Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Antioxid Redox Signal ; 22(8): 651-68, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25401197

RESUMEN

AIMS: Ventilator-induced lung injury (VILI) contributes to mortality in patients with acute respiratory distress syndrome, the most severe form of acute lung injury (ALI). Absence of activating transcription factor 3 (ATF3) confers susceptibility to ALI/VILI. To identify cell-specific ATF3-dependent mechanisms of susceptibility to ALI/VILI, we generated ATF3 chimera by adoptive bone marrow (BM) transfer and randomized to inhaled saline or lipopolysacharide (LPS) in the presence of mechanical ventilation (MV). Adenovirus vectors to silence or overexpress ATF3 were used in primary human bronchial epithelial cells and murine BM-derived macrophages from wild-type or ATF3-deficient mice. RESULTS: Absence of ATF3 in myeloid-derived cells caused increased pulmonary cellular infiltration. In contrast, absence of ATF3 in parenchymal cells resulted in loss of alveolar-capillary membrane integrity and increased exudative edema. ATF3-deficient macrophages were unable to limit the expression of pro-inflammatory mediators. Knockdown of ATF3 in resident cells resulted in decreased junctional protein expression and increased paracellular leak. ATF3 overexpression abrogated LPS induced membrane permeability. Despite release of ATF3-dependent Nrf2 transcriptional inhibition, mice that lacked ATF3 expression in resident cells had increased Nrf2 protein degradation. INNOVATION: In our model, in the absence of ATF3 in parenchymal cells increased Nrf2 degradation is the result of increased Keap-1 expression and loss of DJ-1 (Parkinson disease [autosomal recessive, early onset] 7), previously not known to play a role in lung injury. CONCLUSION: Results suggest that ATF3 confers protection to lung injury by preventing inflammatory cell recruitment and barrier disruption in a cell-specific manner, opening novel opportunities for cell specific therapy for ALI/VILI.


Asunto(s)
Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Pulmón/citología , Factor 2 Relacionado con NF-E2/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo , Animales , Línea Celular , Permeabilidad de la Membrana Celular , Quimera , Células Epiteliales , Femenino , Humanos , Inflamación/metabolismo , Pulmón/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Estrés Oxidativo , Transducción de Señal
2.
BMC Nephrol ; 15: 126, 2014 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-25073618

RESUMEN

BACKGROUND: Intratracheal aspiration and sepsis are leading causes of acute lung injury that frequently necessitate mechanical ventilation (MV), which may aggravate lung injury thereby potentially increasing the risk of acute kidney injury (AKI). We compared the effects of ventilation strategies and underlying conditions on the development of AKI. METHODS: Spraque Dawley rats were challenged by intratracheal acid instillation or 24 h of abdominal sepsis, followed by MV with a low tidal volume (LVT) and 5 cm H2O positive end-expiratory pressure (PEEP) or a high tidal volume (HVT) and no PEEP, which is known to cause more lung injury after acid instillation than in sepsis. Rats were ventilated for 4 hrs and kidney function and plasma mediator levels were measured. Kidney injury was assessed by microscopy; apoptosis was quantified by TUNEL staining. RESULTS: During sepsis, but not after acid instillation, MV with HVT caused more renal apoptosis than MV with LVT. Increased plasma active plasminogen activator inhibitor-1 correlated to kidney apoptosis in the cortex and medulla. Increased apoptosis after HVT ventilation during sepsis was associated with a 40% decrease in creatinine clearance. CONCLUSIONS: AKI is more likely to develop after MV induced lung injury during an indirect (as in sepsis) than after a direct (as after intra-tracheal instillation) insult to the lungs, since it induces kidney apoptosis during sepsis but not after acid instillation, opposite to the lung injury it caused. Our findings thus suggest using protective ventilatory strategies in human sepsis, even in the absence of overt lung injury, to protect the kidney.


Asunto(s)
Lesión Renal Aguda/patología , Apoptosis , Ácido Clorhídrico/toxicidad , Riñón/patología , Respiración Artificial/efectos adversos , Sepsis/patología , Lesión Renal Aguda/etiología , Lesión Renal Aguda/fisiopatología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Ácido Clorhídrico/administración & dosificación , Intubación Intratraqueal/efectos adversos , Riñón/efectos de los fármacos , Riñón/fisiopatología , Masculino , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Sepsis/inducido químicamente
3.
Crit Care ; 18(1): R22, 2014 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-24456613

RESUMEN

INTRODUCTION: Experimental work provides insight into potential lung protective strategies. The objective of this study was to evaluate markers of ventilator-induced lung injury after two different ventilation approaches: (1) a "conventional" lung-protective strategy (volume control (VC) with low tidal volume, positive end-expiratory pressure (PEEP) and paralysis), (2) a physiological approach with spontaneous breathing, permitting synchrony, variability and a liberated airway. For this, we used non-invasive Neurally Adjusted Ventilatory Assist (NIV-NAVA), with the hypothesis that liberation of upper airways and the ventilator's integration with lung protective reflexes would be equally lung protective. METHODS: In this controlled and randomized in vivo laboratory study, 25 adult White New Zealand rabbits were studied, including five non-ventilated control animals. The twenty animals with aspiration-induced lung injury were randomized to ventilation with either VC (6 mL/kg, PEEP 5 cm H2O, and paralysis) or NIV-NAVA for six hours (PEEP = zero because of leaks). Markers of lung function, lung injury, vital signs and ventilator parameters were assessed. RESULTS: At the end of six hours of ventilation (n = 20), there were no significant differences between VC and NIV-NAVA for vital signs, PaO2/FiO2 ratio, lung wet-to-dry ratio and broncho-alveolar Interleukin 8 (Il-8). Plasma IL-8 was higher in VC (P <0.05). Lung injury score was lower for NIV-NAVA (P = 0.03). Dynamic lung compliance recovered after six hours in NIV-NAVA but not in VC (P <0.05). During VC, peak pressures increased from 9.2 ± 2.4 cm H2O (hour 1) to 12.3 ± 12.3 cm H2O (hour 6) (P <0.05). During NIV-NAVA, the tracheal end-expiratory pressure was similar to the end-expiratory pressure during VC. Two animals regurgitated during NIV-NAVA, without clinical consequences, and survived the protocol. CONCLUSIONS: In experimental acute lung injury, NIV-NAVA is as lung-protective as VC 6 ml/kg with PEEP.


Asunto(s)
Lesión Pulmonar Aguda/fisiopatología , Respiración con Presión Positiva/métodos , Lesión Pulmonar Inducida por Ventilación Mecánica/prevención & control , Lesión Pulmonar Aguda/terapia , Animales , Modelos Animales de Enfermedad , Pulmón/patología , Conejos , Distribución Aleatoria , Respiración , Volumen de Ventilación Pulmonar , Lesión Pulmonar Inducida por Ventilación Mecánica/patología
4.
PLoS One ; 7(10): e45506, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23071521

RESUMEN

OBJECTIVES: To perform a meta-analysis of gene expression microarray data from animal studies of lung injury, and to identify an injury-specific gene expression signature capable of predicting the development of lung injury in humans. METHODS: We performed a microarray meta-analysis using 77 microarray chips across six platforms, two species and different animal lung injury models exposed to lung injury with or/and without mechanical ventilation. Individual gene chips were classified and grouped based on the strategy used to induce lung injury. Effect size (change in gene expression) was calculated between non-injurious and injurious conditions comparing two main strategies to pool chips: (1) one-hit and (2) two-hit lung injury models. A random effects model was used to integrate individual effect sizes calculated from each experiment. Classification models were built using the gene expression signatures generated by the meta-analysis to predict the development of lung injury in human lung transplant recipients. RESULTS: Two injury-specific lists of differentially expressed genes generated from our meta-analysis of lung injury models were validated using external data sets and prospective data from animal models of ventilator-induced lung injury (VILI). Pathway analysis of gene sets revealed that both new and previously implicated VILI-related pathways are enriched with differentially regulated genes. Classification model based on gene expression signatures identified in animal models of lung injury predicted development of primary graft failure (PGF) in lung transplant recipients with larger than 80% accuracy based upon injury profiles from transplant donors. We also found that better classifier performance can be achieved by using meta-analysis to identify differentially-expressed genes than using single study-based differential analysis. CONCLUSION: Taken together, our data suggests that microarray analysis of gene expression data allows for the detection of "injury" gene predictors that can classify lung injury samples and identify patients at risk for clinically relevant lung injury complications.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Biomarcadores/metabolismo , Lesión Pulmonar Aguda/genética , Animales , Línea Celular , Bases de Datos Factuales , Perfilación de la Expresión Génica , Humanos , Pulmón/metabolismo , Trasplante de Pulmón/efectos adversos , Análisis por Micromatrices , Modelos Animales , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas
5.
Am J Pathol ; 181(5): 1681-92, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23083833

RESUMEN

Although bone marrow-derived mesenchymal stem cell (MSC) systemic administration reduces sepsis-associated inflammation, organ injury, and mortality in clinically relevant models of polymicrobial sepsis, the cellular and molecular mechanisms mediating beneficial effects are controversial. This study identifies the molecular mechanisms of MSC-conferred protection in sepsis by interrogating transcriptional responses of target organs to MSC therapy. Sepsis was induced in C57Bl/6J mice by cecal ligation and puncture, followed 6 hours later by an i.v. injection of either MSCs or saline. Total RNA from lungs, hearts, kidneys, livers, and spleens harvested 28 hours after cecal ligation and puncture was hybridized to mouse expression bead arrays. Common transcriptional responses were analyzed using a network knowledge-based approach. A total of 4751 genes were significantly changed between placebo- and MSC-treated mice (adjusted P ≤ 0.05). Transcriptional responses identified three common effects of MSC administration in all five organs examined: i) attenuation of sepsis-induced mitochondrial-related functional derangement, ii down-regulation of endotoxin/Toll-like receptor innate immune proinflammatory transcriptional responses, and iii) coordinated expression of transcriptional programs implicated in the preservation of endothelial/vascular integrity. Transcriptomic analysis indicates that the protective effect of MSC therapy in sepsis is not limited to a single mediator or pathway but involves a range of complementary activities affecting biological networks playing critical roles in the control of host cell metabolism and inflammatory response.


Asunto(s)
Perfilación de la Expresión Génica , Redes Reguladoras de Genes/genética , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Sepsis/genética , Sepsis/terapia , Transcripción Genética , Animales , Ciego/lesiones , Comunicación Celular/genética , Metabolismo Energético/genética , Femenino , Regulación de la Expresión Génica , Inmunidad/genética , Inflamación/genética , Inflamación/patología , Ligadura , Ratones , Ratones Endogámicos C57BL , Mitocondrias/genética , Especificidad de Órganos/genética , Sepsis/inducido químicamente , Sepsis/prevención & control , Transducción de Señal/genética
6.
Intensive Care Med ; 38(9): 1548-55, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22711173

RESUMEN

PURPOSE: Gamma-aminobutyric acid (GABA) is the major inhibitory neurotransmitter through activation of GABA receptors. Volatile anesthetics activate type-A (GABA(A)) receptors resulting in inhibition of synaptic transmission. Lung epithelial cells have been recently found to express GABA(A) receptors that exert anti-inflammatory properties. We hypothesized that the volatile anesthetic sevoflurane (SEVO) attenuates lung inflammation through activation of lung epithelial GABA(A) receptors. METHODS: Sprague-Dawley rats were anesthetized with SEVO or ketamine/xylazine (KX). Acute lung inflammation was induced by intratracheal instillation of endotoxin, followed by mechanical ventilation for 4 h at a tidal volume of 15 mL/kg without positive end-expiratory pressure (two-hit lung injury model). To examine the specific effects of GABA, healthy human lung epithelial cells (BEAS-2B) were challenged with endotoxin in the presence and absence of GABA with and without addition of the GABA(A) receptor antagonist picrotoxin. RESULTS: Anesthesia with SEVO improved oxygenation and reduced pulmonary cytokine responses compared to KX. This phenomenon was associated with increased expression of the π subunit of GABA(A) receptors and glutamic acid decarboxylase (GAD). The endotoxin-induced cytokine release from BEAS-2B cells was attenuated by the treatment with GABA, which was reversed by the administration of picrotoxin. CONCLUSION: Anesthesia with SEVO suppresses pulmonary inflammation and thus protects the lung from the two-hit injury. The anti-inflammatory effect of SEVO is likely due to activation of pulmonary GABA(A) signaling pathways.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Anestesia/efectos adversos , Anestésicos Generales/efectos adversos , Inflamación/etiología , Pulmón/patología , Respiración Artificial/efectos adversos , Lesión Pulmonar Aguda/patología , Análisis de Varianza , Anestésicos Disociativos/efectos adversos , Anestésicos por Inhalación/efectos adversos , Animales , Lavado Broncoalveolar , Modelos Animales de Enfermedad , Hemodinámica , Humanos , Inflamación/patología , Ketamina/efectos adversos , Pulmón/citología , Éteres Metílicos/efectos adversos , Ratas , Ratas Sprague-Dawley , Riesgo , Sevoflurano , Xilazina/efectos adversos , Ácido gamma-Aminobutírico
7.
Crit Care Med ; 40(6): 1896-907, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22610192

RESUMEN

OBJECTIVES: We hypothesized that resveratrol administration would reverse sepsis-dependent downregulation of peroxisome proliferator activated receptor-γ coactivator 1α, preserve mitochondrial integrity, and rescue animals from sepsis-induced myocardial failure. SETTING: Teaching hospital research laboratory. INTERVENTIONS: Cecal ligation and puncture in mice was performed to induce sepsis. Mice that underwent cecal ligation and puncture were randomly assigned to receive resveratrol (30 mg/kg or 60 mg/kg) or vehicle 1 mL sodium chloride 0.9% subcutaneously in the scruff of the neck directly after surgery and at 16, 24, and 40 hrs, respectively. MEASUREMENTS AND RESULTS: Forty-eight hrs after cecal ligation and puncture, cardiac performance was established using echocardiography. Mitochondrial integrity was evaluated with electron microscopy, and changes in gene expression were evaluated with microarray analysis. Survival at 48 hrs was just under 50% and comparable between groups. Myocardial contractile function significantly improved after resveratrol treatment. Resveratrol-treated mice developed focal areas of edema, whereas vehicle-treated mice developed significant, diffuse myocardial edema. Electron microscopy revealed widespread swollen mitochondria with ruptured outer membranes, autophagosomes, and vacuolation of the internal compartment, which were significantly attenuated in resveratrol-treated animals. Resveratrol treatment significantly increased cardiac expression of peroxisome proliferator-activated receptor-γ coactivator 1a. Microarray analysis revealed that resveratrol treatment resulted in upregulation of the peroxisome proliferator-activated receptor-γ coactivator gene set containing genes known to be regulated by this transcriptional coactivator. Our data strongly suggest that administration of resveratrol modulates bioenergy metabolism, substrate utilization, oxidative stress, and detoxification pathways associated with both mitochondrial and cardiac pathological conditions, but does not alter mortality from sepsis. CONCLUSIONS: The salutary effects of resveratrol on cecal ligation and puncture-induced myocardial dysfunction are associated with increased peroxisome proliferator-activated receptor-γ coactivator 1a abundance and function. Preservation of myocardial energy production capacity, prevention of secondary injury, mitigation of inflammation, and reversal of sepsis-induced myocardial remodeling are likely to underlie its beneficial effects. This however, does not result in improved survival.


Asunto(s)
Insuficiencia Cardíaca/prevención & control , Contracción Miocárdica/efectos de los fármacos , Sepsis/complicaciones , Estilbenos/farmacología , Transactivadores/metabolismo , Vasodilatadores/farmacología , Animales , Cardiomiopatías/etiología , Ciego , Regulación hacia Abajo/efectos de los fármacos , Edema/etiología , Expresión Génica/efectos de los fármacos , Insuficiencia Cardíaca/etiología , Ligadura , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/ultraestructura , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Distribución Aleatoria , Resveratrol , Transactivadores/genética , Factores de Transcripción
8.
BMC Anesthesiol ; 11: 26, 2011 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-22204611

RESUMEN

BACKGROUND: To examine whether acute lung injury from direct and indirect origins differ in susceptibility to ventilator-induced lung injury (VILI) and resultant systemic inflammatory responses. METHODS: Rats were challenged by acid instillation or 24 h of sepsis induced by cecal ligation and puncture, followed by mechanical ventilation (MV) with either a low tidal volume (Vt) of 6 mL/kg and 5 cm H2O positive end-expiratory pressure (PEEP; LVt acid, LVt sepsis) or with a high Vt of 15 mL/kg and no PEEP (HVt acid, HVt sepsis). Rats sacrificed immediately after acid instillation and non-ventilated septic animals served as controls. Hemodynamic and respiratory variables were monitored. After 4 h, lung wet to dry (W/D) weight ratios, histological lung injury and plasma mediator concentrations were measured. RESULTS: Oxygenation and lung compliance decreased after acid instillation as compared to sepsis. Additionally, W/D weight ratios and histological lung injury scores increased after acid instillation as compared to sepsis. MV increased W/D weight ratio and lung injury score, however this effect was mainly attributable to HVt ventilation after acid instillation. Similarly, effects of HVt on oxygenation were only observed after acid instillation. HVt during sepsis did not further affect oxygenation, compliance, W/D weight ratio or lung injury score. Plasma interleukin-6 and tumour necrosis factor-α concentrations were increased after acid instillation as compared to sepsis, but plasma intercellular adhesion molecule-1 concentration increased during sepsis only. In contrast to lung injury parameters, no additional effects of HVt MV after acid instillation on plasma mediator concentrations were observed. CONCLUSIONS: During MV more severe lung injury develops after acid instillation as compared to sepsis. HVt causes VILI after acid instillation, but not during sepsis. However, this differential effect was not observed in the systemic release of mediators.

9.
Curr Opin Anaesthesiol ; 24(2): 214-8, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21293265

RESUMEN

PURPOSE OF REVIEW: It has become clear from experimental data that prolonged mechanical ventilation can induce diaphragm dysfunction, also known as ventilator-induced diaphragm dysfunction. In this article we will discuss most recent understanding on ventilator-induced diaphragm dysfunction and data on diaphragm dysfunction in patients. RECENT FINDINGS: Over the last year several studies confirmed the existence of diaphragm dysfunction in patients. Known atrophy pathways are activated in patients undergoing prolonged conventional ventilation resulting in muscle proteolysis and a decrease in myofiber content. The loss of diaphragm force is time-dependent, but current data do not distinguish between the role played by other factors involved in diaphragm dysfunction. SUMMARY: Diaphragm dysfunction occurs in patients, especially when ventilated with controlled modes of ventilation that minimize diaphragm activity. Time on the ventilator seems to be one of the biggest risk factors resulting in difficulties in weaning patients and prolonging time on the ventilator. Future trials should investigate whether improved patient-ventilator synchrony can reduce ventilator-induced diaphragm dysfunction and decrease weaning failure.


Asunto(s)
Diafragma/lesiones , Diafragma/fisiopatología , Respiración Artificial/efectos adversos , Animales , Diafragma/metabolismo , Humanos , Proteínas Musculares/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/fisiopatología , Lesión Pulmonar Inducida por Ventilación Mecánica/prevención & control
10.
Intensive Care Med ; 37(2): 334-42, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21072499

RESUMEN

OBJECTIVE: Long pentraxin PTX3 is an inflammatory mediator and a component of the humoral arm of innate immunity. PTX3 expression is increased in animals with acute lung injury (ALI) and in patients with sepsis or acute respiratory distress syndrome and is considered to be a potential biomarker for these diseases. However, the role of PTX3 in the pathogenesis of ALI is not fully understood. We hypothesized that PTX3, as an important immune modulator, may determine the severity of ALI. METHODS: Lipopolysaccharide (LPS) was intra-tracheally administrated to PTX3 knock-out (PTX3-KO) and wild-type (WT) mice. Lung injury, neutrophil infiltration, cell death, fibrin deposition, and tissue factor expression in the lung were determined. Local and systemic inflammatory responses were assessed by measuring cytokines in the lung and plasma. RESULTS: LPS instillation induced ALI in both PTX3-KO and WT mice. Interestingly, PTX3 deficiency significantly increased the magnitude/extent of lung injury compared to that in WT mice. The severe lung injury was accompanied by elevated neutrophil infiltration, cell death, and fibrin deposition in the lung. PTX3 deficiency also enhanced LPS-induced tissue factor expression/activation in the lung and increased tumor necrosis factor-alpha and monocyte chemoattractant protein-1 levels in the plasma. CONCLUSION: Our data suggest that the endogenously expressed PTX3 plays a protective role in the pathogenesis of ALI and that a lack of PTX3 may enhance neutrophil recruitment, cell death, activation of coagulation cascades, and inflammatory responses in the lung.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/fisiopatología , Proteína C-Reactiva/metabolismo , Componente Amiloide P Sérico/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Animales , Animales Modificados Genéticamente , Coagulación Sanguínea/fisiología , Muerte Celular , Técnicas de Inactivación de Genes , Inflamación , Lipopolisacáridos , Ratones , Infiltración Neutrófila , Índice de Severidad de la Enfermedad
11.
Am J Respir Crit Care Med ; 183(4): 471-82, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20870758

RESUMEN

RATIONALE: Ventilator-induced lung injury (VILI) contributes to the mortality in patients with acute lung injury by increasing inflammation. Recent evidence suggests that stimulation of the cholinergic antiinflammatory pathway may be an attractive way to attenuate inflammatory injury. OBJECTIVES: To determine the role of vagus nerve signaling in VILI and establish whether stimulation of the vagus reflex can mitigate VILI. METHODS: We performed bilateral vagotomy in a mouse model of high-tidal volume-induced lung injury. We performed pharmacological and electrical vagus nerve stimulation in a rat model of VILI following ischemia/reperfusion injury. To determine the contribution of the alpha 7 acetylcholine nicotinic receptor to pulmonary cell injury, we exposed human bronchial epithelial cells to cyclic stretch in the presence of specific agonist or antagonist of the alpha 7 receptor. MEASUREMENTS AND MAIN RESULTS: Vagotomy exacerbates lung injury from VILI in mice as demonstrated by increased wet-to-dry ratio, infiltration of neutrophils, and increased IL-6. Vagal stimulation attenuates lung injury in rats after ischemia/reperfusion injury ventilated with high-volume strategies. Treatment of both mice and rats with the vagus mimetic drug semapimod resulted in decreased lung injury. Vagotomy also increased pulmonary apoptosis, whereas vagus stimulation (electrical and pharmacological) attenuated VILI-induced apoptosis. In vitro studies suggest that vagus-dependent effects on inflammation and apoptosis are mediated via the α7 nicotinc acetylcholine receptor-dependent effects on cyclic stretch-dependent signaling pathways c-jun N-terminal kinase and tumor necrosis factor receptor superfamily, member 6. CONCLUSIONS: Stimulation of the cholinergic antiinflammatory reflex may represent a promising alternative for the treatment of VILI.


Asunto(s)
Neuroinmunomodulación/inmunología , Lesión Pulmonar Inducida por Ventilación Mecánica/inmunología , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/inmunología , Animales , Apoptosis , Células Cultivadas , Modelos Animales de Enfermedad , Estimulación Eléctrica , Humanos , Hidrazonas/administración & dosificación , Inmunosupresores/administración & dosificación , Mediadores de Inflamación/inmunología , Interleucina-6/inmunología , Pulmón/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Receptores Nicotínicos/inmunología , Daño por Reperfusión , Nervio Vago/efectos de los fármacos , Nervio Vago/inmunología , Lesión Pulmonar Inducida por Ventilación Mecánica/complicaciones
12.
Am J Respir Crit Care Med ; 182(8): 1047-57, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20558630

RESUMEN

RATIONALE: Sepsis refers to the clinical syndrome of severe systemic inflammation precipitated by infection. Despite appropriate antimicrobial therapy, sepsis-related morbidity and mortality remain intractable problems in critically ill patients. Moreover, there is no specific treatment strategy for the syndrome of sepsis-induced multiple organ dysfunction. OBJECTIVES: We hypothesized that mesenchymal stem cells (MSCs), which have been shown to have immunomodulatory properties, would reduce sepsis-induced inflammation and improve survival in a polymicrobial model of sepsis. METHODS: Sepsis was induced in C57Bl/6J mice by cecal ligation and puncture (CLP), followed 6 hours later by an intravenous injection of MSCs or saline. Twenty-eight hours after CLP, plasma, bronchoalveolar lavage fluid and tissues were collected for analyses. Longer-term studies were performed with antibiotic coadministration to assess the effect of MSCs on survival. MEASUREMENTS AND MAIN RESULTS: MSC treatment significantly reduced mortality in septic mice receiving appropriate antimicrobial therapy. MSCs alone reduced systemic and pulmonary cytokine levels in mice with CLP-induced sepsis, preventing acute lung injury and organ dysfunction, despite the low levels of cell persistence. Microarray data highlighted an overall down-regulation of inflammation and inflammation-related genes (such as IL-10, IL-6) and a shift toward up-regulation of genes involved in promoting phagocytosis and bacterial killing. Finally, bacterial clearance was significantly greater in MSC-treated mice, in part due to enhanced phagocytotic activity of the host immune cells. CONCLUSIONS: These data demonstrate that MSCs have beneficial effects on experimental sepsis, possibly by paracrine mechanisms, and suggest that immunomodulatory cell therapy may be an effective adjunctive treatment to reduce sepsis-related morbidity and mortality.


Asunto(s)
Lesión Pulmonar Aguda/terapia , Trasplante de Células Madre Mesenquimatosas , Insuficiencia Multiorgánica/terapia , Sepsis/terapia , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/inmunología , Animales , Antibacterianos/uso terapéutico , Terapia Combinada , Femenino , Regulación de la Expresión Génica , Inmunomodulación , Inflamación/genética , Inflamación/terapia , Ratones , Ratones Endogámicos C57BL , Insuficiencia Multiorgánica/genética , Insuficiencia Multiorgánica/inmunología , Sepsis/genética , Sepsis/inmunología , Análisis de Supervivencia
13.
Am J Respir Crit Care Med ; 182(4): 489-500, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20413626

RESUMEN

RATIONALE: Ventilator-induced lung injury (VILI) significantly contributes to mortality in patients with acute respiratory distress syndrome, the most severe form of acute lung injury. Understanding the molecular basis for response to cyclic stretch (CS) and its derangement during high-volume ventilation is of high priority. OBJECTIVES: To identify specific molecular regulators involved in the development of VILI. METHODS: We undertook a comparative examination of cis-regulatory sequences involved in the coordinated expression of CS-responsive genes using microarray analysis. Analysis of stretched versus nonstretched cells identified significant enrichment for genes containing putative binding sites for the transcription factor activating transcription factor 3 (ATF3). To determine the role of ATF3 in vivo, we compared the response of ATF3 gene-deficient mice to wild-type mice in an in vivo model of VILI. MEASUREMENTS AND MAIN RESULTS: ATF3 protein expression and nuclear translocation is increased in the lung after mechanical ventilation in wild-type mice. ATF3-deficient mice have greater sensitivity to mechanical ventilation alone or in conjunction with inhaled endotoxin, as demonstrated by increased cell infiltration and proinflammatory cytokines in the lung and bronchoalveolar lavage, and increased pulmonary edema and indices of tissue injury. The expression of stretch-responsive genes containing putative ATF3 cis-regulatory regions was significantly altered in ATF3-deficient mice. CONCLUSIONS: ATF3 deficiency confers increased sensitivity to mechanical ventilation alone or in combination with inhaled endotoxin. We propose ATF3 acts to counterbalance CS and high volume-induced inflammation, dampening its ability to cause injury and consequently protecting animals from injurious CS.


Asunto(s)
Factor de Transcripción Activador 3/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/prevención & control , Animales , Western Blotting/métodos , Líquido del Lavado Bronquioalveolar , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Perfilación de la Expresión Génica/métodos , Humanos , Pulmón/metabolismo , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Respiración Artificial/efectos adversos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
14.
BMC Pulm Med ; 10: 6, 2010 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-20158912

RESUMEN

BACKGROUND: Preventing ventilator-associated lung injury (VALI) has become pivotal in mechanical ventilation of patients with acute lung injury (ALI) or its more severe form, acute respiratory distress syndrome (ARDS). In the present study we investigated whether plasma levels of lung-specific biological markers can be used to evaluate lung injury in patients with ALI/ARDS and patients without lung injury at onset of mechanical ventilation. METHODS: Plasma levels of surfactant protein D (SP-D), Clara Cell protein (CC16), KL-6 and soluble receptor for advanced glycation end-products (sRAGE) were measured in plasma samples obtained from 36 patients - 16 patients who were intubated and mechanically ventilated because of ALI/ARDS and 20 patients without lung injury at the onset of mechanical ventilation and during conduct of the study. Patients were ventilated with either a lung-protective strategy using lower tidal volumes or a potentially injurious strategy using conventional tidal volumes. Levels of biological markers were measured retrospectively at baseline and after 2 days of mechanical ventilation. RESULTS: Plasma levels of CC16 and KL-6 were higher in ALI/ARDS patients at baseline as compared to patients without lung injury. SP-D and sRAGE levels were not significantly different between these patients. In ALI/ARDS patients, SP-D and KL-6 levels increased over time, which was attenuated by lung-protective mechanical ventilation using lower tidal volumes (P = 0.02 for both biological markers). In these patients, with either ventilation strategy no changes over time were observed for plasma levels of CC16 and sRAGE. In patients without lung injury, no changes of plasma levels of any of the measured biological markers were observed. CONCLUSION: Plasma levels of SP-D and KL-6 rise with potentially injurious ventilator settings, and thus may serve as biological markers of VALI in patients with ALI/ARDS.


Asunto(s)
Biomarcadores/sangre , Mucina-1/sangre , Proteína D Asociada a Surfactante Pulmonar/sangre , Respiración Artificial/efectos adversos , Síndrome de Dificultad Respiratoria , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad Crítica , Femenino , Humanos , Tiempo de Internación , Masculino , Persona de Mediana Edad , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/sangre , Síndrome de Dificultad Respiratoria/sangre , Síndrome de Dificultad Respiratoria/diagnóstico , Síndrome de Dificultad Respiratoria/etiología , Estudios Retrospectivos , Volumen de Ventilación Pulmonar , Resultado del Tratamiento , Uteroglobina/sangre
15.
Eur J Cardiothorac Surg ; 35(2): 304-12; discussion 312, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19109031

RESUMEN

OBJECTIVE: Lung ischemia-reperfusion injury (LIRI) is a risk factor for primary acute graft failure following lung transplantation. LIRI hereby contributes to morbidity and mortality after lung transplantation. We have previously shown that surfactant pretreatment ameliorates LIRI up to 1 week after reperfusion. However, the impact of surfactant pretreatment on long-term outcome following LIRI is unknown. Therefore, the objective of this study was to investigate the effect of surfactant pretreatment on long-term outcome following LIRI. METHODS: Male Sprague-Dawley rats (n=63) were randomized to receive intratracheally administered porcine surfactant (400mg/kg) or no pretreatment. One hour thereafter, animals underwent 120min of warm ischemia by clamping the bronchus, pulmonary artery and vein of the left lung. A third group was sham-operated; a fourth group served as unoperated controls. Animals were killed on day 30 or 90 after surgery. Arterial oxygenation and lung compliance were determined. Broncho-alveolar lavage fluid (BALf) was collected to assess surfactant function and alveolar protein. Leukocyte infiltration was determined by flowcytometry in BALf, lung tissue and thoracic lymph nodes. Lungs of three animals per group were used for histological assessment. RESULTS: Lung compliance was lower on day 30 and day 90 after LIRI than in sham-operated controls (day 30 V(max) 6.1+/-2.1 vs 12.6+/-1.3, day 90 6.9+/-3.0 vs 12.1+/-1.6; C(max) day 30 0.49+/-0.17 vs 1.08+/-0.21, day 90 0.67+/-0.31 vs 1.11+/-0.17). Furthermore, the number of CD45RA(+)-lymphocytes in left lung tissue was decreased on day 90 compared to unoperated animals (230.633+/-96.770 vs 696.347+/-202.909) and the number of macrophages elevated in left BALf on day 90. HE slides of LIRI animals were scored as fibroproliferative with moderate atelectasis. Surfactant pretreatment improved lung compliance (V(max) day 30 11.7+/-1.8, day 90 11.1+/-1.2; C(max) day 30 1.04+/-0.23, day 90 1.16+/-0.21) and normalized the number of CD45RA(+)-lymphocytes (769.555+/-421.016) in left lung tissue. Furthermore lung architecture on HE slides was on return to normal. However, more CD5(+)CD4(+)-lymphocytes on day 30 (754.788+/-97.269 vs 430.409+/-109.909) and more macrophages on day 90 (2.144.000+/-630.633 vs 867.454+/-383.220) were measured in pretreated lung tissue compared to LIRI animals. CONCLUSIONS: Severe LIRI caused extensive pulmonary injury up to 90 days postoperatively. Surfactant pretreatment normalized pulmonary function, but resulted in an increased number of CD5(+)CD4(+)-cells and macrophages in lung tissue.


Asunto(s)
Trasplante de Pulmón/patología , Pulmón/irrigación sanguínea , Surfactantes Pulmonares/uso terapéutico , Daño por Reperfusión/prevención & control , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Dióxido de Carbono/sangre , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Pulmón/inmunología , Pulmón/patología , Rendimiento Pulmonar , Trasplante de Pulmón/inmunología , Trasplante de Pulmón/fisiología , Ganglios Linfáticos/inmunología , Subgrupos Linfocitarios/inmunología , Macrófagos Alveolares/inmunología , Masculino , Oxígeno/sangre , Presión Parcial , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/sangre , Daño por Reperfusión/inmunología , Daño por Reperfusión/fisiopatología
16.
Shock ; 31(3): 262-6, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18650784

RESUMEN

Ventilator-induced lung injury is mediated, at least in part, by TNF-alpha. We determined the effect of a recombinant human soluble TNF receptor fusion protein (etanercept) on mechanical ventilation (MV)-induced changes in a murine ventilator-induced lung injury model. After pretreatment with etanercept or placebo, C57Bl/6 mice were anesthetized and randomized to MV with either low tidal volumes (VT, approximately 7.5 mL/kg) or high VT ( approximately 15 mL/kg) for 5 h. Instrumented but spontaneously breathing mice served as controls. End points were lung wet-to-dry ratios, lung histopathology scores, protein levels, neutrophil cell counts and thrombin-antithrombin complex levels in bronchoalveolar lavage fluid (BALF), and cytokine levels in lung homogenates. The number of caspase 3-positive cells was used as a measure for apoptosis. Etanercept treatment attenuated MV-induced changes, in particular, in MV with high VT. Compared with placebo, etanercept reduced the number of neutrophils in BALF and thrombin-antithrombin complex levels in BALF and cytokine levels in lung homogenates. Lung wet-to-dry ratios, histopathology scores, and local protein levels in BALF, however, were not influenced by etanercept treatment. The number of caspase 3-positive cells was significantly higher in etanercept-treated animals. Inhibition of TNF by etanercept attenuates, in part, MV-induced changes.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Inmunoglobulina G/farmacología , Pulmón/inmunología , Proteínas Recombinantes de Fusión/farmacología , Factor de Necrosis Tumoral alfa/inmunología , Lesión Pulmonar Inducida por Ventilación Mecánica/prevención & control , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Caspasa 3/inmunología , Modelos Animales de Enfermedad , Etanercept , Femenino , Humanos , Pulmón/patología , Ratones , Receptores del Factor de Necrosis Tumoral , Lesión Pulmonar Inducida por Ventilación Mecánica/inmunología , Lesión Pulmonar Inducida por Ventilación Mecánica/patología
17.
Semin Thromb Hemost ; 34(5): 475-84, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18956288

RESUMEN

Disturbed alveolar fibrin turnover is intrinsic to acute lung injury/acute respiratory distress syndrome (ALI/ARDS) and pneumonia and is important to its pathogenesis. Recent studies also suggest disturbed alveolar fibrin turnover to be a feature of ventilator-induced lung injury (VILI). The mechanisms that contribute to alveolar coagulopathy are localized tissue factor-mediated thrombin generation, impaired activity of natural coagulation inhibitors, and depression of bronchoalveolar urokinase plasminogen activator-mediated fibrinolysis, caused by the increase of plasminogen activator inhibitors. Administration of anticoagulant agents (including activated protein C, antithrombin, tissue factor-factor VIIa pathway inhibitors, and heparin) and profibrinolytic agents (including plasminogen activators) attenuate pulmonary coagulopathy. Several preclinical studies show additional anti-inflammatory effects of these therapies in ALI/ARDS and pneumonia. In this article, we review the involvement of coagulation and fibrinolysis in the pathogenesis of ALI/ARDS pneumonia and VILI and the potential of anticoagulant and profibrinolytic strategies to reverse pulmonary coagulopathy and pulmonary inflammatory responses.


Asunto(s)
Lesión Pulmonar Aguda/sangre , Bronquios/fisiopatología , Fibrina/metabolismo , Hemostasis , Alveolos Pulmonares/fisiopatología , Síndrome de Dificultad Respiratoria/sangre , Lesión Pulmonar Aguda/metabolismo , Bronquios/metabolismo , Citocinas/metabolismo , Fibrinólisis , Humanos , Activadores Plasminogénicos/metabolismo , Alveolos Pulmonares/metabolismo , Receptores de Trombina/metabolismo , Síndrome de Dificultad Respiratoria/metabolismo , Trombina/metabolismo
18.
Am J Physiol Endocrinol Metab ; 295(3): E658-64, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18628355

RESUMEN

Sepsis is a multifactorial, and often fatal, disorder typically characterized by widespread inflammation and immune activation with resultant endothelial activation. In the present study, we postulated that the adipokine adiponectin serves as a critical modulator of survival and endothelial activation in sepsis. To this aim, we evaluated both loss-of-function (adiponectin gene-deficient mice) and subsequent gain-of-function (recombinant adiponectin reconstitution) strategies in two well-established inflammatory models, cecal ligation perforation (CLP) and thioglyocollate-induced peritonitis. Adipoq(-/-) mice, subjected to CLP, exhibited a profound ( approximately 8-fold) reduction in survival compared with their wild-type Adipoq(+/+) littermates after 48 h. Furthermore, compared with wild-type controls, thioglycollate challenge resulted in a markedly greater influx of peritoneal neutrophils in Adipoq(-/-) mice accompanied by an excess production of key chemoattractant cytokines (IL-12p70, TNFalpha, MCP-1, and IL-6) and upregulation of aortic endothelial adhesion molecule VCAM-1 and ICAM-1 expressions. Importantly, all of these effects were blunted by recombinant total adiponectin administration given 3 days prior to thioglycollate challenge. The protective effects of adiponectin were ascribed largely to higher-order adiponectin oligomers, since administration of recombinant C39A trimeric adiponectin did not attenuate endothelial adhesion molecule expression in thioglycollate-challenged Adipoq(-/-) mice. These data suggest a critical role of adiponectin as a modulator of survival and endothelial inflammation in experimental sepsis and a potential mechanistic link between adiposity and increased sepsis.


Asunto(s)
Adiponectina/deficiencia , Endotelio/fisiología , Sepsis/mortalidad , Sepsis/fisiopatología , Adiponectina/genética , Adiponectina/farmacología , Animales , Ciego/fisiología , Citocinas/biosíntesis , Inflamación/inducido químicamente , Inflamación/patología , Perforación Intestinal/patología , Recuento de Leucocitos , Ligadura , Ratones , Ratones Noqueados , Peritonitis/patología , Proteínas Recombinantes/farmacología , Sepsis/genética , Análisis de Supervivencia , Tioglicolatos
20.
Biochim Biophys Acta ; 1781(6-7): 314-20, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18485924

RESUMEN

Lung surfactant is a lipid-protein-film covering the inner alveolar surface. We have previously shown that double knock-out (d-ko) mice lacking both the epidermal-type (E-) and the heart-type (H-) fatty acid binding protein (FABP) exhibit a defect of surfactant synthesis in alveolar type II cells that can be corrected by feeding pioglitazone, a drug that activates peroxisome proliferator-activated receptor gamma (PPARgamma). Here, we demonstrate first that healthy surfactant at collapse pressure produces protrusions composed of bilayers but not folds, second that the d-ko effect profoundly perturbs lipid/hydrophobic protein composition, pressure-area isotherm, and structural organisation of the surfactant at nanoscale, parameters that are critical for the normal breathing cycle. In support of these data in vivo measurements of lung function reveal that maximum compliance in d-ko vs. wild-type mice is significantly reduced. Further, we show that the biophysical phenotype can be corrected substantially with pioglitazone. Finally, we show that d-ko alveolar cells up-regulate liver-type (L-) FABP, a member of the FABP family that we have previously shown to interact with PPARgamma. Taken together, these data suggest that PPARgamma agonists could be a tool to repair surfactant damage caused by dysfunctional alveolar lipid metabolism, and provide in vivo support for L-FABP aided signaling.


Asunto(s)
Proteínas de Unión a Ácidos Grasos/genética , PPAR gamma/agonistas , Surfactantes Pulmonares/metabolismo , Animales , Secuencia de Bases , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Hipoglucemiantes/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pioglitazona , Tiazolidinedionas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA