Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Genet Metab ; 123(4): 501-510, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29478818

RESUMEN

BACKGROUND: Gaucher disease is characterized by the activation of splenic and hepatic macrophages, accompanied by dramatically increased levels of angiotensin-converting enzyme (ACE). To evaluate the source of the elevated blood ACE, we performed complete ACE phenotyping using blood, spleen and liver samples from patients with Gaucher disease and controls. METHODS: ACE phenotyping included 1) immunohistochemical staining for ACE; 2) measuring ACE activity with two substrates (HHL and ZPHL); 3) calculating the ratio of the rates of substrate hydrolysis (ZPHL/HHL ratio); 4) assessing the conformational fingerprint of ACE by evaluating the pattern of binding of monoclonal antibodies to 16 different ACE epitopes. RESULTS: We show that in patients with Gaucher disease, the dramatically increased levels of ACE originate from activated splenic and/or hepatic macrophages (Gaucher cells), and that both its conformational fingerprint and kinetic characteristics (ZPHL/HHL ratio) differ from controls and from patients with sarcoid granulomas. Furthermore, normal spleen was found to produce high levels of endogenous ACE inhibitors and a novel, tightly-bound 10-30 kDa ACE effector which is deficient in Gaucher spleen. CONCLUSIONS: The conformation of ACE is tissue-specific. In Gaucher disease, ACE produced by activated splenic macrophages differs from that in hepatic macrophages, as well as from macrophages and dendritic cells in sarcoid granulomas. The observed differences are likely due to altered ACE glycosylation or sialylation in these diseased organs. The conformational differences in ACE may serve as a specific biomarker for Gaucher disease.


Asunto(s)
Células Dendríticas/enzimología , Enfermedad de Gaucher/enzimología , Enfermedad de Gaucher/patología , Granuloma/enzimología , Macrófagos/enzimología , Peptidil-Dipeptidasa A/metabolismo , Células Cultivadas , Humanos , Hígado/enzimología , Fenotipo , Bazo/enzimología
2.
J Vasc Res ; 53(5-6): 349-357, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27997923

RESUMEN

High-mobility group box 1 protein (HMGB1) has been implicated in inflammatory responses, and is also associated with cerebral vasospasm after subarachnoid hemorrhage (SAH). However, there are no direct evident links between HMGB1 and cerebral vasospasm. We therefore investigated the effects of HMGB1 on pial arteriole reactivity following SAH in rats. We initially found that SAH induced a significant decrease in pial arteriole dilating responses to sciatic nerve stimulation (SNS), hypercapnia (CO2), and the topical suffusion of acetylcholine (ACh), adenosine (ADO), and s-nitroso-N-acetylpenicillamine (SNAP) over a 7-day period after SAH. The percent change of arteriolar diameter was decreased to the lowest point at 48 h after SAH, in response to dilating stimuli (i.e., it decreased from 41.0 ± 19.0% in the sham group to 11.00 ± 0.70% after SNS) (n = 5, p < 0.01). HMGB1 infusion in the lateral ventricle in normal rats for 48 h did not change the pial arteriole dilating response. In addition, inhibitors of HMGB1-receptor for advanced glycation end-product or HMGB1-toll-like receptor 2/4 interaction, or the HMBG1 antagonist did not improve pial arteriole reactivity 48 h after SAH. These findings suggest that HMGB1 may not be a major player in cerebral vascular dilating dysfunction after SAH.


Asunto(s)
Arteriolas/metabolismo , Proteína HMGB1/metabolismo , Piamadre/irrigación sanguínea , Hemorragia Subaracnoidea/metabolismo , Vasodilatación , Animales , Arteriolas/efectos de los fármacos , Arteriolas/fisiopatología , Modelos Animales de Enfermedad , Estimulación Eléctrica , Proteína HMGB1/antagonistas & inhibidores , Proteína HMGB1/farmacología , Hipercapnia/metabolismo , Hipercapnia/fisiopatología , Masculino , Ratas Sprague-Dawley , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Nervio Ciático/fisiopatología , Transducción de Señal , Hemorragia Subaracnoidea/fisiopatología , Factores de Tiempo , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
3.
J Immunol ; 190(7): 3590-9, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23436933

RESUMEN

Mechanical ventilation of lungs is capable of activating the innate immune system and inducing sterile inflammatory response. The proinflammatory cytokine IL-1ß is among the definitive markers for accurately identifying ventilator-induced lung inflammation. However, mechanisms of IL-1ß release during mechanical ventilation are unknown. In this study, we show that cyclic stretch activates the nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasomes and induces the release of IL-1ß in mouse alveolar macrophages via caspase-1- and TLR4-dependent mechanisms. We also observed that NADPH oxidase subunit gp91(phox) was dispensable for stretch-induced cytokine production, whereas mitochondrial generation of reactive oxygen species was required for stretch-induced NLRP3 inflammasome activation and IL-1ß release. Further, mechanical ventilation activated the NLRP3 inflammasomes in mouse alveolar macrophages and increased the production of IL-1ß in vivo. IL-1ß neutralization significantly reduced mechanical ventilation-induced inflammatory lung injury. These findings suggest that the alveolar macrophage NLRP3 inflammasome may sense lung alveolar stretch to induce the release of IL-1ß and hence may contribute to the mechanism of lung inflammatory injury during mechanical ventilation.


Asunto(s)
Proteínas Portadoras/metabolismo , Inflamasomas/metabolismo , Macrófagos Alveolares/metabolismo , Neumonía/etiología , Animales , Caspasa 1/metabolismo , Modelos Animales de Enfermedad , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Macrófagos Alveolares/inmunología , Masculino , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Neumonía/inmunología , Neumonía/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Receptor Toll-Like 4/metabolismo , Ácido Úrico/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/inmunología , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo
4.
Crit Care ; 19: 45, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25887642

RESUMEN

INTRODUCTION: Severe sepsis is associated with approximately 50% mortality and accounts for tremendous healthcare costs. Most patients require ventilatory support and propofol is commonly used to sedate mechanically ventilated patients. Volatile anesthetics have been shown to attenuate inflammation in a variety of different settings. We therefore hypothesized that volatile anesthetic agents may offer beneficial immunomodulatory effects during the course of long-term intra-abdominal sepsis in rats under continuous sedation and ventilation for up to 24 hours. METHODS: Sham operation or cecal ligation and puncture (CLP) was performed in adult male Wistar rats followed by mechanical ventilation. Animals were sedated for 24 hours with propofol (7 to 20 mg/kg/h), sevoflurane, desflurane or isoflurane (0.7 minimal alveolar concentration each). RESULTS: Septic animals sedated with propofol showed a mean survival time of 12 hours, whereas >56% of all animals in the volatile groups survived 24 hours (P <0.001). After 18 hours, base excess in propofol + CLP animals (-20.6 ± 2.0) was lower than in the volatile groups (isoflurane + CLP: -11.7 ± 4.2, sevoflurane + CLP: -11.8 ± 3.5, desflurane + CLP -14.2 ± 3.7; all P <0.03). Plasma endotoxin levels reached 2-fold higher levels in propofol + CLP compared to isoflurane + CLP animals at 12 hours (P <0.001). Also blood levels of inflammatory mediators (tumor necrosis factor-α, interleukin-1ß, interleukin-10, CXCL-2, interferon-γ and high mobility group protein-1) were accentuated in propofol + CLP rats compared to the isoflurane + CLP group at the same time point (P <0.04). CONCLUSIONS: This is the first study to assess prolonged effects of sepsis and long-term application of volatile sedatives compared to propofol on survival, cardiovascular, inflammatory and end organ parameters. Results indicate that volatile anesthetics dramatically improved survival and attenuate systemic inflammation as compared to propofol. The main mechanism responsible for adverse propofol effects could be an enhanced plasma endotoxin concentration, leading to profound hypotension, which was unresponsive to fluid resuscitation.


Asunto(s)
Anestésicos Intravenosos/efectos adversos , Propofol/efectos adversos , Respiración Artificial , Sepsis/mortalidad , Animales , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Wistar , Sepsis/complicaciones
5.
Am J Physiol Lung Cell Mol Physiol ; 307(2): L173-85, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24838752

RESUMEN

The inflammatory response is a primary mechanism in the pathogenesis of ventilator-induced lung injury. Autophagy is an essential, homeostatic process by which cells break down their own components. We explored the role of autophagy in the mechanisms of mechanical ventilation-induced lung inflammatory injury. Mice were subjected to low (7 ml/kg) or high (28 ml/kg) tidal volume ventilation for 2 h. Bone marrow-derived macrophages transfected with a scrambled or autophagy-related protein 5 small interfering RNA were administered to alveolar macrophage-depleted mice via a jugular venous cannula 30 min before the start of the ventilation protocol. In some experiments, mice were ventilated in the absence and presence of autophagy inhibitors 3-methyladenine (15 mg/kg ip) or trichostatin A (1 mg/kg ip). Mechanical ventilation with a high tidal volume caused rapid (within minutes) activation of autophagy in the lung. Conventional transmission electron microscopic examination of lung sections showed that mechanical ventilation-induced autophagy activation mainly occurred in lung macrophages. Autophagy activation in the lungs during mechanical ventilation was dramatically attenuated in alveolar macrophage-depleted mice. Selective silencing of autophagy-related protein 5 in lung macrophages abolished mechanical ventilation-induced nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome activation and lung inflammatory injury. Pharmacological inhibition of autophagy also significantly attenuated the inflammatory responses caused by lung hyperinflation. The activation of autophagy in macrophages mediates early lung inflammation during mechanical ventilation via NLRP3 inflammasome signaling. Inhibition of autophagy activation in lung macrophages may therefore provide a novel and promising strategy for the prevention and treatment of ventilator-induced lung injury.


Asunto(s)
Autofagia/fisiología , Proteínas Portadoras/metabolismo , Inflamasomas/fisiología , Macrófagos Alveolares/fisiología , Lesión Pulmonar Inducida por Ventilación Mecánica/fisiopatología , Adenina/análogos & derivados , Adenina/farmacología , Animales , Autofagia/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia , Ácidos Hidroxámicos/farmacología , Macrófagos/fisiología , Macrófagos Alveolares/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/deficiencia , Proteína con Dominio Pirina 3 de la Familia NLR , Neumonía/patología , Neumonía/prevención & control , Especies Reactivas de Oxígeno/metabolismo , Respiración Artificial/efectos adversos , Transducción de Señal , Estrés Mecánico
6.
Anesthesiology ; 120(6): 1414-28, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24525631

RESUMEN

BACKGROUND: Pulmonary endothelial barrier dysfunction mediated in part by Src-kinase activation plays a crucial role in acute inflammatory disease. Proinflammatory cytokines, such as tumor necrosis factor-α (TNFα), activate Src via phosphatidylinositide 3-kinase/Akt-dependent nitric oxide generation, a process initiated by recruitment of phosphatidylinositide 3-kinase regulatory subunit p85 to TNF-receptor-1. Because amide-linked local anesthetics have well-established anti-inflammatory effects, the authors hypothesized that ropivacaine and lidocaine attenuate inflammatory Src signaling by disrupting the phosphatidylinositide 3-kinase-Akt-nitric oxide pathway, thus blocking Src-dependent neutrophil adhesion and endothelial hyperpermeability. METHODS: Human lung microvascular endothelial cells, incubated with TNFα in the absence or presence of clinically relevant concentrations of ropivacaine and lidocaine, were analyzed by Western blot, probing for phosphorylated/activated Src, endothelial nitric oxide synthase, Akt, intercellular adhesion molecule-1, and caveolin-1. The effect of ropivacaine on TNFα-induced nitric oxide generation, co-immunoprecipitation of TNF-receptor-1 with p85, neutrophil adhesion, and endothelial barrier disruption were assessed. RESULTS: Ropivacaine and lidocaine attenuated TNFα-induced Src activation (half-maximal inhibitory concentration [IC50] = 8.611 × 10 M for ropivacaine; IC50 = 5.864 × 10 M for lidocaine) and endothelial nitric oxide synthase phosphorylation (IC50 = 7.572 × 10 M for ropivacaine; IC50 = 6.377 × 10 M for lidocaine). Akt activation (n = 7; P = 0.006) and stimulus-dependent binding of TNF-receptor-1 and p85 (n = 6; P = 0.043) were blocked by 1 nM of ropivacaine. TNFα-induced neutrophil adhesion and disruption of endothelial monolayers via Src-dependent intercellular adhesion molecule-1- and caveolin-1-phosphorylation, respectively, were also attenuated. CONCLUSIONS: Ropivacaine and lidocaine effectively blocked inflammatory TNFα signaling in endothelial cells by attenuating p85 recruitment to TNF-receptor-1. The resultant decrease in Akt, endothelial nitric oxide synthase, and Src phosphorylation reduced neutrophil adhesion and endothelial hyperpermeability. This novel anti-inflammatory "side-effect" of ropivacaine and lidocaine may provide therapeutic benefit in acute inflammatory disease.


Asunto(s)
Amidas/farmacología , Anestésicos Locales/farmacología , Endotelio Vascular/efectos de los fármacos , Lidocaína/farmacología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Familia-src Quinasas/metabolismo , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/enzimología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Humanos , Pulmón/efectos de los fármacos , Pulmón/enzimología , Microcirculación/efectos de los fármacos , Microcirculación/fisiología , Ropivacaína , Factor de Necrosis Tumoral alfa/administración & dosificación , Familia-src Quinasas/fisiología
7.
BMC Anesthesiol ; 14: 57, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25097454

RESUMEN

BACKGROUND: Acute lung injury (ALI) is associated with high mortality due to the lack of effective therapeutic strategies. Mechanical ventilation itself can cause ventilator-induced lung injury. Pulmonary vascular barrier function, regulated in part by Src kinase-dependent phosphorylation of caveolin-1 and intercellular adhesion molecule-1 (ICAM-1), plays a crucial role in the development of protein-/neutrophil-rich pulmonary edema, the hallmark of ALI. Amide-linked local anesthetics, such as ropivacaine, have anti-inflammatory properties in experimental ALI. We hypothesized ropivacaine may attenuate inflammation in a "double-hit" model of ALI triggered by bacterial endotoxin plus hyperinflation via inhibition of Src-dependent signaling. METHODS: C57BL/6 (WT) and ICAM-1 (-/-) mice were exposed to either nebulized normal saline (NS) or lipopolysaccharide (LPS, 10 mg) for 1 hour. An intravenous bolus of 0.33 mg/kg ropivacaine or vehicle was followed by mechanical ventilation with normal (7 ml/kg, NTV) or high tidal volume (28 ml/kg, HTV) for 2 hours. Measures of ALI (excess lung water (ELW), extravascular plasma equivalents, permeability index, myeloperoxidase activity) were assessed and lungs were homogenized for Western blot analysis of phosphorylated and total Src, ICAM-1 and caveolin-1. Additional experiments evaluated effects of ropivacaine on LPS-induced phosphorylation/expression of Src, ICAM-1 and caveolin-1 in human lung microvascular endothelial cells (HLMVEC). RESULTS: WT mice treated with LPS alone showed a 49% increase in ELW compared to control animals (p = 0.001), which was attenuated by ropivacaine (p = 0.001). HTV ventilation alone increased measures of ALI even more than LPS, an effect which was not altered by ropivacaine. LPS plus hyperinflation ("double-hit") increased all ALI parameters (ELW, EVPE, permeability index, MPO activity) by 3-4 fold compared to control, which were again decreased by ropivacaine. Western blot analyses of lung homogenates as well as HLMVEC treated in culture with LPS alone showed a reduction in Src activation/expression, as well as ICAM-1 expression and caveolin-1 phosphorylation. In ICAM-1 (-/-) mice, neither addition of LPS to HTV ventilation alone nor ropivacaine had an effect on the development of ALI. CONCLUSIONS: Ropivacaine may be a promising therapeutic agent for treating the cause of pulmonary edema by blocking inflammatory Src signaling, ICAM-1 expression, leukocyte infiltration, and vascular hyperpermeability.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Amidas/farmacología , Anestésicos Locales/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Lesión Pulmonar Aguda/etiología , Animales , Caveolina 1/genética , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Inflamación/tratamiento farmacológico , Inflamación/patología , Molécula 1 de Adhesión Intercelular/genética , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación/efectos de los fármacos , Edema Pulmonar/prevención & control , Ropivacaína , Transducción de Señal/efectos de los fármacos , Lesión Pulmonar Inducida por Ventilación Mecánica/prevención & control , Familia-src Quinasas/metabolismo
8.
Anesthesiology ; 119(4): 901-6, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23867232

RESUMEN

BACKGROUND: Sepsis remains a leading cause of death in intensive care units. There is growing evidence that volatile anesthetics have beneficial immunomodulatory effects on complex inflammation-mediated conditions. The authors investigated the effect of volatile anesthetics on the overall survival of mice in a sepsis model of cecal ligation and puncture (CLP). METHODS: Mice (N = 12 per treatment group) were exposed to anesthetic concentrations of desflurane, isoflurane, and sevoflurane either during induction of sepsis or when the mice showed pronounced symptoms of inflammation. Overall survival, as well as organ function and inflammation was compared with the CLP group without intervention. RESULTS: With desflurane and sevoflurane conditioning (1.2 minimal alveolar concentration for 2 h immediately after induction of CLP) overall survival was improved to 58% and 83%, respectively, compared with 17% in the untreated CLP group. Isoflurane did not significantly affect outcome. Application of sevoflurane 24 h after sepsis induction significantly improved overall survival to 66%. CONCLUSIONS: Administration of the volatile anesthetics desflurane and sevoflurane reduced CLP-induced mortality. Anesthesia may be a critical confounder when comparing study data where different anesthesia protocols were used.


Asunto(s)
Anestésicos por Inhalación/farmacología , Ciego/lesiones , Sepsis/mortalidad , Anestésicos por Inhalación/metabolismo , Animales , Desflurano , Modelos Animales de Enfermedad , Inflamación/complicaciones , Inflamación/metabolismo , Isoflurano/análogos & derivados , Isoflurano/metabolismo , Isoflurano/farmacología , Ligadura , Masculino , Éteres Metílicos/metabolismo , Éteres Metílicos/farmacología , Ratones , Ratones Endogámicos C57BL , Sepsis/complicaciones , Sepsis/metabolismo , Sevoflurano , Heridas Punzantes
9.
Am J Physiol Lung Cell Mol Physiol ; 302(4): L370-9, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22140070

RESUMEN

Lung inflammatory responses in the absence of infection are considered to be one of primary mechanisms of ventilator-induced lung injury. Here, we determined the role of calpain in the pathogenesis of lung inflammation attributable to mechanical ventilation. Male C57BL/6J mice were subjected to high (28 ml/kg) tidal volume ventilation for 2 h in the absence and presence of calpain inhibitor I (10 mg/kg). To address the isoform-specific functions of calpain 1 and calpain 2 during mechanical ventilation, we utilized a liposome-based delivery system to introduce small interfering RNAs targeting each isoform in pulmonary vasculature in vivo. Mechanical ventilation with high tidal volume induced rapid (within minutes) and persistent calpain activation and lung inflammation as evidenced by neutrophil recruitment, production of TNF-α and IL-6, pulmonary vascular hyperpermeability, and lung edema formation. Pharmaceutical calpain inhibition significantly attenuated these inflammatory responses caused by lung hyperinflation. Depletion of calpain 1 or calpain 2 had a protective effect against ventilator-induced lung inflammatory responses. Inhibition of calpain activity by means of siRNA silencing or pharmacological inhibition also reduced endothelial nitric oxide (NO) synthase (NOS-3)-mediated NO production and subsequent ICAM-1 phosphorylation following high tidal volume ventilation. These results suggest that calpain activation mediates early lung inflammation during ventilator-induced lung injury via NOS-3/NO-dependent ICAM-1 phosphorylation and neutrophil recruitment. Inhibition of calpain activation may therefore provide a novel and promising strategy for the prevention and treatment of ventilator-induced lung injury.


Asunto(s)
Calpaína/metabolismo , Inflamación/enzimología , Pulmón/enzimología , Infiltración Neutrófila , Lesión Pulmonar Inducida por Ventilación Mecánica/enzimología , Animales , Calpaína/antagonistas & inhibidores , Calpaína/genética , Activación Enzimática , Técnicas de Silenciamiento del Gen , Glicoproteínas/farmacología , Glicoproteínas/uso terapéutico , Molécula 1 de Adhesión Intercelular/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Interferencia de ARN , Lesión Pulmonar Inducida por Ventilación Mecánica/patología , Lesión Pulmonar Inducida por Ventilación Mecánica/prevención & control , Ventiladores Mecánicos/efectos adversos
10.
J Neuroinflammation ; 9: 272, 2012 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-23253693

RESUMEN

BACKGROUND: Inhalational anesthetics have been shown to influence T cell functions both in vitro and in vivo, in many cases inducing T cell death, suggesting that exposure to these drugs could modify the course of an autoimmune disease. We tested the hypothesis that in mice immunized to develop experimental autoimmune encephalomyelitis (EAE), a well established model of multiple sclerosis (MS), treatment with the commonly used inhalational anesthetic sevoflurane would attenuate disease symptoms. METHODS: C57Bl6 female mice were immunized with myelin oligodendrocyte glycoprotein (MOG) peptide residues 35 to 55 to induce a chronic demyelinating disease. At day 10 after immunization, the mice were subjected to 2 h of 2.5% sevoflurane in 100% oxygen, or 100% oxygen, alone. Following treatment, clinical scores were monitored up to 4 weeks, after which brain histology was performed to measure the effects on astrocyte activation and lymphocyte infiltration. Effects of sevoflurane on T cell activation were studied using splenic T cells isolated from MOG peptide-immunized mice, restimulated ex vivo with MOG peptide or with antibodies to CD3 and CD28, and in the presence of different concentrations of sevoflurane. T cell responses were assessed 1 day later by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay for proliferation, lactate dehydrogenase (LDH) release for cell death, and inflammatory activation by production of interleukin (IL)-17 and interferon (IFN)γ. RESULTS: Clinical scores in the oxygen-treated group increased until day 28 at which time they showed moderate to severe disease (average clinical score of 2.9). In contrast, disease progression in the sevoflurane-treated group increased to 2.1 at day 25, after which it remained unchanged until the end of the study. Immunohistochemical analysis revealed reduced numbers of infiltrating leukocytes and CD4+ cells in the CNS of the sevoflurane-treated mice, as well as reduced glial cell activation. In splenic T cells, low doses of sevoflurane reduced IFNγ production, cell proliferation, and increased LDH release. CONCLUSIONS: These results are the first to show attenuation of EAE disease by an inhaled anesthetic and are consistent with previous reports that inhaled anesthetics, including sevoflurane, can suppress T cell activation that, in the context of autoimmune diseases such as MS, could lead to reduced clinical progression.


Asunto(s)
Encéfalo/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Activación de Linfocitos/efectos de los fármacos , Éteres Metílicos/uso terapéutico , Linfocitos T/efectos de los fármacos , Animales , Encéfalo/inmunología , Encéfalo/patología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Activación de Linfocitos/inmunología , Éteres Metílicos/farmacología , Ratones , Ratones Endogámicos C57BL , Sevoflurano , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/patología , Linfocitos T/inmunología , Linfocitos T/patología
11.
Anesthesiology ; 117(3): 548-59, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22846676

RESUMEN

BACKGROUND: Retrospective analysis of patients undergoing cancer surgery suggests the use of regional anesthesia may reduce cancer recurrence and improve survival. Amide-linked local anesthetics have antiinflammatory properties, although the mechanism of action in this regard is unclear. As inflammatory processes involving Src tyrosine protein kinase and intercellular adhesion molecule-1 are important in tumor growth and metastasis, we hypothesized that amide-linked local anesthetics may inhibit inflammatory Src-signaling involved in migration of adenocarcinoma cells. METHODS: NCI-H838 lung cancer cells were incubated with tumor necrosis factor-α in absence/presence of ropivacaine, lidocaine, or chloroprocaine (1 nM-100 µM). Cell migration and total cell lysate Src-activation and intercellular adhesion molecule-1 phosphorylation were assessed. The role of voltage-gated sodium-channels in the mechanism of local anesthetic effects was also evaluated. RESULTS: Ropivacaine treatment (100 µM) of H838 cells for 20 min decreased basal Src activity by 62% (P=0.003), and both ropivacaine and lidocaine coadministered with tumor necrosis factor-α statistically significantly decreased Src-activation and intercellular adhesion molecule-1 phosphorylation, whereas chloroprocaine had no such effect. Migration of these cells at 4 h was inhibited by 26% (P=0.005) in presence of 1 µM ropivacaine and 21% by 1 µM lidocaine (P=0.004). These effects of ropivacaine and lidocaine were independent of voltage-gated sodium-channel inhibition. CONCLUSIONS: This study indicates that amide-, but not ester-linked, local anesthetics may provide beneficial antimetastatic effects. The observed inhibition of NCI-H838 cell migration by lidocaine and ropivacaine was associated with the inhibition of tumor necrosis factor-α-induced Src-activation and intercellular adhesion molecule-1 phosphorylation, providing the first evidence of a molecular mechanism that appears to be independent of their known role as sodium-channel blockers.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Anestésicos Locales/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Bloqueadores de los Canales de Sodio/farmacología , Familia-src Quinasas/fisiología , Adenocarcinoma/patología , Amidas/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Lidocaína/farmacología , Neoplasias Pulmonares/patología , Metástasis de la Neoplasia/prevención & control , Fosforilación , Procaína/análogos & derivados , Procaína/farmacología , Estudios Retrospectivos , Ropivacaína , Transducción de Señal , Tetrodotoxina/farmacología , Veratridina/farmacología
12.
Am J Physiol Lung Cell Mol Physiol ; 301(2): L197-206, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21571907

RESUMEN

Lung hyperinflation is known to be an important contributing factor in the pathogenesis of ventilator-induced lung injury. Mechanical stretch causes epithelial barrier dysfunction and an increase in alveolar permeability, although the precise mechanisms have not been completely elucidated. p120-catenin is an adherens junction-associated protein that regulates cell-cell adhesion. In this study, we determined the role of p120-catenin in cyclic stretch-induced alveolar epithelial barrier dysfunction. Cultured alveolar epithelial cells (MLE-12) were subjected to uniform cyclic (0.5 Hz) biaxial stretch from 0 to 8 or 20% change in surface area for 0, 1, 2, or 4 h. At the end of the experiments, cells were lysed to determine p120-catenin expression by Western blot analysis. Immunofluorescence staining of p120-catenin and F-actin was performed to assess the integrity of monolayers and interepithelial gap formation. Compared with unstretched control cells, 20% stretch caused a significant loss in p120-catenin expression, which was coupled to interepithelial gap formation. p120-Catenin knockdown with small interfering RNA (siRNA) dose dependently increased stretch-induced gap formation, whereas overexpression of p120-catenin abolished stretch-induced gap formation. Furthermore, pharmacological calpain inhibition or depletion of calpain-1 with a specific siRNA prevented p120-catenin loss and subsequent stretch-induced gap formation. Our findings demonstrate that p120-catenin plays a critical protective role in cyclic stretch-induced alveolar barrier dysfunction, and, thus, maintenance of p120-catenin expression may be a novel therapeutic strategy for the prevention and treatment of ventilator-induced lung injury.


Asunto(s)
Barrera Alveolocapilar/fisiopatología , Calpaína/metabolismo , Cateninas/metabolismo , Alveolos Pulmonares/fisiopatología , Estrés Fisiológico , Animales , Western Blotting , Calpaína/antagonistas & inhibidores , Calpaína/genética , Cateninas/genética , Línea Celular , Relación Dosis-Respuesta a Droga , Células Epiteliales , Epitelio/patología , Epitelio/fisiopatología , Técnica del Anticuerpo Fluorescente , Espacio Intracelular , Ratones , Alveolos Pulmonares/patología , ARN Interferente Pequeño/administración & dosificación , Coloración y Etiquetado , Transfección , Regulación hacia Arriba , Catenina delta
13.
J Proteome Res ; 9(11): 5782-93, 2010 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-20873814

RESUMEN

Fine epitope mapping of monoclonal antibodies (mAbs) to 16 epitopes on human angiotensin I-converting enzyme (ACE) revealed that the epitopes of all mAbs contained putative glycosylation sites. ACE glycosylation is both cell- and tissue-specific and, therefore, the local conformation of ACE produced by different cells could be also unique. The pattern of ACE binding by a set of mAbs to 16 epitopes of human ACE - "conformational fingerprint of ACE" - is the most sensitive marker of ACE conformation and could be cell- and tissue-specific. The recognition of ACEs by mAbs to ACE was estimated using an immune-capture enzymatic plate precipitation assay. Precipitation patterns of soluble recombinant ACE released from Chinese hamster ovary (CHO)-ACE cells was influenced by conditions that alter ACE glycosylation. This pattern was also strongly cell type specific. Patients with sarcoidosis exhibited conformational fingerprints of tissue ACE (lungs and lymph nodes), as well as blood ACE, which were distinct from controls. Conformational fingerprinting of ACE may detect ACE originated from the cells other than endothelial cells in the blood and when combined with elevated blood ACE levels in patients with sarcoidosis may potentially reflect extrapulmonary sarcoidosis involvement (bone marrow, spleen, liver). If proven true, this would serve as a biomarker of enormous potential clinical significance.


Asunto(s)
Peptidil-Dipeptidasa A/química , Sarcoidosis/enzimología , Animales , Anticuerpos Monoclonales , Línea Celular , Mapeo Epitopo/métodos , Epítopos , Glicosilación , Humanos , Peptidil-Dipeptidasa A/inmunología , Conformación Proteica , Distribución Tisular
14.
Microvasc Res ; 80(2): 250-7, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20430040

RESUMEN

Reduced lung capillary expression of angiotensin I-converting enzyme (ACE), a key enzyme in cardiovascular pathophysiology, and of caveolin-1, an important regulator of endothelial cell signalling, has been demonstrated in various models of pulmonary arterial hypertension (PAH). We addressed the relationship between PAH and ACE expression in caveolin-1 knockout mice (Cav1(-/-)), which have moderate PAH. Tissue ACE activity was reduced by 50% in lungs from 3-month-old Cav1(-/-) mice compared to wild type (WT). A similar reduction in lung endothelial ACE expression was observed by measuring the lung uptake of (125)I-labeled monoclonal anti-ACE antibody and by quantitative immunohistochemistry. These alterations in ACE are limited to capillary segments of the pulmonary circulation. Functionally, the increase in pulmonary artery pressure (PAP) in response to ACE conversion of angiotensin I to angiotensin II in isolated, perfused mouse lungs was reduced significantly in Cav1(-/-) mice compared to WT. Thus, these complementary approaches demonstrate the dependence of lung microvascular endothelial cell ACE protein expression on caveolin-1 expression and underscore the vital role of caveolin-1-regulated pulmonary vascular homeostasis on endothelial ACE expression and activity. In summary, we have revealed a novel role of caveolin-1 in the regulation of ACE expression in pulmonary capillary endothelial cells. Further understanding of the mechanism by which reduced caveolin-1 expression leads altered pulmonary vascular development, PAH, and reduced ACE expression may have important clinical implications in patients with these severe lung diseases.


Asunto(s)
Caveolina 1/genética , Hipertensión Pulmonar/enzimología , Pulmón/enzimología , Peptidil-Dipeptidasa A/metabolismo , Angiotensina I/metabolismo , Angiotensina I/farmacología , Angiotensina II/metabolismo , Angiotensina II/farmacología , Animales , Presión Sanguínea , Capilares/enzimología , Capilares/patología , Caveolina 1/metabolismo , Células Endoteliales/enzimología , Células Endoteliales/patología , Regulación Enzimológica de la Expresión Génica , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Inmunohistoquímica , Pulmón/irrigación sanguínea , Pulmón/patología , Ratones , Ratones Noqueados , Perfusión , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/enzimología , Arteria Pulmonar/fisiopatología , Transducción de Señal
15.
Circ Res ; 102(12): e120-31, 2008 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-18511851

RESUMEN

We investigated the role of caveolae in the mechanism of increased pulmonary vascular permeability and edema formation induced by the activation of polymorphonuclear neutrophils (PMNs). We observed that the increase in lung vascular permeability induced by the activation of PMNs required caveolin-1, the caveolae scaffold protein. The permeability increase induced by PMN activation was blocked in caveolin-1 knockout mice and by suppressing caveolin-1 expression in rats. The response was also dependent on Src phosphorylation of caveolin-1 known to activate caveolae-mediated endocytosis in endothelial cells. To address the role of PMN interaction with endothelial cells, we used an intercellular adhesion molecule (ICAM)-1 blocking monoclonal antibody. Preventing the ICAM-1-mediated PMN binding to endothelial cells abrogated Src phosphorylation of caveolin-1, as well as the increase in endothelial permeability. Direct ICAM-1 activation by crosslinking recapitulated these responses, suggesting that ICAM-1 activates caveolin-1 signaling responsible for caveolae-mediated endothelial hyperpermeability. Our results provide support for the novel concept that a large component of pulmonary vascular hyperpermeability induced by activation of PMNs adherent to the vessel wall is dependent on signaling via caveolin-1 and increased caveolae-mediated transcytosis. Thus, it is important to consider the role of the transendothelial vesicular permeability pathway that contributes to edema formation in developing therapeutic interventions against PMN-mediated inflammatory diseases such as acute lung injury.


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Caveolas/fisiología , Caveolina 1/fisiología , Células Endoteliales/efectos de los fármacos , Molécula 1 de Adhesión Intercelular/fisiología , Neutrófilos/fisiología , Animales , Anticuerpos Monoclonales/farmacología , Caveolina 1/antagonistas & inhibidores , Caveolina 1/biosíntesis , Caveolina 1/deficiencia , Caveolina 1/genética , Adhesión Celular/fisiología , Células Cultivadas/citología , Células Cultivadas/efectos de los fármacos , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Células Endoteliales/citología , Pulmón/irrigación sanguínea , Ratones , Ratones Noqueados , Neutrófilos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Proto-Oncogénicas pp60(c-src)/fisiología , Edema Pulmonar/fisiopatología , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Albúmina Sérica/farmacocinética , beta-Ciclodextrinas/farmacología
16.
J Cardiovasc Pharmacol Ther ; 22(4): 374-386, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28587581

RESUMEN

BACKGROUND: Angiotensin-converting enzyme (ACE) inhibitors (ACEI) are widely used in the management of cardiovascular diseases but with significant interindividual variability in the patient's response. OBJECTIVES: To investigate whether interindividual variability in the response to ACE inhibitors is explained by the "ACE phenotype"-for example, variability in plasma ACE concentration, activity, and conformation and/or the degree of ACE inhibition in each individual. METHODS: The ACE phenotype was determined in plasma of 14 patients with hypertension treated chronically for 4 weeks with 40 mg enalapril (E) or 20 mg E + 16 mg candesartan (EC) and in 20 patients with hypertension treated acutely with a single dose (20 mg) of E with or without pretreatment with hydrochlorothiazide. The ACE phenotyping included (1) plasma ACE concentration; (2) ACE activity (with 2 substrates: Hip-His-Leu and Z-Phe-His-Leu and calculation of their ratio); (3) detection of ACE inhibitors in patient's blood (indicator of patient compliance) and the degree of ACE inhibition (ie, adherence); and (4) ACE conformation. RESULTS: Enalapril reduced systolic and diastolic blood pressure in most patients; however, 20% of patients were considered nonresponders. Chronic treatment results in 40% increase in serum ACE concentrations, with the exception of 1 patient. There was a trend toward better response to ACEI among patients who had a higher plasma ACE concentration. CONCLUSION: Due to the fact that "20% of patients do not respond to ACEI by blood pressure drop," the initial blood ACE level could not be a predictor of blood pressure reduction in an individual patient. However, ACE phenotyping provides important information about conformational and kinetic changes in ACE of individual patients, and this could be a reason for resistance to ACE inhibitors in some nonresponders.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Antihipertensivos/uso terapéutico , Presión Sanguínea/efectos de los fármacos , Enalapril/uso terapéutico , Hipertensión/tratamiento farmacológico , Peptidil-Dipeptidasa A/genética , Variantes Farmacogenómicas , Medicina de Precisión , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/efectos adversos , Inhibidores de la Enzima Convertidora de Angiotensina/farmacocinética , Antihipertensivos/efectos adversos , Antihipertensivos/farmacocinética , Bencimidazoles/uso terapéutico , Compuestos de Bifenilo , Estudios Cruzados , Método Doble Ciego , Monitoreo de Drogas , Resistencia a Medicamentos/genética , Quimioterapia Combinada , Enalapril/efectos adversos , Enalapril/farmacocinética , Genotipo , Humanos , Hidroclorotiazida/uso terapéutico , Hipertensión/enzimología , Hipertensión/genética , Hipertensión/fisiopatología , Selección de Paciente , Peptidil-Dipeptidasa A/metabolismo , Farmacogenética , Pruebas de Farmacogenómica , Fenotipo , Valor Predictivo de las Pruebas , Inhibidores de los Simportadores del Cloruro de Sodio/uso terapéutico , Tetrazoles/uso terapéutico , Resultado del Tratamiento
17.
PLoS One ; 12(7): e0180213, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28671983

RESUMEN

A patient's recovery from lung inflammatory injury or development of multi-system organ failure is determined by the host's ability to resolve inflammation and repair tissue damage, both of which require the clearance of apoptotic neutrophils by macrophages (efferocytosis). Here, we investigated the effects of isoflurane on macrophage efferocytosis and resolution of lung inflammatory injury. Treatment of murine bone marrow-derived macrophages (BMDMs) or alveolar macrophages with isoflurane dramatically enhanced phagocytosis of apoptotic neutrophils. Isoflurane significantly increased the surface expression of the receptor tyrosine kinase Mer in macrophages, but markedly decreased the levels of a soluble form of Mer protein in the medium. Isoflurane treatment also caused a decrease in a disintegrin and metalloproteinase 17 (ADAM17) on the cell surface and a concomitant increase in its cytoplasmic fraction. These responses induced by isoflurane were completely reversed by a pharmacological inhibitor or genetic deletion of AMP-activated protein kinase (AMPK). In a mouse model of lipopolysaccharide-induced lung injury, isoflurane accelerated the recovery of lung inflammation and injury that was coupled with an increase in the number of alveolar macrophages containing apoptotic bodies. In alveolar macrophage-depleted mice, administration of isoflurane-pretreated BMDMs facilitated resolution of lung inflammation following lipopolysaccharide challenge. Thus, isoflurane promoted resolution of lipopolysaccharide-induced lung inflammatory injury via enhancement of macrophage efferocytosis. Increased macrophage efferocytosis following isoflurane treatment correlates with upregulation of Mer surface expression through AMPK-mediated blockade of ADAM17 trafficking to the cell membrane.


Asunto(s)
Proteína ADAM17/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Antígenos de Diferenciación de Linfocitos B/metabolismo , Apoptosis , Isoflurano/farmacología , Neutrófilos/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Receptores Inmunológicos/metabolismo , Transducción de Señal , Proteínas Quinasas Activadas por AMP/genética , Lesión Pulmonar Aguda/inducido químicamente , Animales , Células Cultivadas , Técnicas de Silenciamiento del Gen , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Neutrófilos/inmunología
18.
Curr Med Chem ; 23(15): 1571-96, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27048377

RESUMEN

The calcium binding protein S100B has attracted great attention as a biomarker for a variety of diseases. S100B is mainly expressed in glial cells and functions through intracellular and extracellular signaling pathways. The biological roles of S100B have been closely associated with its concentrations and its physiological states. The released S100B can bind to the receptor of advanced glycation end products and induce the initiation of multiple cell signaling transductions. The regulation of S100B bioactivities has been suggested through phosphoinositide 3 kinase/Akt, p53, mitogen-activated protein kinases, transcriptional factors including nuclear factor-kappaB, and cyclic adenosine monophosphate. The levels of S100B in the blood may function to predict the progress or the prognosis of many kinds of diseases, such as cerebrovascular diseases, neurodegenerative diseases, motor neuron diseases, traumatic brain injury, schizophrenia, depression, diabetes mellitus, myocardial infarction, cancer, and infectious diseases. Given that the activity of S100B has been implicated in the pathological process of these diseases, S100B should not be simply regarded as a biomarker, it may also function as therapeutic target for these diseases. Further elucidation of the roles of S100B may formulate innovative therapeutic strategies for multiple diseases.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Enfermedad , Subunidad beta de la Proteína de Unión al Calcio S100/antagonistas & inhibidores , Animales , Biomarcadores/análisis , Biomarcadores/metabolismo , Humanos , Subunidad beta de la Proteína de Unión al Calcio S100/análisis , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo
19.
Sci Rep ; 6: 34913, 2016 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-27734897

RESUMEN

Angiotensin I-converting enzyme (ACE) hydrolyzes numerous peptides and is a critical participant in blood pressure regulation and vascular remodeling. Elevated tissue ACE levels are associated with increased risk for cardiovascular and respiratory disorders. Blood ACE concentrations are determined by proteolytic cleavage of ACE from the endothelial cell surface, a process that remains incompletely understood. In this study, we identified a novel ACE gene mutation (Arg532Trp substitution in the N domain of somatic ACE) that increases blood ACE activity 7-fold and interrogated the mechanism by which this mutation significantly increases blood ACE levels. We hypothesized that this ACE mutation disrupts the binding site for blood components which may stabilize ACE conformation and diminish ACE shedding. We identified the ACE-binding protein in the blood as lysozyme and also a Low Molecular Weight (LMW) ACE effector, bilirubin, which act in concert to regulate ACE conformation and thereby influence ACE shedding. These results provide mechanistic insight into the elevated blood level of ACE observed in patients on ACE inhibitor therapy and elevated blood lysozyme and ACE levels in sarcoidosis patients.


Asunto(s)
Bilirrubina/química , Muramidasa/química , Peptidil-Dipeptidasa A/química , Animales , Anticuerpos Monoclonales/química , Células CHO , Estudios de Casos y Controles , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Citometría de Flujo , Humanos , Péptidos y Proteínas de Señalización Intercelular , Ratones , Mutación , Péptidos/química , Fenotipo , Unión Proteica , Dominios Proteicos , Proteína C Asociada a Surfactante Pulmonar , Sarcoidosis/sangre , Resonancia por Plasmón de Superficie
20.
Sci Rep ; 5: 13457, 2015 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-26307438

RESUMEN

We have demonstrated a printed electronic tag that monitors time-integrated sensor signals and writes to nonvolatile memories for later readout. The tag is additively fabricated on flexible plastic foil and comprises a thermistor divider, complementary organic circuits, and two nonvolatile memory cells. With a supply voltage below 30 V, the threshold temperatures can be tuned between 0 °C and 80 °C. The time-temperature dose measurement is calibrated for minute-scale integration. The two memory bits are sequentially written in a thermometer code to provide an accumulated dose record.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA