Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
FASEB J ; 35(6): e21675, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34038004

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease associated with mitochondrial oxidative stress. Mitochondrial reactive oxygen species (mtROS) are important for cell homeostasis by regulating mitochondrial dynamics. Here, we show that IPF BAL cells exhibited increased mitochondrial biogenesis that is, in part, due to increased nuclear expression of peroxisome proliferator-activated receptor-É£ (PPARÉ£) coactivator (PGC)-1α. Increased PPARGC1A mRNA expression directly correlated with reduced pulmonary function in IPF subjects. Oxidant-mediated activation of the p38 MAPK via Akt1 regulated PGC-1α activation to increase mitochondrial biogenesis in monocyte-derived macrophages. Demonstrating the importance of PGC-1α in fibrotic repair, mice harboring a conditional deletion of Ppargc1a in monocyte-derived macrophages or mice administered a chemical inhibitor of mitochondrial division had reduced biogenesis and increased apoptosis, and the mice were protected from pulmonary fibrosis. These observations suggest that Akt1-mediated regulation of PGC-1α maintains mitochondrial homeostasis in monocyte-derived macrophages to induce apoptosis resistance, which contributes to the pathogenesis of pulmonary fibrosis.


Asunto(s)
Macrófagos Alveolares/patología , Mitocondrias/patología , Dinámicas Mitocondriales , Estrés Oxidativo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Procesamiento Proteico-Postraduccional , Fibrosis Pulmonar/patología , Adolescente , Adulto , Anciano , Animales , Apoptosis , Femenino , Homeostasis , Humanos , Macrófagos Alveolares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Mitocondrias/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/química , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Fosforilación , Fibrosis Pulmonar/etiología , Fibrosis Pulmonar/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Adulto Joven
2.
Respir Res ; 21(1): 83, 2020 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-32293449

RESUMEN

BACKGROUND: The outbreak of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in China has been declared a public health emergency of international concern. The cardiac injury is a common condition among the hospitalized patients with COVID-19. However, whether N terminal pro B type natriuretic peptide (NT-proBNP) predicted outcome of severe COVID-19 patients was unknown. METHODS: The study initially enrolled 102 patients with severe COVID-19 from a continuous sample. After screening out the ineligible cases, 54 patients were analyzed in this study. The primary outcome was in-hospital death defined as the case fatality rate. Research information and following-up data were obtained from their medical records. RESULTS: The best cut-off value of NT-proBNP for predicting in-hospital death was 88.64 pg/mL with the sensitivity for 100% and the specificity for 66.67%. Patients with high NT-proBNP values (> 88.64 pg/mL) had a significantly increased risk of death during the days of following-up compared with those with low values (≤88.64 pg/mL). After adjustment for potential risk factors, NT-proBNP was independently correlated with in-hospital death. CONCLUSION: NT-proBNP might be an independent risk factor for in-hospital death in patients with severe COVID-19. TRIAL REGISTRATION: ClinicalTrials, NCT04292964. Registered 03 March 2020.


Asunto(s)
Infecciones por Coronavirus , Mortalidad Hospitalaria , Péptido Natriurético Encefálico/análisis , Pandemias , Fragmentos de Péptidos/análisis , Neumonía Viral , Adulto , Anciano , Betacoronavirus , COVID-19 , Prueba de COVID-19 , Técnicas de Laboratorio Clínico , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/mortalidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mortalidad , Neumonía Viral/diagnóstico , Neumonía Viral/mortalidad , Valor Predictivo de las Pruebas , Pronóstico , Valores de Referencia , Estudios Retrospectivos , Factores de Riesgo , SARS-CoV-2
3.
FASEB J ; 34(2): 3305-3317, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31916311

RESUMEN

Pulmonary edema associated with increased vascular permeability is a severe complication of Pseudomonas (P.) aeruginosa-induced acute lung injury. The mechanisms underlying P aeruginosa-induced vascular permeability are not well understood. In the present study, we investigated the role of neuronal Wiskott Aldrich syndrome protein (N-WASP) in modulating P aeruginosa-induced vascular permeability. Using lung microvascular endothelial and alveolar epithelial cells, we demonstrated that N-WASP downregulation attenuated P aeruginosa-induced actin stress fiber formation and prevented paracellular permeability. P aeruginosa-induced dissociation between VE-cadherin and ß-catenin, but increased association between N-WASP and VE-cadherin, suggesting a role for N-WASP in promoting P aeruginosa-induced adherens junction rupture. P aeruginosa increased N-WASP-Y256 phosphorylation, which required the activation of Rho GTPase and focal adhesion kinase. Increased N-WASP-Y256 phosphorylation promotes N-WASP and integrin αVß6 association as well as TGF-ß-mediated permeability across alveolar epithelial cells. Inhibition of N-WASP-Y256 phosphorylation by N-WASP-Y256F overexpression blocked N-WASP effects in P aeruginosa-induced actin stress fiber formation and increased paracellular permeability. In vivo, N-WASP knockdown attenuated the development of pulmonary edema and improved survival in a mouse model of P aeruginosa pneumonia. Together, our data demonstrate that N-WASP plays an essential role in P aeruginosa-induced vascular permeability and pulmonary edema through the modulation of actin cytoskeleton dynamics.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Permeabilidad Capilar , Pulmón/metabolismo , Neumonía/metabolismo , Infecciones por Pseudomonas/metabolismo , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Uniones Adherentes/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Cadherinas/metabolismo , Células Cultivadas , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Integrinas/metabolismo , Pulmón/microbiología , Ratones , Pseudomonas aeruginosa/patogenicidad , Ratas , Factor de Crecimiento Transformador beta/metabolismo , Proteína del Síndrome de Wiskott-Aldrich/genética , beta Catenina/metabolismo , Proteínas de Unión al GTP rho/metabolismo
4.
J Clin Invest ; 129(11): 4962-4978, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31609245

RESUMEN

Macrophages are important in mounting an innate immune response to injury as well as in repair of injury. Gene expression of Rho proteins is known to be increased in fibrotic models; however, the role of these proteins in idiopathic pulmonary fibrosis (IPF) is not known. Here, we show that BAL cells from patients with IPF have a profibrotic phenotype secondary to increased activation of the small GTPase Rac1. Rac1 activation requires a posttranslational modification, geranylgeranylation, of the C-terminal cysteine residue. We found that by supplying more substrate for geranylgeranylation, Rac1 activation was substantially increased, resulting in profibrotic polarization by increasing flux through the mevalonate pathway. The increased flux was secondary to greater levels of acetyl-CoA from metabolic reprogramming to ß oxidation. The polarization mediated fibrotic repair in the absence of injury by enhancing macrophage/fibroblast signaling. These observations suggest that targeting the mevalonate pathway may abrogate the role of macrophages in dysregulated fibrotic repair.


Asunto(s)
Fibrosis Pulmonar Idiopática/metabolismo , Macrófagos/metabolismo , Ácido Mevalónico/metabolismo , Acetilcoenzima A/genética , Acetilcoenzima A/metabolismo , Adolescente , Adulto , Anciano , Animales , Femenino , Humanos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/patología , Macrófagos/patología , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Neuropéptidos/genética , Neuropéptidos/metabolismo , Oxidación-Reducción , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
5.
FASEB J ; 30(7): 2557-69, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27025963

RESUMEN

TGF-ß1 induces an increase in paracellular permeability and actin stress fiber formation in lung microvascular endothelial and alveolar epithelial cells via small Rho GTPase. The molecular mechanism involved is not fully understood. Neuronal Wiskott-Aldrich syndrome protein (N-WASP) has an essential role in actin structure dynamics. We hypothesized that N-WASP plays a critical role in these TGF-ß1-induced responses. In these cell monolayers, we demonstrated that N-WASP down-regulation by short hairpin RNA prevented TGF-ß1-mediated disruption of the cortical actin structure, actin stress filament formation, and increased permeability. Furthermore, N-WASP down-regulation blocked TGF-ß1 activation mediated by IL-1ß in alveolar epithelial cells, which requires actin stress fiber formation. Control short hairpin RNA had no effect on these TGF-ß1-induced responses. TGF-ß1-induced phosphorylation of Y256 of N-WASP via activation of small Rho GTPase and focal adhesion kinase mediates TGF-ß1-induced paracellular permeability and actin cytoskeleton dynamics. In vivo, compared with controls, N-WASP down-regulation increases survival and prevents lung edema in mice induced by bleomycin exposure-a lung injury model in which TGF-ß1 plays a critical role. Our data indicate that N-WASP plays a crucial role in the development of TGF-ß1-mediated acute lung injury by promoting pulmonary edema via regulation of actin cytoskeleton dynamics.-Wagener, B. M., Hu, M., Zheng, A., Zhao, X., Che, P., Brandon, A., Anjum, N., Snapper, S., Creighton, J., Guan, J.-L., Han, Q., Cai, G.-Q., Han, X., Pittet, J.-F., Ding, Q. Neuronal Wiskott-Aldrich syndrome protein regulates TGF-ß1-mediated lung vascular permeability.


Asunto(s)
Permeabilidad Capilar/fisiología , Células Endoteliales/fisiología , Pulmón/irrigación sanguínea , Factor de Crecimiento Transformador beta1/metabolismo , Proteína Neuronal del Síndrome de Wiskott-Aldrich/metabolismo , Animales , Bleomicina/toxicidad , Células Cultivadas , Regulación de la Expresión Génica/fisiología , Lesión Pulmonar/inducido químicamente , Ratones , Neuronas , Ratas , Factor de Crecimiento Transformador beta1/genética , Proteína Neuronal del Síndrome de Wiskott-Aldrich/genética
6.
Sci Rep ; 5: 8453, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25677816

RESUMEN

S100A4 expression is associated with poor clinical outcomes of patients with pancreatic cancer. The effects of loss or gain of S100A4 were examined in pancreatic cancer cell lines. S100A4 downregulation remarkably reduces cell migration and invasion, inhibits proliferation, and induces apoptosis in pancreatic tumor cells. S100A4 downregulation results in significant cell growth inhibition and apoptosis in response to TGF-ß1, supporting a non-canonical role of S100A4 in pancreatic cancer. The role of S100A4 in tumor progression was studied by using an orthotopic human pancreatic cancer xenograft mouse model. Tumor mass is remarkably decreased in animals injected with S100A4-deficient pancreatic tumor cells. P27(Kip1) expression and cleaved caspase-3 are increased, while cyclin E expression is decreased, in S100A4-deficient pancreatic tumors in vivo. S100A4-deficient tumors have lower expression of vascular endothelial growth factor, suggesting reduced angiogenesis. Biochemical assays revealed that S100A4 activates Src and focal adhesion kinase (FAK) signaling events, and inhibition of both kinases is required to maximally block the tumorigenic potential of pancreatic cancer cells. These findings support that S100A4 plays an important role in pancreatic cancer progression in vivo and S100A4 promotes tumorigenic phenotypes of pancreatic cancer cells through the Src-FAK mediated dual signaling pathway.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Proteínas S100/metabolismo , Familia-src Quinasas/metabolismo , Animales , Caspasa 3/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ciclina E/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación hacia Abajo , Femenino , Humanos , Ratones , Ratones SCID , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Proteína de Unión al Calcio S100A4 , Proteínas S100/antagonistas & inhibidores , Proteínas S100/genética , Transducción de Señal , Factor de Crecimiento Transformador beta1/farmacología , Trasplante Heterólogo , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
J Pharmacol Exp Ther ; 351(1): 87-95, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25047515

RESUMEN

Myofibroblasts are effector cells in fibrotic disorders that synthesize and remodel the extracellular matrix (ECM). This study investigated the role of the Src kinase pathway in myofibroblast activation in vitro and fibrogenesis in vivo. The profibrotic cytokine, transforming growth factor ß1 (TGF-ß1), induced rapid activation of Src kinase, which led to myofibroblast differentiation of human lung fibroblasts. The Src kinase inhibitor AZD0530 (saracatinib) blocked TGF-ß1-induced Src kinase activation in a dose-dependent manner. Inhibition of Src kinase significantly reduced α-smooth muscle actin (α-SMA) expression, a marker of myofibroblast differentiation, in TGF-ß1-treated lung fibroblasts. In addition, the induced expression of collagen and fibronectin and three-dimensional collagen gel contraction were also significantly inhibited in AZD0530-treated fibroblasts. The therapeutic efficiency of Src kinase inhibition in vivo was tested in the bleomycin murine lung fibrosis model. Src kinase activation and collagen accumulation were significantly reduced in the lungs of AZD0530-treated mice when compared with controls. Furthermore, the total fibrotic area and expression of α-SMA and ECM proteins were significantly decreased in lungs of AZD0530-treated mice. These results indicate that Src kinase promotes myofibroblast differentiation and activation of lung fibroblasts. Additionally, these studies provide proof-of-concept for targeting the noncanonical TGF-ß signaling pathway involving Src kinase as an effective therapeutic strategy for lung fibrosis.


Asunto(s)
Benzodioxoles/farmacología , Diferenciación Celular , Inhibidores Enzimáticos/farmacología , Miofibroblastos/efectos de los fármacos , Fibrosis Pulmonar/tratamiento farmacológico , Quinazolinas/farmacología , Familia-src Quinasas/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Benzodioxoles/uso terapéutico , Línea Celular , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Inhibidores Enzimáticos/uso terapéutico , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Miofibroblastos/citología , Miofibroblastos/enzimología , Quinazolinas/uso terapéutico , Factor de Crecimiento Transformador beta/farmacología , Familia-src Quinasas/antagonistas & inhibidores
8.
Am J Pathol ; 182(5): 1572-84, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23499373

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease whose underlying molecular mechanisms are largely unknown. Herein, we show that focal adhesion kinase-related nonkinase (FRNK) plays a key role in limiting the development of lung fibrosis. Loss of FRNK function in vivo leads to increased lung fibrosis in an experimental mouse model. The increase in lung fibrosis is confirmed at the histological, biochemical, and physiological levels. Concordantly, loss of FRNK function results in increased fibroblast migration and myofibroblast differentiation and activation of signaling proteins that drive these phenotypes. FRNK-deficient murine lung fibroblasts also have an increased capacity to produce and contract matrix proteins. Restoration of FRNK expression in vivo and in vitro reverses these profibrotic phenotypes. These data demonstrate the multiple antifibrotic actions of FRNK. More important, FRNK expression is down-regulated in human IPF, and down-regulation of FRNK in normal human lung fibroblasts recapitulates the profibrotic phenotype seen in FRNK-deficient cells. The effect of loss and gain of FRNK in the experimental model, when taken together with its down-regulation in human IPF, suggests that FRNK acts as an endogenous negative regulator of lung fibrosis by repressing multiple profibrotic responses.


Asunto(s)
Proteínas Tirosina Quinasas/metabolismo , Fibrosis Pulmonar/enzimología , Fibrosis Pulmonar/patología , Adulto , Animales , Bleomicina , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Pulmón/efectos de los fármacos , Pulmón/enzimología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Miofibroblastos/efectos de los fármacos , Miofibroblastos/enzimología , Miofibroblastos/patología , Proteínas Tirosina Quinasas/deficiencia , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/farmacología
9.
Am J Physiol Lung Cell Mol Physiol ; 303(8): L692-702, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22886502

RESUMEN

Myofibroblasts are implicated in pathological stromal responses associated with lung fibrosis. One prominent phenotypic marker of fully differentiated myofibroblasts is the polymerized, thick cytoplasmic filaments containing newly synthesized α-smooth muscle actin (α-SMA). These α-SMA-containing cytoplasmic filaments are important for myofibroblast contractility during tissue remodeling. However, the molecular mechanisms regulating the formation and maturation of α-SMA-containing filaments have not been defined. This study demonstrates a critical role for neuronal Wiskott-Aldrich syndrome protein (N-WASP) in regulating the formation of α-SMA-containing cytoplasmic filaments during myofibroblast differentiation and in myofibroblast contractility. Focal adhesion kinase (FAK) is activated by transforming growth factor-ß1 (TGF-ß1) and is required for phosphorylation of tyrosine residue 256 (Y256) of N-WASP. Phosphorylation of Y256 of N-WASP is essential for TGF-ß1-induced formation of α-SMA-containing cytoplasmic filaments in primary human lung fibroblasts. In addition, we demonstrate that actin-related protein (Arp) 2/3 complex is downstream of N-WASP and mediates the maturation of α-SMA-containing cytoplasmic filaments. Together, this study supports a critical role of N-WASP in integrating FAK and Arp2/3 signaling to mediate formation of α-SMA-containing cytoplasmic filaments during myofibroblast differentiation and maturation.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Fibroblastos/metabolismo , Fibrosis Pulmonar/metabolismo , Proteína Neuronal del Síndrome de Wiskott-Aldrich/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Proteína 3 Relacionada con la Actina/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Colágeno/metabolismo , Citoplasma/metabolismo , Fibroblastos/citología , Quinasa 1 de Adhesión Focal/metabolismo , Pulmón/citología , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Cultivo Primario de Células , Fibrosis Pulmonar/patología , ARN Interferente Pequeño/genética , Factor de Crecimiento Transformador beta1/farmacología , Tirosina/metabolismo , Proteína Neuronal del Síndrome de Wiskott-Aldrich/genética
10.
Biochim Biophys Acta ; 1812(4): 565-71, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21094253

RESUMEN

Recent studies recognize that Hypocretin system (also known as Orexin) plays a critical role in sleep/wake disorders and feeding behaviors. However, little is known about the regulation of the Hypocretin system. It is also known that tumor necrosis factor alpha (TNF-α) is involved in the regulation of sleep/wake cycle. Here, we test our hypothesis that the Hypocretin system is regulated by TNF-α. Prepro-Hypocretin and Hypocretin receptor 2 (HcrtR2) can be detected at a very low level in rat B35 neuroblastoma cells. In response to TNF-α, Prepro-Hypocretin mRNA and protein levels are down-regulated, and also HcrtR2 protein level is down-regulated in B35 cells. To investigate the mechanism, exogenous rat Prepro-Hypocretin and rat HcrtR2 were overexpressed in B35 cells. In response to TNF-α, protein and mRNA of Prepro-Hypocretin are significantly decreased (by 93% and 94%, respectively), and the half-life of Prepro-Hypocretin mRNA is decreased in a time- and dose-dependent manner. The level of HcrtR2 mRNA level is not affected by TNF-α treatment; however, HcrtR2 protein level is significantly decreased (by 86%) through ubiquitination in B35 cells treated with TNF-α. Downregulation of cellular inhibitor of apoptosis protein-1 and -2 (cIAP-1 and -2) abrogates the HcrtR2 ubiquitination induced by TNF-α. The control green fluorescent protein (GFP) expression is not affected by TNF-α treatment. These studies demonstrate that TNF-α can impair the function of the Hypocretin system by reducing the levels of both Prepro-Hypocretin and HcrtR2.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neuropéptidos/metabolismo , Estabilidad del ARN , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitinación , Animales , Línea Celular Tumoral , Regulación hacia Abajo , Péptidos y Proteínas de Señalización Intracelular/genética , Neuropéptidos/genética , Receptores de Orexina , Orexinas , ARN Mensajero/genética , Ratas , Receptores Acoplados a Proteínas G/genética , Receptores de Neuropéptido/genética , Trastornos del Sueño-Vigilia/etiología
11.
Exp Cell Res ; 316(9): 1600-9, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20109444

RESUMEN

Fibroblast migration plays an important role in the normal wound healing process; however, dysregulated cell migration may contribute to the progressive formation of fibrotic lesions in the diseased condition. To examine the role of focal-adhesion-kinase (FAK)-related non-kinase (FRNK) in regulation of fibrotic lung fibroblast migration, we examined cell migration, FRNK expression, and activation of focal adhesion kinase (FAK) and Rho GTPase (Rho and Rac) in primary lung fibroblasts derived from both idiopathic pulmonary fibrosis (IPF) patients and normal human controls. Fibrotic (IPF) lung fibroblasts have increased cell migration when compared to control human lung fibroblasts. FRNK expression is significantly reduced in IPF lung fibroblasts, while activation of FAK, Rho and Rac is increased in IPF lung fibroblasts. Endogenous FRNK expression is inversely correlated with FAK activation and cell migration rate in IPF lung fibroblasts. Forced exogenous FRNK expression abrogates the increased cell migration, and blocked the activation of FAK and Rho GTPase (Rho and Rac), in IPF lung fibroblasts. These data for the first time provide evidence that downregulation of endogenous FRNK plays a role in promoting cell migration through FAK and Rho GTPase in fibrotic IPF lung fibroblasts.


Asunto(s)
Movimiento Celular , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Western Blotting , Adhesión Celular , Células Cultivadas , Regulación hacia Abajo , Fibroblastos/metabolismo , Fibroblastos/patología , Técnica del Anticuerpo Fluorescente , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/genética , Pulmón/metabolismo , Pulmón/patología , Fenotipo , Proteínas Tirosina Quinasas/genética , Transducción de Señal , Proteínas de Unión al GTP rho/metabolismo
12.
J Neurosci ; 29(50): 15836-45, 2009 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-20016099

RESUMEN

The network oscillation and synaptic plasticity are known to be regulated by GABAergic inhibition, but how they are affected by changes in the GABA transporter activity remains unclear. Here we show that in the CA1 region of mouse hippocampus, pharmacological blockade or genetic deletion of GABA transporter-1 (GAT1) specifically impaired long-term potentiation (LTP) induced by theta burst stimulation, but had no effect on LTP induced by high-frequency stimulation or long-term depression induced by low-frequency stimulation. The extent of LTP impairment depended on the precise burst frequency, with significant impairment at 3-7 Hz that correlated with the time course of elevated GABAergic inhibition caused by GAT1 disruption. Furthermore, in vivo electrophysiological recordings showed that GAT1 gene deletion reduced the frequency of hippocampal theta oscillation. Moreover, behavioral studies showed that GAT1 knock-out mice also exhibited impaired hippocampus-dependent learning and memory. Together, these results have highlighted the important link between GABAergic inhibition and hippocampal theta oscillation, both of which are critical for synaptic plasticity and learning behaviors.


Asunto(s)
Proteínas Transportadoras de GABA en la Membrana Plasmática/fisiología , Hipocampo/fisiología , Potenciación a Largo Plazo/fisiología , Ritmo Teta , Animales , Conducta Animal/fisiología , Proteínas Transportadoras de GABA en la Membrana Plasmática/deficiencia , Proteínas Transportadoras de GABA en la Membrana Plasmática/genética , Hipocampo/patología , Técnicas In Vitro , Aprendizaje/fisiología , Potenciación a Largo Plazo/genética , Masculino , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Plasticidad Neuronal/genética , Plasticidad Neuronal/fisiología
13.
J Neurosci Res ; 86(2): 465-70, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17918738

RESUMEN

gamma-Aminobutyric acid (GABA) transporters play a key role in the regulation of GABA neurotransmission. We reported previously that overexpression of the GABA transporter subtype 1 (GAT1), the major form of the GABA transporter in the CNS, led to hyperalgesia in mice. In the present study, nociceptive responses of GAT1-knockout mice (GAT1(-/-)) were compared with those of heterozygous (GAT(+/-)) and wild-type (GAT(+/+)) mice by four conventional pain models (tail-immersion test, hot-plate test, acetic acid-induced abdominal constriction test, and formalin test). In addition, the analgesic effects of two GAT1-selective inhibitors, NO-711 and tiagabine, were examined in all three genotypes using the same four models. Our data demonstrated that GAT1 deficiency because of genetic knockout or acute blockade by selective inhibitors leads to hypoalgesia in mice. These results confirmed the crucial role of GAT1 in the regulation of nociceptive threshold and suggested that GAT1 inhibitors have the potential for clinical use in pain therapy.


Asunto(s)
Proteínas Transportadoras de GABA en la Membrana Plasmática/metabolismo , Umbral del Dolor/fisiología , Dolor/fisiopatología , Animales , Antagonistas del GABA/farmacología , Proteínas Transportadoras de GABA en la Membrana Plasmática/efectos de los fármacos , Proteínas Transportadoras de GABA en la Membrana Plasmática/genética , Ratones , Ratones Noqueados , Ácidos Nipecóticos/farmacología , Oximas/farmacología , Umbral del Dolor/efectos de los fármacos , Tiagabina
14.
J Neurosci Res ; 85(3): 649-55, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17183588

RESUMEN

Dysregulation of the brain GABAergic system has been implicated in the pathophysiology of violence and aggression. As a key regulator of central GABAergic activity, dysfunction of the GABA transporter subtype 1 (GAT1) represents a potential mechanism mediating pathologic aggression. We provide evidence that GAT1-/- mice and GAT1+/- mice exhibit lower aggressive behavior both in home cage resident-intruder test and neutral arena resident-intruder test, compared to wild-type mice (GAT1+/+). The pharmacologic effects of the GAT1 inhibitor, tiagabine and the GABA(A) receptor antagonist, bicuculline have been assessed in GAT1+/+ mice: tiagabine inhibits attacks but bicuculline induces attacks. Compared to GAT1+/- and +/+ mice, the GAT1-/- mice displayed a normal circadian pattern of home cage activity, but more activity overall. Meanwhile, reduced testosterone concentration was found in GAT1-/- mice compared to GAT1+/+ mice but not in GAT1+/+ mice treated with tiagabine, suggesting that testosterone is not directly involved in GAT1 mediated aggressive behavior regulation. These results showed that GAT1 is an important target involved in the regulation of aggressive behavior in mice, and long-term dysfunction of GAT1 may also result in the alteration of testosterone secretion.


Asunto(s)
Agresión/fisiología , Proteínas Transportadoras de GABA en la Membrana Plasmática/deficiencia , Animales , Proteínas Transportadoras de GABA en la Membrana Plasmática/genética , Proteínas Transportadoras de GABA en la Membrana Plasmática/fisiología , Vivienda para Animales , Masculino , Ratones , Ratones Noqueados , Actividad Motora/fisiología , Conducta Social , Testosterona/sangre
15.
Neuropsychopharmacology ; 32(7): 1531-9, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17164814

RESUMEN

Gamma-aminobutyric acid (GABA) transporter subtype 1 (GAT1), which transports extracellular GABA into presynaptic neurons, plays an important regulatory role in the function of GABAergic systems. However, the contributions of the GAT1 in regulating mental status are not fully understood. In this paper, we observed the behavioral alterations of GAT1 knockout (GAT1(-/-)) mice using several depression- and anxiety-related models (eg, the forced-swimming test and the tail-suspension test for testing depression-related behaviors; the open-field test, the dark-light exploration test, the emergence test, and the elevated plus maze (EPM) test for anxiety-related behaviors). Here we found that GAT1(-/-) mice showed a lower level of depression- and anxiety-like behaviors in comparison to wild-type mice. Furthermore, GAT1(-/-) mice exhibited measurable insensitivity to selected antidepressants and anxiolytics such as fluoxetine, amitriptyline, buspirone, diazepam, and tiagabine in the tail-suspension test and/or the EPM test. Moreover, the basal level of corticosterone was found to be significantly lower in GAT1(-/-) mice. These results showed that the absence of GAT1 affects mental status through enhancing the GABAergic system, as well as modifying the serotonergic system and the hypothalamic-pituitary-adrenal (HPA) activity in mice.


Asunto(s)
Trastornos de Ansiedad/metabolismo , Encéfalo/metabolismo , Trastorno Depresivo/metabolismo , Proteínas Transportadoras de GABA en la Membrana Plasmática/genética , Ácido gamma-Aminobutírico/metabolismo , Animales , Ansiolíticos/farmacología , Antidepresivos/farmacología , Trastornos de Ansiedad/genética , Trastornos de Ansiedad/fisiopatología , Conducta Animal/fisiología , Encéfalo/fisiopatología , Química Encefálica/genética , Corticosterona/sangre , Corticosterona/metabolismo , Trastorno Depresivo/genética , Trastorno Depresivo/fisiopatología , Resistencia a Medicamentos/genética , Femenino , Proteínas Transportadoras de GABA en la Membrana Plasmática/deficiencia , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pruebas Neuropsicológicas , Serotonina/metabolismo , Estrés Psicológico/metabolismo , Estrés Psicológico/fisiopatología
16.
J Neurosci Res ; 84(2): 255-67, 2006 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16683252

RESUMEN

It is widely accepted that the GABAergic system plays an important role in the action of ethanol in vivo. GABA transporter subtype 1 (GAT1) constructs high affinity reuptake sites in the CNS and regulates GABAergic transmissions. In this study, mice lacking the GAT1 were developed by homologous recombination. Both hetero- and homozygous GAT1 mutant mice were tested for ethanol, saccharin or quinine consumption, ethanol-conditioned place preference, ethanol-conditioned taste aversion, ethanol-simulated motor activity, and ethanol-induced sedation/hypnosis. The GAT1(-/-) mice showed decreased ethanol aversion and ethanol reward, and insensitivity to both the sedative/hypnotic and the motor stimulant effects of ethanol, along with increased avoidance of quinine preference and consumption. GAT1(+/-) mice showed significantly increased consumption of ethanol and saccharin, however, enhanced the rewarding and preference effect of ethanol, increased avoidance of quinine, and higher sensitivity to the motor stimulant effect of ethanol. These results demonstrate that GAT1, perhaps in a bi-directional way, modulates some behavioral effects of ethanol. The GAT1 mutant mice provided us a very useful model to investigate the mechanisms of ethanol action in vivo.


Asunto(s)
Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Proteínas Transportadoras de GABA en la Membrana Plasmática/genética , Analgésicos no Narcóticos/farmacología , Animales , Northern Blotting , Southern Blotting , Encéfalo/metabolismo , Depresores del Sistema Nervioso Central/metabolismo , Condicionamiento Clásico/efectos de los fármacos , Etanol/metabolismo , Proteínas Transportadoras de GABA en la Membrana Plasmática/biosíntesis , Ratones , Ratones Noqueados , Quinina/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sacarina/farmacología , Edulcorantes/farmacología , Sinaptosomas/efectos de los fármacos , Sinaptosomas/fisiología
17.
Acta Crystallogr C ; 61(Pt 4): m177-9, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15805614

RESUMEN

The title compound, [CuII(2)(C7H3O6S)2(C10H9N3)2][CuI(C10H9N3)2]2.2H2O, consists of anionic CuII moieties, cationic CuI species and uncoordinated water molecules. The anionic dimeric unit consists of one crystallographically independent fully deprotonated 5-sulfosalicylate (2-oxido-5-sulfonatobenzoate) anion, a di-2-pyridylamine group and a CuII atom. Each CuII atom is five-coordinate within a square-pyramidal geometry. The anion lies on a special position of -1 site symmetry. In the cationic monomer, the CuI atom adopts tetrahedral geometry. The cations and anions are connected by O-H...O and N-H...O hydrogen bonds.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...