Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
PLoS One ; 17(1): e0258998, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35081125

RESUMEN

Hepatocellular carcinoma (HCC) is one of the most burdened tumors worldwide, with a complex and multifactorial pathogenesis. Current treatment approaches involve different molecular targets. Phytochemicals have shown considerable promise in the prevention and treatment of HCC. We investigated the efficacy of two natural components, 1,8 cineole (Cin) and ellagic acid (EA), against diethylnitrosamine/2-acetylaminofluorene (DEN/2-AAF) induced HCC in rats. DEN/2-AAF showed deterioration of hepatic cells with an impaired functional capacity of the liver. In addition, the levels of tumor markers including alpha-fetoprotein, arginase-1, alpha-L-fucosidase, and ferritin were significantly increased, whereas the hepatic miR-122 level was significantly decreased in induced-HCC rats. Interestingly, treatment with Cin (100mg/kg) and EA (60mg/kg) powerfully restored these biochemical alterations. Moreover, Cin and EA treatment exhibited significant downregulation in transforming growth factor beta-1 (TGF-ß1), Fascin-1 (FSCN1), vascular endothelial growth factor (VEGF), matrix metalloproteinase-9 (MMP-9), and epithelial-mesenchymal transition (EMT) key marker, vimentin, along with a restoration of histopathological findings compared to HCC group. Such effects were comparable to Doxorubicin (DOX) (2mg/kg); however, a little additive effect was evident through combining these phytochemicals with DOX. Altogether, this study highlighted 1,8 cineole and ellagic acid for the first time as promising phytochemicals for the treatment of hepatocarcinogenesis via regulating multiple targets.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Hepatocelular , Ácido Elágico , Eucaliptol , Fitoquímicos/farmacología , Animales , Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Proteínas Portadoras/efectos de los fármacos , Proteínas Portadoras/metabolismo , Modelos Animales de Enfermedad , Ácido Elágico/administración & dosificación , Ácido Elágico/farmacología , Eucaliptol/administración & dosificación , Eucaliptol/farmacología , Humanos , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/metabolismo , MicroARNs/efectos de los fármacos , MicroARNs/metabolismo , Proteínas de Microfilamentos/efectos de los fármacos , Proteínas de Microfilamentos/metabolismo , Ratas , Factor de Crecimiento Transformador beta1/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Vimentina/efectos de los fármacos , Vimentina/metabolismo
2.
Neuropharmacology ; 196: 108693, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34229013

RESUMEN

Several lines of evidence have strongly implicated neuroinflammation in Parkinson's disease (PD) progression and l-dopa-induced dyskinesia. The present study investigated whether early subchronic pretreatment with the serotonin 5-HT1A/1B receptor agonist eltoprazine plus the adenosine A2A receptor antagonist preladenant counteracted l-dopa-induced abnormal involuntary movements (AIMs, index of dyskinesia), and neuroinflammation, in unilateral 6-hydroxydopamine(6-OHDA)-lesioned rat model of PD. The immunoreactivity of glial fibrillary acidic protein (GFAP), and the colocalization of ionized calcium binding adaptor molecule-1 (IBA-1), with interleukin (IL)-1ß, tumor-necrosis-factor-α (TNF-α) and IL-10 were evaluated in the denervated caudate-putamen (CPu) and substantia nigra pars-compacta (SNc). The combined subchronic pretreatment with l-dopa plus eltoprazine and preladenant reduced AIMs induced by acute l-dopa challenge in these rats and decreased GFAP and IBA-1 immunoreactivity induced by the drug in both CPu and SNc, with reduction in IL-1ß in IBA-1-positive cells in both CPu and SNc, and in TNF-α in IBA-1-positive cells in SNc. Moreover, a significant increase in IL-10 in IBA-1-positive cells was observed in SNc. Evaluation of immediate early-gene zif-268 (index of neuronal activation) after l-dopa challenge, showed an increase in its expression in denervated CPu of rats pretreated with l-dopa or l-dopa plus preladenant compared with vehicle, whereas rats pretreated with eltoprazine, with or without preladenant, had lower zif-268 expression. Finally, tyrosine hydroxylase and dopamine transporter examined to evaluate neurodegeneration, showed a significant equal decrease in all experimental groups. The present findings suggest that combination of l-dopa with eltoprazine and preladenant may be promising therapeutic strategy for delaying the onset of dyskinesia, preserving l-dopa efficacy and reducing neuroinflammation markers in nigrostriatal system of 6-OHDA-lesioned rats.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/farmacología , Antiparkinsonianos/efectos adversos , Discinesia Inducida por Medicamentos/fisiopatología , Levodopa/efectos adversos , Trastornos Parkinsonianos/fisiopatología , Piperazinas/farmacología , Pirimidinas/farmacología , Agonistas del Receptor de Serotonina 5-HT1/farmacología , Triazoles/farmacología , Animales , Proteínas de Unión al Calcio/efectos de los fármacos , Proteínas de Unión al Calcio/metabolismo , Núcleo Caudado/efectos de los fármacos , Núcleo Caudado/metabolismo , Discinesia Inducida por Medicamentos/etiología , Discinesia Inducida por Medicamentos/metabolismo , Proteína Ácida Fibrilar de la Glía/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Interleucina-10/metabolismo , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/metabolismo , Proteínas de Microfilamentos/efectos de los fármacos , Proteínas de Microfilamentos/metabolismo , Enfermedades Neuroinflamatorias/metabolismo , Oxidopamina/toxicidad , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/metabolismo , Porción Compacta de la Sustancia Negra/efectos de los fármacos , Porción Compacta de la Sustancia Negra/metabolismo , Putamen/efectos de los fármacos , Putamen/metabolismo , Ratas , Receptor de Serotonina 5-HT1A , Receptor de Serotonina 5-HT1B , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
3.
Eur J Pharmacol ; 900: 174038, 2021 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-33737008

RESUMEN

Subarachnoid hemorrhage (SAH) due to rupture of an intracranial aneurysm leads to vasospasm resulting in delayed cerebral ischemia. Therapeutic options are currently limited to hemodynamic optimization and nimodipine, which have marginal clinical efficacy. Nitric oxide (NO) modulates cerebral blood flow through activation of the cGMP-Protein Kinase G (PKG) pathway. Our hypothesis is that SAH results in downregulation of signaling components in the NO-PKG pathway which could explain why treatments for vasospasm targeting this pathway lack efficacy and that treatment with a cell permeant phosphopeptide mimetic of downstream effector prevents delayed vasospasm after SAH. Using a rat endovascular perforation model, reduced levels of NO-PKG pathway molecules were confirmed. Additionally, it was determined that expression and phosphorylation of a PKG substrate: Vasodilator-stimulated phosphoprotein (VASP) was downregulated. A family of cell permeant phosphomimetic of VASP (VP) was wasdesigned and shown to have vasorelaxing property that is synergistic with nimodipine in intact vascular tissuesex vivo. Hence, treatment targeting the downstream effector of the NO signaling pathway, VASP, may bypass receptors and signaling elements leading to vasorelaxation and that treatment with VP can be explored as a therapeutic strategy for SAH induced vasospasm and ameliorate neurological deficits.


Asunto(s)
Fosfopéptidos/uso terapéutico , Hemorragia Subaracnoidea/tratamiento farmacológico , Vasodilatadores/uso terapéutico , Vasoespasmo Intracraneal/tratamiento farmacológico , Animales , Moléculas de Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/efectos de los fármacos , Regulación hacia Abajo , Diseño de Fármacos , Sinergismo Farmacológico , Proteínas de Microfilamentos/efectos de los fármacos , Proteínas de Microfilamentos/metabolismo , Imitación Molecular , Nimodipina/farmacología , Óxido Nítrico/metabolismo , Fosfopéptidos/farmacocinética , Fosfoproteínas/efectos de los fármacos , Fosfoproteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Hemorragia Subaracnoidea/metabolismo , Porcinos , Vasodilatadores/farmacocinética
4.
Cell Biol Int ; 45(5): 1030-1037, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33404163

RESUMEN

Antimetastatic effect of Metformin has been documented in epithelial ovarian cancer (EOC). Presently, we investigated the regulatory mechanism of Metformin in EOC metastasis. First, Girdin was significantly enhanced in EOC tumorous tissues and cell lines. Seconded, knockdown of Girdin significantly suppressed EOC cell viability, migration, and invasion, while upregulation of Girdin produced the opposite effects in vitro and facilitated lung metastasis in EOC cell xenograft in vivo. In addition, we confirmed that the inhibitory effect of Metformin on Girdin expression. Mechanistically, the oncogenic effects of Girdin could be reversed by LY294002 (an AKT pathway inhibitor) and Metformin. These results suggested that Metformin attenuated EOC metastasis through Girdin and targeting Girdin may be a promising therapeutic strategy for EOC in the future.


Asunto(s)
Carcinoma Epitelial de Ovario/metabolismo , Proteínas de Microfilamentos/genética , Metástasis de la Neoplasia/tratamiento farmacológico , Proteínas de Transporte Vesicular/genética , Adulto , Animales , Carcinoma Epitelial de Ovario/genética , Carcinoma Epitelial de Ovario/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Supervivencia Celular/genética , Transición Epitelial-Mesenquimal , Femenino , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Metformina/metabolismo , Metformina/farmacología , Ratones Desnudos , Proteínas de Microfilamentos/efectos de los fármacos , Proteínas de Microfilamentos/metabolismo , Persona de Mediana Edad , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Transcriptoma/genética , Proteínas de Transporte Vesicular/efectos de los fármacos , Proteínas de Transporte Vesicular/metabolismo
5.
BMC Womens Health ; 20(1): 256, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33213437

RESUMEN

BACKGROUND: Fascin is an actin-binding protein and highly expressed in ovarian cancer cells. It is associated with metastasis of cancer and may be a useful prognostic factor. Anticancer activity of curcumin is related to its effect on several signaling mechanisms. Although there have been many reports regarding the anticancer properties of curcumin, its inhibitory effects on migration and invasion of ovarian cancer cells, particularly in the context of fascin expression, have not been reported. The purpose of this study was to investigate the effect of curcumin on fascin expression in ovarian cancer cells and to propose a possible mechanism for the anticancer activity of curcumin through reduced fascin expression. METHODS: SKOV3, human epithelial ovary cancer cell line, was cultured with curcumin at various dose and duration. The fascin was quantified using cell viability test and Western blot. To determine the effect of curcumin on the upstream pathway of fascin expression, the signal transducer and activator of transcription 3 (STAT3) was analyzed by sandwich-ELISA. Attachment assay, migration assay and invasion assay were analyzed to approve the change of cellular invasiveness of ovary cancer after curcumin. To determine the morphological changes of ovarian cancer cells by curcumin, immunofluorescence was performed. RESULTS: MTS assays showed that cell viability was different at various concentration of curcumin, and as concentration increased, cell viability tended to decrease. Curcumin appears to suppress fascin expression, even with a minimal concentration and short exposure time. Also, curcumin may suppress fascin expression in ovarian cancer cells through STAT3 downregulation. The attachment assay, migration assay and invasion assay of the ovarian cancer cells exhibited a statistically significant decrease. Immunofluorescence revealed a change of cell shape from a typical form of uninfluenced cells to a more polygonal appearance, with a significant reduction in filopodia formation. CONCLUSIONS: Curcumin reduces fascin expression through JAK/STAT3 pathway inhibition, which interferes with the cellular interactions essential for the metastasis and recurrence of ovarian cancer cells. Higher curcumin concentrations and longer exposure times concomitantly decreased fascin expression.


Asunto(s)
Proteínas Portadoras , Curcumina , Proteínas de Microfilamentos , Neoplasias Ováricas , Proteínas Portadoras/efectos de los fármacos , Proteínas Portadoras/metabolismo , Curcumina/farmacología , Femenino , Humanos , Quinasas Janus/metabolismo , Proteínas de Microfilamentos/efectos de los fármacos , Proteínas de Microfilamentos/metabolismo , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
6.
Acta Neuropathol Commun ; 8(1): 166, 2020 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-33076989

RESUMEN

Repeated exposure to mild TBI (mTBI) has been linked to an increased risk of Alzheimer's disease (AD), chronic traumatic encephalopathy (CTE) and other neurodegenerative diseases. Some pathological features typically observed in AD have been found in postmortem brains of TBI and CTE, hence treatments tested for AD have a potential to be effective against r-mTBI outcomes. Neuroinflammation may present a possible answer due to its central role both in acute brain injury and in chronic degenerative-like disorders. Our previous studies have shown that drug nilvadipine, acting as an inhibitor of spleen tyrosine kinase (SYK), is effective at reducing inflammation, tau hyperphosphorylation and amyloid production in AD mouse models. To demonstrate the effect of nilvadipine in the absence of age-related variables, we introduced the same treatment to young r-mTBI mice. We further investigate therapeutic mechanisms of nilvadipine using its racemic properties. Both enantiomers, (+)-nilvadipine and (-)-nilvadipine, can lower SYK activity, whereas (+)-nilvadipine is also a potent L-type calcium channel blocker (CCB) and shown to be anti-hypertensive. All r-mTBI mice exhibited increased neuroinflammation and impaired cognitive performance and motor functions. Treatment with racemic nilvadipine mitigated the TBI-induced inflammatory response and significantly improved spatial memory, whereas (-)-enantiomer decreased microgliosis and improved spatial memory but failed to reduce the astroglial response to as much as the racemate. These results suggest the therapeutic potential of SYK inhibition that is enhanced when combined with the CCB effect, which indicate a therapeutic advantage of multi-action drugs for r-mTBI.


Asunto(s)
Conmoción Encefálica/fisiopatología , Bloqueadores de los Canales de Calcio/farmacología , Nifedipino/análogos & derivados , Aprendizaje Espacial/efectos de los fármacos , Memoria Espacial/efectos de los fármacos , Quinasa Syk/antagonistas & inhibidores , Animales , Antígenos CD/efectos de los fármacos , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/efectos de los fármacos , Antígenos de Diferenciación Mielomonocítica/metabolismo , Conmoción Encefálica/metabolismo , Conmoción Encefálica/psicología , Proteínas de Unión al Calcio/efectos de los fármacos , Proteínas de Unión al Calcio/metabolismo , Proteína Ácida Fibrilar de la Glía/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Inflamación/metabolismo , Ratones , Proteínas de Microfilamentos/efectos de los fármacos , Proteínas de Microfilamentos/metabolismo , Nifedipino/farmacología , Fosforilación , Prueba de Desempeño de Rotación con Aceleración Constante , Aprendizaje Espacial/fisiología , Memoria Espacial/fisiología , Quinasa Syk/efectos de los fármacos , Quinasa Syk/metabolismo
7.
BMC Nephrol ; 21(1): 402, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32948146

RESUMEN

BACKGROUND: Dipeptidyl peptidase-4 (DPP-4) is a serine protease that inhibits the degradation of glucagon-like peptide 1. DPP-4 inhibitors are used worldwide to treat type 2 diabetes mellitus and were recently shown to have pleiotropic effects such as anti-oxidant, anti-inflammatory, and anti-fibrotic actions. DPP-4 inhibitors improve albuminuria and renal injury including glomerular damage independent of its hypoglycemic effect. Although DPP-4 is mainly expressed in the kidney, the physiological function of DPP-4 remains unclear. METHODS: The localization of renal DPP-4 activity was determined in human renal biopsy specimens with glycyl-1-prolyl-4-methoxy-2-naphthylamide and the effects of a DPP-4 inhibitor were examined in human cultured podocyte. RESULTS: DPP-4 activity under normal conditions was observed in some Bowman's capsular epithelial cells and proximal tubules, but not in the glomerulus. DPP-4 activity was observed in crescent formation in anti-neutrophil myeloperoxidase cytoplasmic antigen antibody nephritis, nodular lesions in diabetic nephropathy, and some podocytes in focal segmental glomerulosclerosis. Notably, the DPP-4 inhibitor saxagliptin suppressed DPP-4 activity in podocytes and the proximal tubules. To assess the effect of DPP-4 inhibitor on podocytes, human cultured podocytes were injured by Adriamycin, which increased DPP-4 activity; this activity was dose-dependently suppressed by saxagliptin. Treatment with saxagliptin maintained the structure of synaptopodin and RhoA. Saxagliptin also improved the detachment of podocytes. CONCLUSIONS: DPP-4 activity induces degradation of synaptopodin and reduction of RhoA, resulting in destruction of the podocyte cytoskeleton. Saxagliptin may have pleiotropic effects to prevent podocyte injury.


Asunto(s)
Adamantano/análogos & derivados , Nefropatías Diabéticas/metabolismo , Dipéptidos/farmacología , Dipeptidil Peptidasa 4/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Riñón/metabolismo , Nefritis/metabolismo , Podocitos/efectos de los fármacos , Adamantano/farmacología , Anticuerpos Anticitoplasma de Neutrófilos/inmunología , Cápsula Glomerular/metabolismo , Nefropatías Diabéticas/tratamiento farmacológico , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Doxorrubicina/farmacología , Femenino , Humanos , Técnicas In Vitro , Glomérulos Renales/metabolismo , Túbulos Renales Proximales/metabolismo , Masculino , Proteínas de Microfilamentos/efectos de los fármacos , Proteínas de Microfilamentos/metabolismo , Nefritis/inmunología , Podocitos/metabolismo , Proteína de Unión al GTP rhoA/efectos de los fármacos , Proteína de Unión al GTP rhoA/metabolismo
8.
Am J Hypertens ; 33(8): 775-783, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32301965

RESUMEN

BACKGROUND: Aberrant activation of epidermal growth factor receptor (EGFR) signaling pathway is associated with the pathogenesis of pulmonary hypertension (PH). However, the effect of icotinib, a first generation of EGFR tyrosine kinase inhibitor (EGFR-TKI), on PH remains to be elucidated. METHODS: PH rat model was established by a single intraperitoneal injection of monocrotaline (MCT, 60 mg/kg). Icotinib (15, 30, and 60 mg/kg/day) was administered by oral gavage from the day of MCT injection. After 4 weeks, hemodynamic parameters and histological changes of the pulmonary arterial vessels were assessed, and the phenotypic switching of pulmonary arterial smooth muscle cells (PASMCs) was determined in vivo. Moreover, the effects of icotinib (10 µM) on epidermal growth factor (EGF, 50 ng/ml)-stimulated proliferation, migration, and phenotypic switching of human PASMCs were explored in vitro. RESULTS: Icotinib significantly reduced the right ventricular systolic pressure and right ventricle hypertrophy index in rats with MCT-induced PH. Moreover, icotinib improved MCT-induced pulmonary vascular remodeling. The expression of contractile marker (smooth muscle 22 alpha (SM22α)) and synthetic markers (osteopontin (OPN) and vimentin) in pulmonary artery was restored by icotinib treatment. In vitro, icotinib suppressed EGF-induced PASMCs proliferation and migration. Meanwhile, icotinib inhibited EGF-induced downregulation of α-smooth muscle actin and SM22α and upregulation of OPN and Collagen I in PASMCs, suggesting that icotinib could inhibit EGF-induced phenotypic switching of PASMCs. Mechanistically, these effects of icotinib were associated with the inhibition of EGFR-Akt/ERK signaling pathway. CONCLUSIONS: Icotinib can attenuate MCT-induced pulmonary vascular remodeling and improve PH. This effect of icotinib might be attributed to preventing PASMC dysfunction by inhibiting EGFR-Akt/ERK signaling pathway.


Asunto(s)
Éteres Corona/farmacología , Receptores ErbB/antagonistas & inhibidores , Hipertensión Pulmonar/fisiopatología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Arteria Pulmonar/efectos de los fármacos , Quinazolinas/farmacología , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Factor de Crecimiento Epidérmico/farmacología , Hipertensión Pulmonar/inducido químicamente , Técnicas In Vitro , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas de Microfilamentos/efectos de los fármacos , Proteínas de Microfilamentos/metabolismo , Monocrotalina/toxicidad , Proteínas Musculares/efectos de los fármacos , Proteínas Musculares/metabolismo , Músculo Liso Vascular/fisiopatología , Osteopontina/efectos de los fármacos , Osteopontina/metabolismo , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Arteria Pulmonar/fisiopatología , Ratas , Transducción de Señal , Remodelación Vascular/efectos de los fármacos , Función Ventricular Derecha/efectos de los fármacos , Presión Ventricular/efectos de los fármacos , Vimentina/efectos de los fármacos , Vimentina/metabolismo
9.
Behav Brain Res ; 384: 112520, 2020 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-32006563

RESUMEN

Cerebral ischemia/reperfusion (I/R) injury is a leading cause of learning and memory dysfunction. Hydrogen sulfide (H2S) has been shown to confer neuroprotection in various neurodegenerative diseases, including cerebral I/R-induced hippocampal CA1 injury. However, the underlying mechanisms have not been completely understood. In the present study, rats were pretreated with SAM/NaHS (SAM, an H2S agonist, and NaHS, an H2S donor) only or SAM/NaHS combined with CaM (an activator of CaMKII) prior to cerebral ischemia. The Morris water maze test demonstrated that SAM/NaHS could alleviate learning and memory impairment induced by cerebral I/R injury. Cresyl violet staining was used to show the survival of hippocampal CA1 pyramidal neurons. SAM/NaHS significantly increased the number of surviving cells, whereas CaM weakened the protection induced by SAM/NaHS. The immunohistochemistry results indicated that the number of Iba1-positive microglia significantly increased after cerebral I/R. Compared with the I/R group, the number of Iba1-positive microglia in the SAM/NaHS groups significantly decreased. Co-Immunoprecipitation and immunoblotting were conducted to demonstrate that SAM/NaHS suppressed the assembly of CaMKII with the ASK1-MKK3-p38 signal module after cerebral I/R, which decreased the phosphorylation of p38. In contrast, CaM significantly inhibited the effects of SAM/NaHS. Taken together, the results suggested that SAM/NaHS could suppress cerebral I/R injury by downregulating p38 phosphorylation via decreasing the assembly of CaMKII with the ASK1-MKK3-p38 signal module.


Asunto(s)
Región CA1 Hipocampal/efectos de los fármacos , Calmodulina/farmacología , Sulfuro de Hidrógeno/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Trastornos de la Memoria/metabolismo , Daño por Reperfusión/metabolismo , S-Adenosilmetionina/farmacología , Sulfuros/farmacología , Animales , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/patología , Proteínas de Unión al Calcio/efectos de los fármacos , Proteínas de Unión al Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Regulación hacia Abajo , Accidente Cerebrovascular Isquémico/fisiopatología , Aprendizaje/efectos de los fármacos , MAP Quinasa Quinasa 3/efectos de los fármacos , MAP Quinasa Quinasa 3/metabolismo , MAP Quinasa Quinasa Quinasa 5/efectos de los fármacos , MAP Quinasa Quinasa Quinasa 5/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Trastornos de la Memoria/fisiopatología , Proteínas de Microfilamentos/efectos de los fármacos , Proteínas de Microfilamentos/metabolismo , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Prueba del Laberinto Acuático de Morris , Fosforilación , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Ratas , Daño por Reperfusión/fisiopatología , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Plant Physiol ; 181(1): 161-178, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31311831

RESUMEN

Auxin transport inhibitors are essential tools for understanding auxin-dependent plant development. One mode of inhibition affects actin dynamics; however, the underlying mechanisms remain unclear. In this study, we characterized the action of 2,3,5-triiodobenzoic acid (TIBA) on actin dynamics in greater mechanistic detail. By surveying mutants for candidate actin-binding proteins with reduced TIBA sensitivity, we determined that Arabidopsis (Arabidopsis thaliana) villins contribute to TIBA action. By directly interacting with the C-terminal headpiece domain of villins, TIBA causes villin to oligomerize, driving excessive bundling of actin filaments. The resulting changes in actin dynamics impair auxin transport by disrupting the trafficking of PIN-FORMED auxin efflux carriers and reducing their levels at the plasma membrane. Collectively, our study provides mechanistic insight into the link between the actin cytoskeleton, vesicle trafficking, and auxin transport.


Asunto(s)
Actinas/metabolismo , Proteínas de Arabidopsis/metabolismo , Arabidopsis/efectos de los fármacos , Ácidos Indolacéticos/metabolismo , Proteínas de Microfilamentos/antagonistas & inhibidores , Reguladores del Crecimiento de las Plantas/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Arabidopsis/genética , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Transporte Biológico/efectos de los fármacos , Membrana Celular/metabolismo , Proteínas de Microfilamentos/efectos de los fármacos , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Mutación , Transporte de Proteínas/efectos de los fármacos , Ácidos Triyodobenzoicos/farmacología
11.
Endocr J ; 66(1): 65-74, 2019 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-30369516

RESUMEN

The human homologue of flightless-I (FLII) belong to the gelsolin protein family and contain a gelsolin-like domain at the C-terminus and a leucine-rich repeat (LRR) domain at the N-terminus. FLII regulates estrogen receptor alpha (ERα) and glucocorticoid receptor (GR)-mediated transcription by direct interaction through different domains, suggestive of its potential role in the crosstalk between the ERα and GR signaling pathway. Here, we demonstrate that FLII plays a critical role in GR-mediated repression of ERα target gene expression. In FLII-depleted cells, the reduction in 17-ß-estradiol (E2)-induced ERα occupancy following treatment with dexamethasone (Dex) at the estrogen responsive element (ERE) site of the ERα target gene was significantly inhibited. The ERE binding of GR by the cotreatment with E2 and Dex was significantly inhibited by FLII depletion, indicating that FLII is required for the recruitment of GR at the ERE sites of ERα target genes. In addition, the recruitment of ERα-induced FLII to ERE sites was significantly reduced by Dex treatment. In protein binding assays, GR inhibited the E2-induced interaction between ERα and FLII, suggesting that GR interferes with the binding of ERα and FLII at the ERα target genes, resulting in the release of ERα and FLII from EREs. Taken together, our data reveal an unknown mechanism by which the transcription coactivator FLII regulates the GR-mediated repression of ERα target gene expression in MCF-7 cells.


Asunto(s)
Neoplasias de la Mama/genética , Receptor alfa de Estrógeno/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Microfilamentos/genética , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Glucocorticoides/genética , Neoplasias de la Mama/metabolismo , Dexametasona/farmacología , Estradiol/farmacología , Receptor alfa de Estrógeno/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Estrógenos/farmacología , Glucocorticoides/farmacología , Humanos , Células MCF-7 , Proteínas de Microfilamentos/efectos de los fármacos , Proteínas de Microfilamentos/metabolismo , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Glucocorticoides/efectos de los fármacos , Receptores de Glucocorticoides/metabolismo , Elementos de Respuesta , Transactivadores
12.
J Neurosci Res ; 96(12): 1887-1899, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30242877

RESUMEN

Stachybotrys microspora triprenyl phenol (SMTP)-44D has both anti-oxidative and anti-inflammatory activities, but its efficacy has not been proved in relation to the pathological changes of neurovascular unit (NVU) and neurovascular trophic coupling (NVTC) in ischemic stroke. Here, the present study was designed to assess the efficacies of SMTP-44D, moreover, compared with the standard neuroprotective reagent edaravone in ischemic brains. ICR mice were subjected to transient middle cerebral artery occlusion (tMCAO) for 60 min, SMTP-44D (10 mg/kg) or edaravone (3 mg/kg) was intravenously administrated through subclavian vein just after the reperfusion, and these mice were examined at 1, 3, and 7 d after reperfusion. Compared with the vehicle group, SMTP-44D treatment revealed obvious ameliorations in clinical scores and infarct volume, meanwhile, markedly suppressed the accumulations of 4-HNE, 8-OHdG, nitrotyrosine, RAGE, TNF-α, Iba-1, and cleaved caspase-3 after tMCAO. In addition, SMTP-44D significantly prevented the dissociation of NVU and improved the intensity of NAGO/BDNF and the number of BDNF/TrkB and BDNF/NeuN double positive cells. These effects of SMTP-44D in reducing oxidative and inflammatory stresses were similar to or stronger than those of edaravone. The present study demonstrated that SMTP-44D showed strong anti-oxidative, anti-inflammatory, and anti-apoptotic effects, moreover, the drug also significantly improved the NVU damage and NVTC in the ischemic brain.


Asunto(s)
Infarto Encefálico/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Fenoles/farmacología , Accidente Cerebrovascular/tratamiento farmacológico , Acetilglucosamina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Vasos Sanguíneos/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteínas de Unión al Calcio/efectos de los fármacos , Caspasa 3/efectos de los fármacos , Proteínas de Unión al ADN , Fibrinolíticos/farmacología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Masculino , Ratones , Ratones Endogámicos ICR , Proteínas de Microfilamentos/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Estrés Oxidativo/efectos de los fármacos , Piroptosis/efectos de los fármacos , Stachybotrys , Activador de Tejido Plasminógeno/farmacología , Factor de Necrosis Tumoral alfa/efectos de los fármacos
13.
Med Sci Monit ; 24: 3840-3847, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29875353

RESUMEN

BACKGROUND Molecular hydrogen (H2) has been widely reported to have benefiicial effects in diverse animal models and human disease through reduction of oxidative stress and inflammation. The aim of this study was to investigate whether hydrogen gas could ameliorate endotoxin-induced uveitis (EIU) in rats. MATERIAL AND METHODS Male Sprague-Dawley rats were divided into a normal group, a model group, a nitrogen-oxygen (N-O) group, and a hydrogen-oxygen (H-O) group. EIU was induced in rats of the latter 3 groups by injection of lipopolysaccharide (LPS). After that, rats in the N-O group inhaled a gas mixture of 67% N2 and 33% O2, while those in the H-O group inhaled a gas mixture of 67% H2 and 33% O2. All rats were graded according to the signs of uveitis after electroretinography (ERG) examination. Protein concentration in the aqueous humor (AqH) was measured. Furthermore, hematoxylin-eosin staining and immunostaining of anti-ionized calcium-binding adapter molecule 1 (Iba1) in the iris and ciliary body (ICB) were carried out. RESULTS No statistically significant differences existed in the graded score of uveitis and the b-wave peak time in the Dark-adapted 3.0 ERG among the model, N-O, and H-O groups (P>0.05), while rats of the H-O group showed a lower concentration of AqH protein than that of the model or N-O group (P<0.05). The number of the infiltrating cells in the ICB of rats from the H-O group was not significantly different from that of the model or N-O group (P>0.05), while the activation of microglia cells in the H-O group was somewhat reduced (P<0.05). CONCLUSIONS Post-treatment hydrogen gas inhalation did not ameliorate the clinical signs, or reduce the infiltrating cells of EIU. However, it inhibited the elevation of protein in the AqH and reduced the microglia activation.


Asunto(s)
Hidrógeno/uso terapéutico , Uveítis/terapia , Animales , Humor Acuoso/efectos de los fármacos , Proteínas de Unión al Calcio/efectos de los fármacos , Cuerpo Ciliar/efectos de los fármacos , Modelos Animales de Enfermedad , Endotoxinas/efectos adversos , Hidrógeno/administración & dosificación , Hidrógeno/fisiología , Iris/efectos de los fármacos , Lipopolisacáridos/farmacología , Masculino , Proteínas de Microfilamentos/efectos de los fármacos , Microglía/efectos de los fármacos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ratas , Ratas Sprague-Dawley , Uveítis/inducido químicamente
14.
Am J Ther ; 24(2): e162-e166, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27415979

RESUMEN

BACKGROUND: Proton pump inhibition (PPI) administrated together with adenosine diphosphate (ADP) receptor blockers (ADPRB) significantly reduces the risk of gastrointestinal bleeding. Nevertheless, there is a heated discussion about an interaction between PPI and ADPRB that leads to high on-treatment platelet reactivity (HTPR). STUDY QUESTION: Is there a relationship between pantoprazole PPI and HTPR on ADPRB therapy in patients with acute ST-elevation myocardial infarction (STEMI). METHODS: Single center pilot study in patients with acute STEMI was performed. This study enrolled totally 87 patients (34 clopidogrel-treated and 53 new ADPRB-treated patients). Pantoprazole was administrated in 33 patients. HTPR was detected with ADP-induced light transmission aggregometry and vasodilator-stimulated phosphoprotein phosphorylation analysis. Samples were taken before coronary angiography (sample 1) and on the next day after the procedure (sample 2). RESULTS: No significant differences were found in pantoprazole-treated patients and patients without PPI neither in sample 1 (59.2 ± 29.5% vs. 54.9 ± 22.7%, P = 0.49) nor in sample 2 (43.8 ± 27.2% vs. 37.0 ± 22.9%, P = 0.30). Similarly, there were no significant differences in the platelet reactivity index of vasodilator-stimulated phosphoprotein phosphorylation in both samples (sample 1: 53.3 ± 29.8% vs. 65.0 ± 20.5%, P = 0.11; sample 2: 30.8 ± 27.1% vs. 40.6 ± 27.5%, P = 0.19). A comparison of clopidogrel and new ADP receptor blockers in patients on pantoprazole PPI did not reveal significant differences in on-treatment platelet reactivity. CONCLUSIONS: This study did not reveal interaction between pantoprazole and ADPRB in patients with acute STEMI.


Asunto(s)
2-Piridinilmetilsulfinilbencimidazoles/farmacología , Plaquetas/efectos de los fármacos , Moléculas de Adhesión Celular/efectos de los fármacos , Proteínas de Microfilamentos/efectos de los fármacos , Fosfoproteínas/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de la Bomba de Protones/farmacología , Antagonistas del Receptor Purinérgico P2Y/uso terapéutico , Infarto del Miocardio con Elevación del ST/terapia , Adenosina/análogos & derivados , Adenosina/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Moléculas de Adhesión Celular/metabolismo , Clopidogrel , Angiografía Coronaria , Interacciones Farmacológicas , Femenino , Humanos , Masculino , Proteínas de Microfilamentos/metabolismo , Persona de Mediana Edad , Pantoprazol , Intervención Coronaria Percutánea , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Proyectos Piloto , Clorhidrato de Prasugrel/uso terapéutico , Estudios Prospectivos , Ticagrelor , Ticlopidina/análogos & derivados , Ticlopidina/uso terapéutico
15.
Biomarkers ; 22(2): 133-144, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27781498

RESUMEN

OBJECTIVE: Our purpose was to study the molecular basis of infliximab (IFX) effect on colon mucosa in a colitis model and to identify new biomarkers of mucosal healing. METHODS: Healthy rats and rats which were subjected to experimental colitis induced by dextran sulfate sodium, with or without IFX treatment (in the short- and long-term), were studied along with forty-seven IBD patients. Colon mucosal integrity by periodic acid Schiff (PAS) staining, intestinal damage by immunohistochemistry (proliferating cell nuclear antigen, ß-catenin, E-cadherin, phosphotyrosine, p-p38, allograft inflammatory factor-1 (AIF-1) and colonic mucosal apoptosis by TUNEL staining were evaluated in rats while serum and colon AIF-1 levels were determined in IBD patients. RESULTS: In rats with colitis, IFX reestablished the epithelial barrier integrity, recovered mucus production and decreased colon inflammation, as verified by reduced serum and colon AIF-1 levels; colon and serum AIF-1 levels were also lower in inactive IBD patients compare to active ones. P38 activation after IFX treatment tended to induce differentiation/proliferation of epithelial cells along the colonic crypt-villous axis. CONCLUSIONS: These findings support AIF-1 as a new biomarker of mucosal healing in experimental colitis and suggest that p38 activation is involved in the mucosal healing intracellular mechanism induced by IFX treatment.


Asunto(s)
Proteínas de Unión al Calcio/sangre , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Infliximab/uso terapéutico , Mucosa Intestinal/efectos de los fármacos , Proteínas de Microfilamentos/sangre , Animales , Biomarcadores/análisis , Proteínas de Unión al Calcio/efectos de los fármacos , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Proteínas de Unión al ADN/sangre , Proteínas de Unión al ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Humanos , Enfermedades Inflamatorias del Intestino/sangre , Infliximab/farmacología , Mucosa Intestinal/química , Proteínas de Microfilamentos/efectos de los fármacos , Ratas , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
Invest Ophthalmol Vis Sci ; 57(13): 5736-5747, 2016 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-27787561

RESUMEN

PURPOSE: Transforming growth factor-ß-induced epithelial-mesenchymal transition (EMT) is one of the main causes of posterior capsular opacification (PCO) or secondary cataract; however, the signaling events involved in TGF-ß-induced PCO have not been fully characterized. Here, we focus on examining the role of ß-catenin/cyclic AMP response element-binding protein (CREB)-binding protein (CBP) and ß-catenin/T-cell factor (TCF)-dependent signaling in regulating cytoskeletal dynamics during TGF-ß-induced EMT in lens epithelial explants. METHODS: Rat lens epithelial explants were cultured in medium M199 in the absence of serum. Explants were treated with TGF-ß2 in the presence or absence of the ß-catenin/CBP interaction inhibitor, ICG-001, or the ß-catenin/TCF interaction inhibitor, PNU-74654. Western blot and immunofluorescence experiments were carried out and analyzed. RESULTS: An increase in the expression of fascin, an actin-bundling protein, was observed in the lens explants upon stimulation with TGF-ß, and colocalized with F-actin filaments. Inhibition of ß-catenin/CBP interactions, but not ß-catenin/TCF interactions, led to a decrease in TGF-ß-induced fascin and stress fiber formation, as well as a decrease in the expression of known markers of EMT, α-smooth muscle actin (α-SMA) and matrix metalloproteinase 9 (MMP9). In addition, inhibition of ß-catenin/CBP-dependent signaling also prevented TGF-ß-induced downregulation of epithelial cadherin (E-cadherin) in lens explants. CONCLUSIONS: We show that ß-catenin/CBP-dependent signaling regulates fascin, MMP9, and α-SMA expression during TGF-ß-induced EMT. We demonstrate that ß-catenin/CBP-dependent signaling is crucial for TGF-ß-induced EMT in the lens.


Asunto(s)
Opacificación Capsular/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Factor de Crecimiento Transformador beta2/farmacología , beta Catenina/farmacología , Actinas , Animales , Western Blotting , Opacificación Capsular/patología , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/efectos de los fármacos , Movimiento Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Proteínas de Microfilamentos/biosíntesis , Proteínas de Microfilamentos/efectos de los fármacos , Ratas , Ratas Wistar , Proteínas Recombinantes , Transducción de Señal
17.
J Neurosurg Anesthesiol ; 28(1): 51-64, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25844953

RESUMEN

BACKGROUND: Anesthetic agents (eg, isoflurane, propofol) may cause neurodegeneration in the developing brains and impair animals' learning ability. Dexmedetomidine (DEX), a selective alpha 2-adrenoreceptor agonist, has antiapoptotic properties in several brain injury models. Here, we tested whether DEX can protect the brain from neurodegeneration in rats exposed to propofol in utero. MATERIALS AND METHODS: Fetal rats of embryonic day 20 were exposed in utero for 1 hour to propofol anesthesia with DEX or saline, or no anesthesia (control). The fetal brains were harvested 6 hours later. Cleaved caspase-3 levels and the relative number of ionized calcium-binding adaptor molecule 1 (IBA1)-positive cells were assessed by Western blot and immunohistochemistry. Learning and memory functions of the offspring in a separate cohort were assessed at postnatal day 35 by using an 8-arm radial maze. RESULTS: Propofol anesthesia in pregnant rats augmented caspase-3 activation by 217% in the brain tissues of fetal rats and increased the number of IBA1-positive cells in the cortex by 40% and in the thalamus by 270%. Juvenile rats exposed prenatally to propofol were not different than controls on spontaneous locomotor activity, but made more errors of omission and took longer to complete visiting all 8 arms on days 1, 2, and 3 across a 5-day test in the radial arm maze. This neurocognitive deficit was prevented by administration of DEX (5.0 µg/kg, IP), which also significantly inhibited propofol-induced caspase-3 activation and microglial response in the fetal brains. CONCLUSIONS: DEX attenuates neuronal injury induced by maternal propofol anesthesia in the fetal brains, providing neurocognitive protection in the offspring rats.


Asunto(s)
Anestésicos Intravenosos/efectos adversos , Dexmedetomidina/farmacología , Hipnóticos y Sedantes/farmacocinética , Síndromes de Neurotoxicidad/prevención & control , Efectos Tardíos de la Exposición Prenatal/prevención & control , Propofol/efectos adversos , Animales , Western Blotting , Encéfalo/efectos de los fármacos , Proteínas de Unión al Calcio/efectos de los fármacos , Caspasa 3/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Proteínas de Microfilamentos/efectos de los fármacos , Embarazo , Ratas , Ratas Sprague-Dawley
18.
Eur J Clin Invest ; 46(1): 7-14, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26449338

RESUMEN

BACKGROUND: Our recent drug interaction trial with clopidogrel shows that morphine decreases the concentrations and pharmacodynamic effects of clopidogrel, which could lead to treatment failure in susceptible individuals. We hypothesized that the pharmacodynamic consequences of drug-drug interactions would be less between morphine and ticagrelor. MATERIALS AND METHODS: Twenty-four healthy subjects received a loading dose of 180 mg ticagrelor together with placebo or 5 mg morphine intravenously in a randomized, double-blind, placebo-controlled, crossover trial. Pharmacokinetics were determined by liquid chromatography tandem mass spectrometry, and ticagrelor pharmacodynamic effects were measured by platelet function tests (whole blood platelet aggregation: multiplate, platelet plug formation: PFA-100, vasodilator-stimulated phosphoprotein (VASP) phosphorylation assay). RESULTS: Concomitant i.v. injection of morphine slows drug resorption of ticagrelor and its active metabolite (P < 0·05) by 1 h and decreases plasma levels of ticagrelor and its active metabolite by 25-31% (P ≤ 0·03) and the drug exposure (area under the curve) by 22-23% (P ≤ 0·01). Importantly, however, the pharmacodynamic effects of ticagrelor on platelet aggregation in whole blood, platelet plug formation and VASP phosphorylation are not affected by morphine. CONCLUSIONS: Morphine co-administration moderately decreases ticagrelor plasma concentrations but does not inhibit its pharmacodynamic effects in healthy volunteers within 6 h after drug administration. Limitations of our trial include the investigation in healthy volunteers under standardized conditions, which does not necessarily reflect a realistic emergency scenario.


Asunto(s)
Adenosina/análogos & derivados , Analgésicos Opioides/farmacología , Plaquetas/efectos de los fármacos , Morfina/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Adenosina/sangre , Adenosina/farmacología , Adulto , Moléculas de Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Cromatografía Liquida , Estudios Cruzados , Método Doble Ciego , Interacciones Farmacológicas , Femenino , Voluntarios Sanos , Humanos , Masculino , Proteínas de Microfilamentos/efectos de los fármacos , Proteínas de Microfilamentos/metabolismo , Fosfoproteínas/efectos de los fármacos , Fosfoproteínas/metabolismo , Inhibidores de Agregación Plaquetaria/sangre , Pruebas de Función Plaquetaria , Espectrometría de Masas en Tándem , Ticagrelor , Adulto Joven
19.
Psychopharmacology (Berl) ; 233(3): 447-56, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26525565

RESUMEN

RATIONALE AND OBJECTIVES: Cyclic nucleotide phosphodiesterase-2 (PDE2) is a potential therapeutic target for the treatment of cognitive dysfunction. Using the object recognition test (ORT), this study assessed the effects of two PDE2 inhibitors, Bay 60-7550 and ND7001, on learning and memory, and examined underlying mechanisms. METHODS: To assess the role of PDE2 inhibition on phases of memory, Bay 60-7550 (3 mg/kg) was administered: 30 min prior to training; 0, 1, or 3 h after training; or 30 min prior to recall testing. To assess cyclic nucleotide involvement in PDE2 inhibitor-enhanced memory consolidation, either the nitric oxide synthase inhibitor NG-nitro-L-arginine methyl ester (L-NAME; 20 mg/kg; intraperitoneal (IP)), soluble guanylyl cyclase inhibitor 1H-[-1,2,4]oxadiazolo-[4,3-a]quinoxalin-1-one (ODQ; 20 mg/kg; IP), protein kinase G inhibitor KT5823 (2.5 µg; intracerebroventricular (ICV)), or protein kinase A inhibitor H89 (1 µg; ICV) was administered 30 min prior to the PDE2 inhibitor Bay 60-7550 (3 mg/kg) or ND7001 (3 mg/kg). Changes in the phosphorylation of 3'5'-cyclic adenosine monophosphate (cAMP) response element binding protein (CREB) at Ser-133 and vasodilator-stimulated phosphoprotein (VASP) at Ser-239 were determined to confirm activation of cAMP and 3'5'-cyclic guanosine monophosphate (cGMP) signaling. RESULTS: Bay 60-7550 (3 mg/kg) enhanced memory of mice in the ORT when given 30 min prior to training, immediately after training, or 30 min prior to recall. Inhibitors of the cGMP pathway blocked the memory-enhancing effects of both Bay 60-7550 (3 mg/kg) and ND7001 (3 mg/kg) on early consolidation processes. Bay 60-7550 (3 mg/kg) enhanced phosphorylation of CREB and VASP, both targets of cGMP-dependent protein kinase (PKG). CONCLUSIONS: These results confirm a potential of PDE2, or components of its signaling pathway, as a therapeutic target for drug discovery focused on restoring memory function.


Asunto(s)
GMP Cíclico/fisiología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/antagonistas & inhibidores , Memoria/efectos de los fármacos , Inhibidores de Fosfodiesterasa/farmacología , Reconocimiento en Psicología/efectos de los fármacos , Animales , Carbazoles/farmacología , Moléculas de Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Guanilato Ciclasa/antagonistas & inhibidores , Imidazoles/farmacología , Masculino , Ratones Endogámicos ICR , Proteínas de Microfilamentos/efectos de los fármacos , Proteínas de Microfilamentos/metabolismo , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Oxadiazoles/farmacología , Fosfoproteínas/efectos de los fármacos , Fosfoproteínas/metabolismo , Inhibidores de Proteínas Quinasas , Quinoxalinas/farmacología , Triazinas/farmacología
20.
Clin Nephrol ; 84(2): 95-9, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26103965

RESUMEN

OBJECTIVE: To investigate the effect of BMP7 on the transdifferentiation and Smad7 expression of podocytes induced by high glucose in vitro and to explore its possible protective mechanisms. METHODS: Mouse podocytes were cultured and divided into normal glucose group (NG), high glucose group (HG), mannitol group, NG+BMP7 group, and HG+BMP7 group. Real-time PCR and Western blot were applied respectively to detect the mRNA and protein expression levels of synaptopodin, desmin, and Smad7. RESULTS: The cells significantly up-regulated the mRNA and protein expression of desmin and reduced the expression of both synaptopodin and Smad7 after 48 hours (vs. NG, p < 0.01). BMP7 dramatically suppressed the mRNA and protein expression of desmin and protected the expression of synaptopodin and Smad7 after incubation with high glucose for 48 hours (vs. HG, p < 0.01). CONCLUSIONS: BMP7 can inhibit the epithelial-to-mesenchymal cell transformation (EMT) of podocytes induced by high glucose; Smad7 may mediate the blunting effects of BMP7 on high glucose in podocytes.


Asunto(s)
Proteína Morfogenética Ósea 7/farmacología , Hiperglucemia/fisiopatología , Podocitos/efectos de los fármacos , Proteína smad7/efectos de los fármacos , Animales , Técnicas de Cultivo de Célula , Línea Celular , Transdiferenciación Celular/efectos de los fármacos , Desmina/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Glucosa/farmacología , Manitol/farmacología , Ratones , Proteínas de Microfilamentos/efectos de los fármacos , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA