103-mediated apoptosis in HLMVECs. In vivo, the down-regulation of NOX4 with NOX4 siRNA attenuated PA103-induced lung vascular permeability. The deletion of NOX2 in mice exerted no effect on permeability, but offered significant resistance to P. aeruginosa-induced lung inflammation. These data show that P. aeruginosa lung infection up-regulates NOX2 and NOX4 expression and ROS generation, which play distinct roles in regulating lung inflammation, apoptosis, and permeability.


Asunto(s)
Permeabilidad Capilar , Glicoproteínas de Membrana/biosíntesis , NADPH Oxidasas/biosíntesis , NADP/metabolismo , Neumonía Bacteriana/enzimología , Infecciones por Pseudomonas/enzimología , Pseudomonas aeruginosa , Animales , Barrera Alveolocapilar/enzimología , Barrera Alveolocapilar/patología , Células Cultivadas , Regulación Enzimológica de la Expresión Génica/genética , Humanos , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , NADP/genética , NADPH Oxidasa 2 , NADPH Oxidasa 4 , NADPH Oxidasas/genética , Neumonía Bacteriana/genética , Neumonía Bacteriana/patología , Infecciones por Pseudomonas/genética , Infecciones por Pseudomonas/patología , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA