Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Cereb Cortex ; 28(7): 2391-2404, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28591834

RESUMEN

Grb2-associated-binding protein 1 (Gab1) is a docking/scaffolding molecule known to play an important role in cell growth and survival. Here, we report that Gab1 is decreased in cholinergic neurons in Alzheimer's disease (AD) patients and in a mouse model of AD. In mice, selective ablation of Gab1 in cholinergic neurons in the medial septum impaired learning and memory and hippocampal long-term potentiation. Gab1 ablation also inhibited SK channels, leading to an increase in firing in septal cholinergic neurons. Gab1 overexpression, on the other hand, improved cognitive function and restored hippocampal CaMKII autorphosphorylation in AD mice. These results suggest that Gab1 plays an important role in the pathophysiology of AD and may represent a novel therapeutic target for diseases involving cholinergic dysfunction.


Asunto(s)
Enfermedad de Alzheimer/patología , Corteza Cerebral/patología , Neuronas Colinérgicas/fisiología , Cognición/fisiología , Regulación de la Expresión Génica/genética , Fosfoproteínas/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Proteínas Adaptadoras Transductoras de Señales , Anciano de 80 o más Años , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/diagnóstico por imagen , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/citología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Colina O-Acetiltransferasa/genética , Colina O-Acetiltransferasa/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Mutación/genética , Fosfoproteínas/genética , Presenilina-1/genética , Presenilina-1/metabolismo
2.
Hepatobiliary Pancreat Dis Int ; 17(6): 510-516, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30135046

RESUMEN

BACKGROUD: Transarterial chemoembolization (TACE) is the primary palliative treatment for patients with unresectable hepatocellular carcinoma (HCC). However, it is often accompanied by postoperative pain which hinder patient recovery. This study was to examine whether preemptive parecoxib and sufentanil-based patient controlled analgesia (PCA) could improve the pain management in patients receiving TACE for inoperable HCC. METHODS: From June to December 2016, 84 HCC patients undergoing TACE procedure were enrolled. Because of the willingness of the individuals, it is difficult to randomize the patients to different groups. We matched the patients' age, gender and pain scores, and divided the patients into the multimodal group (n = 42) and control group (n = 42). Patients in the multimodal group received 40 mg of parecoxib, 30 min before TACE, followed by 48 h of sufentanil-based PCA. Patients in the control group received a routine analgesic regimen, i.e., 5 mg of dezocine during operation, and 100 mg of tramadol or equivalent intravenous opioid according to patient's complaints and pain intensity. Postoperative pain intensity, percentage of patients as per the pain category, adverse reaction, duration of hospital stay, cost-effectiveness, and patient's satisfaction were all taken into consideration when evaluated. RESULTS: Compared to the control group, the visual analogue scale scores for pain intensity was significantly lower at 2, 4, 6, and 12 h (all P < 0.05) in the multimodal group and a noticeably lower prevalence of post-operative nausea and vomiting in the multimodal group (31.0% vs. 59.5%). Patient's satisfaction in the multimodal group was also significantly higher than that in the control group (95.2% vs. 69.0%). No significant difference was observed in the duration of hospital stay between the two groups. CONCLUSION: Preemptive parecoxib and sufentanil-based multimodal analgesia regime is a safe, efficient and cost-effective regimen for postoperative pain control in HCC patients undergoing TACE.


Asunto(s)
Analgesia Controlada por el Paciente , Carcinoma Hepatocelular/terapia , Quimioembolización Terapéutica , Neoplasias Hepáticas/terapia , Dolor Postoperatorio/terapia , Adulto , Anciano , Quimioembolización Terapéutica/efectos adversos , Análisis Costo-Beneficio , Femenino , Costos de la Atención en Salud , Humanos , Isoxazoles/administración & dosificación , Isoxazoles/efectos adversos , Masculino , Persona de Mediana Edad , Satisfacción del Paciente , Náusea y Vómito Posoperatorios/prevención & control , Sufentanilo/administración & dosificación , Sufentanilo/efectos adversos
3.
J Pineal Res ; 63(4)2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28776759

RESUMEN

Severe hypoglycemia has a detrimental impact on the cerebrovasculature, but the molecular events that lead to the disruption of the integrity of the tight junctions remain unclear. Here, we report that the microvessel integrity was dramatically compromised (59.41% of wild-type mice) in TP53-induced glycolysis and apoptosis regulator (TIGAR) transgenic mice stressed by hypoglycemia. Melatonin, a potent antioxidant, protects against hypoglycemic stress-induced brain endothelial tight junction injury in the dosage of 400 nmol/L in vitro. FRET (fluorescence resonance energy transfer) imaging data of endothelial cells stressed by low glucose revealed that TIGAR couples with calmodulin to promote TIGAR tyrosine nitration. A tyrosine 92 mutation interferes with the TIGAR-dependent NADPH generation (55.60% decreased) and abolishes its protective effect on tight junctions in human brain microvascular endothelial cells. We further demonstrate that the low-glucose-induced disruption of occludin and Caludin5 as well as activation of autophagy was abrogated by melatonin-mediated blockade of nitrosative stress in vitro. Collectively, we provide information on the detailed molecular mechanisms for the protective actions of melatonin on brain endothelial tight junctions and suggest that this indole has translational potential for severe hypoglycemia-induced neurovascular damage.


Asunto(s)
Antioxidantes/farmacología , Células Endoteliales/efectos de los fármacos , Melatonina/farmacología , Proteínas/metabolismo , Uniones Estrechas/efectos de los fármacos , Animales , Proteínas Reguladoras de la Apoptosis , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Línea Celular , Humanos , Hipoglucemia/complicaciones , Péptidos y Proteínas de Señalización Intracelular/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Monoéster Fosfórico Hidrolasas , Proteínas/efectos de los fármacos , Uniones Estrechas/metabolismo
4.
Sci Rep ; 9(1): 18248, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31796756

RESUMEN

Vascular dementia (VaD) is a complex disorder caused by reduced blood flow in the brain. However, there is no effective pharmacological treatment option available until now. Here, we reported that low-dose levamlodipine besylate could reverse the cognitive impairment in VaD mice model of right unilateral common carotid arteries occlusion (rUCCAO). Oral administration of levamlodipine besylate (0.1 mg/kg) could reduce the latency to find the hidden platform in the MWM test as compared to the vehicle group. Furthermore, vehicle-treated mice revealed reduced phospho-CaMKII (Thr286) levels in the hippocampus, which can be partially restored by levamlodipine besylate (0.1 mg/kg and 0.5 mg/kg) treatment. No significant outcome on microglia and astrocytes were observed following levamlodipine besylate treatment. This data reveal novel findings of the therapeutic potential of low-dose levamlodipine besylate that could considerably enhance the cognitive function in VaD mice.


Asunto(s)
Amlodipino/administración & dosificación , Demencia Vascular/tratamiento farmacológico , Niacina/análogos & derivados , Nootrópicos/administración & dosificación , Amlodipino/farmacología , Animales , Astrocitos/efectos de los fármacos , Vasos Sanguíneos/efectos de los fármacos , Modelos Animales de Enfermedad , Ratones , Microglía/efectos de los fármacos , Niacina/administración & dosificación , Niacina/uso terapéutico , Nootrópicos/farmacología
5.
Theranostics ; 9(19): 5672-5680, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31534510

RESUMEN

Autophagy is a dynamic and complicated catabolic process. Imaging autophagic flux can clearly advance knowledge of its pathophysiology significance. While the most common way autophagy is imaged relies on fluorescent protein-based probes, this method requires substantial genetic manipulation that severely restricts the application. Small fluorescent probes capable of tracking autophagic flux with good spatiotemporal resolution are highly demanable. Methods: In this study, we developed a small-molecule fluorogenic probe (AFG-1) that facilitates real-time imaging of autophagic flux in both intact cells and live mice. AFG-1 is inspired by the cascading nitrosative and acidic microenvironments evolving during autophagy. It operates over two sequential steps. In the first step, AFG-1 responds to the up-regulated peroxynitrite at the initiation of autophagy by its diphenylamino group being oxidatively dearylated to yield a daughter probe. In the second step, the daughter probe responds to the acidic autolysosomes at the late stage of autophagy by being protonated. Results: This pathway-dependent mechanism has been confirmed first by sequentially sensing ONOO- and acid in aqueous solution, and then by imaging autophagic flux in live cells. Furthermore, AFG-1 has been successfully applied to visualize autophagic flux in real-time in live mice following brain ischemic injury, justifying its robustness. Conclusion: Due to the specificity, easy operation, and the dynamic information yielded, AFG-1 should serve as a potential tool to explore the roles of autophagy under various pathological settings.


Asunto(s)
Autofagia/efectos de los fármacos , Colorantes Fluorescentes/metabolismo , Animales , Línea Celular , Microambiente Celular , Endotelio/metabolismo , Endotelio/patología , Colorantes Fluorescentes/química , Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Sondas Moleculares/química , Sondas Moleculares/metabolismo , Estrés Nitrosativo , Ácido Peroxinitroso/metabolismo
6.
CNS Neurosci Ther ; 23(10): 818-826, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28851042

RESUMEN

AIMS: Vascular dementia (VaD) is a heterogeneous brain disorder for which there are no effective approved pharmacological treatments available. We aimed to evaluate the effect of calmodulin inhibitor, DY-9836, and its loaded nanodrug carrier system on cognitive impairment and gain a better understanding of the protective mechanisms in mice with bilateral carotid artery stenosis (BCAS). RESULTS: DY-9836 (0.5 or 1 mg/kg) or DY-9836 (0.25 mg/kg)-encapsulated polysialic acid-octadecylamine (PSA-ODA) micelles (PSA-ODA/DY) were given to BCAS mice for 4 weeks. Administration of DY-9836 or PSA-ODA/DY reduced escape latency in space exploration and working memory test compared with vehicle group. Vehicle-treated mice showed reduced phospho-CaMKII (Thr286/287) levels in the hippocampus, whereas partially restored by DY-9836 (1 mg/kg) or PSA-ODA/DY (0.25 mg/kg) treatment. In accordance with the pharmacological profile of DY-9836 observed during behavioral studies, experimental molecular and biochemical markers induced by BCAS, such as protein tyrosine nitration, Nod-like receptor protein 3 (NLRP3), caspase-1, and interleukin-1ß, were reduced by DY-9836 and PSA-ODA/DY treatment. CONCLUSIONS: These data disclose novel findings about the therapeutic potential of DY-9836, and its encapsulated nanodrug delivery system significantly enhanced the cognitive function via inhibitory effect on nitrosative stress and NLRP3 signaling in VaD mice.


Asunto(s)
Calmodulina/antagonistas & inhibidores , Estenosis Carotídea/fisiopatología , Demencia Vascular/tratamiento farmacológico , Indazoles/administración & dosificación , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Nootrópicos , Piperazinas/administración & dosificación , Aminas , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Estenosis Carotídea/tratamiento farmacológico , Caspasa 1/metabolismo , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/fisiopatología , Demencia Vascular/fisiopatología , Modelos Animales de Enfermedad , Portadores de Fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Masculino , Ratones Endogámicos C57BL , Micelas , Estrés Nitrosativo/efectos de los fármacos , Estrés Nitrosativo/fisiología , Nootrópicos/administración & dosificación , Fosforilación , Ácidos Siálicos
7.
Mol Neurobiol ; 53(4): 2600-9, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26093380

RESUMEN

Disturbance of neuregulin-1ß/ErbB4 signaling is considered to be associated with brain ischemia, but the mechanisms of this disruption are largely unknown. In the present study, we provide evidence that degradation of ErbB4 is involved in neuronal cell death in response to ischemia. Our data showed that the application of neuregulin-1ß provided significant protection against oxygen-glucose deprivation (OGD)-induced neuronal death as detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, annexin V/propidium iodide flow cytometry analysis and terminal deoxynucleotidyl transferase (TdT) dUTP nick end labeling (TUNEL) staining. Furthermore, neuregulin-1ß treatment significantly reduced the infarct volume of ischemic mice, and this result was not seen in the ErbB4 knockout mice. We found that brain ischemia induced the breakdown of ErbB4 in a time-dependent manner in vivo, but not that of ErbB2. In vitro studies further indicated that recombinant calpain induced the cleavage of ErbB4 in a dose-dependent way, whereas the calpain inhibitor significantly reduced the OGD-induced ErbB4 breakdown. Additionally, OGD-induced apoptosis was partially abolished by transfection with the ErbB4E872K mutant. Taken together, neuregulin-1ß elicits its neuroprotective effect in an ErbB4-dependent manner, and the cleavage of ErbB4 by calpain contributes to a neuronal cell death cascade during brain ischemia.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Calpaína/metabolismo , Neuronas/metabolismo , Neuronas/patología , Receptor ErbB-4/metabolismo , Animales , Isquemia Encefálica/tratamiento farmacológico , Muerte Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Activación Enzimática/efectos de los fármacos , Glucosa/deficiencia , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Neurregulina-1/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Oxígeno , Transfección
8.
ACS Appl Mater Interfaces ; 8(51): 35045-35058, 2016 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-27750011

RESUMEN

Clinical treatment for vascular dementia still remains a challenge mainly due to the blood-brain barrier (BBB). Here, a micelle based on polysialic acid (PSA), which is a hydrophilic and endogenous carbohydrate polymer, was designed to deliver calmodulin antagonist for therapy of vascular dementia. PSA was first chemically conjugated with octadecylamine (ODA), and the obtained PSA-ODA copolymer could self-assemble into micelle in aqueous solution with a 120.0 µg/mL critical micelle concentration. The calmodulin antagonist loaded PSA-ODA micelle, featuring sustained drug release behavior over a period of 72 h with a 3.6% (w/w) drug content and a 107.0 ± 4.0 nm size was then fabricated. The PSA-ODA micelle could cross the BBB mainly via active endocytosis by brain endothelial cells followed by transcytosis. In a water maze test for spatial learning, calmodulin antagonist loaded PSA-ODA micelle significantly reduced the escape latencies of right unilateral common carotid arteries occlusion (rUCCAO) mice with dosage significantly reduced versus free drug. The decrease of hippocampal phospho-CaMKII (Thr286/287) and phospho-synapsin I (Ser603) was partially restored in rUCCAO mice following calmodulin antagonist loaded PSA-ODA micelle treatment. Consistent with the restored CaMKII phosphorylation, the elevation of BrdU/NeuN double-positive cells in the same context was also observed. Overall, the PSA-ODA micelle developed from the endogenous material might promote the development of therapeutic approaches for improving the efficacy of brain-targeted drug delivery and have great potential for vascular dementia treatment.


Asunto(s)
Ácidos Siálicos/química , Animales , Calmodulina , Demencia Vascular , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Ratones , Micelas , Polímeros
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA