Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Drug Deliv Transl Res ; 8(5): 1406-1420, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30117120

RESUMEN

Curcumin (CUR), a natural polyphenolic compound, is considered as one of the most potential candidates against Alzheimer disease (AD) by targeting multiple pathologies such as amyloid-beta, tau phosphorylation, and oxidative stress. Poor physicochemical profile and oral bioavailability (BA) are the major contributors to its failure in clinical trials. Lack of success in numerous drug clinical trials for the treatment of AD urges the need of repositioning of CUR. To overcome its limitation and enhance oral BA, Novel CUR Formulation (NCF) was developed using self-nanomicellizing solid dispersion strategy which displayed 117-fold enhancement in oral BA of CUR. NCF was tested using SH-SY5Y695 APP human neuroblastoma cell line against the cytotoxicity induced by copper metal ion, H2O2, and Aß42 oligomer and compared with CUR control. The safety and efficacy of NCF on mice AD-like behavioral deficits (open field, novel objective recognition, Y-maze, and Morris water maze tests) were assessed in transgenic AD (APPSwe/PS1deE9) mice model. In SH-SY5Y695 APP human neuroblastoma cell line, NCF showed better safety and efficacy against the cytotoxicity due to the significantly enhancement of cellular uptake. It not only prevents the deterioration of cognitive functions of the aged APPSwe/PS1deE9 mice during aging but also reverses the cognitive functions to their much younger age which is also better than the currently available approved options. Moreover, NCF was proved as well tolerated with no appearance of any significant toxicity via oral administration. The results of the study demonstrated the potential of NCF to improve the efficacy of CUR without compromising its safety profile, and pave the way for clinical development for AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Disfunción Cognitiva/prevención & control , Curcumina/administración & dosificación , Nanoestructuras/química , Administración Oral , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/toxicidad , Animales , Línea Celular , Cobre/toxicidad , Curcumina/efectos adversos , Curcumina/química , Modelos Animales de Enfermedad , Composición de Medicamentos , Humanos , Peróxido de Hidrógeno/toxicidad , Masculino , Ratones , Ratones Transgénicos , Micelas , Fragmentos de Péptidos/toxicidad
2.
Drug Des Devel Ther ; 12: 2111-2128, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30022810

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is a devastating neurodegenerative disorder that lacks any disease-modifying drug for the prevention and treatment. Edaravone (EDR), an approved free radical scavenger, has proven to have potential against AD by targeting multiple key pathologies including amyloid-beta (Aß), tau phosphorylation, oxidative stress, and neuroinflammation. To enable its oral use, novel edaravone formulation (NEF) was previously developed. The aim of the present investigation was to evaluate safety and efficacy of NEF by using in vitro/in vivo disease model. MATERIALS AND METHODS: In vitro therapeutic potential of NEF over EDR was studied against the cytotoxicity induced by copper metal ion, H2O2 and Aß42 oligomer, and cellular uptake on SH-SY5Y695 amyloid-ß precursor protein (APP) human neuroblastoma cell line. For in vivo safety and efficacy assessment, totally seven groups of APP/PS1 (five treatment groups, one each as a basal and sham control) and one group of C57BL/6 mice as a positive control for behavior tests were used. Three groups were orally treated for 3 months with NEF at an equivalent dose of EDR 46, 138, and 414 µmol/kg, whereas one group was supplied with each Donepezil (5.27 µM/kg) and Soluplus (amount present in NEF of 414 µmol/kg dose of EDR). Behavior tests were conducted to assess motor function (open-field), anxiety-related behavior (open-field), and cognitive function (novel objective recognition test, Y-maze, and Morris water maze). For the safety assessment, general behavior, adverse effects, and mortality were recorded during the treatment period. Moreover, biochemical, hematological, and morphological parameters were determined. RESULTS: Compared to EDR, NEF showed superior cellular uptake and neuroprotective effect in SH-SY5Y695 APP cell line. Furthermore, it showed nontoxicity of NEF up to 414 µM/kg dose of EDR and its potential to reverse AD-like behavior deficits of APP/PS1 mice in a dose-dependent manner. CONCLUSION: Our results indicate that oral delivery of NEF holds a promise as a safe and effective therapeutic agent for AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Modelos Animales de Enfermedad , Edaravona/uso terapéutico , Nanopartículas/química , Fármacos Neuroprotectores/uso terapéutico , Administración Oral , Animales , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Edaravona/administración & dosificación , Edaravona/química , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Micelas , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/química
3.
Oncotarget ; 7(48): 78813-78826, 2016 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-27705931

RESUMEN

MicroRNA128-1 (miR128-1), as a brain-specific miRNA, is downregulated in glioblastoma multiforme (GBM) and closely associated with the progression of GBM. However, the underlying molecular mechanism of the downregulation and its role in the regulation of tumorigenesis and anticancer drug resistance in GBM remains largely unknown. In the current study,we found that miR128-1 was downregulated in GBM and glioma stem-like cells (GSCs). Intriguingly, treatment with the DNA methylation inhibitors 5-Aza-CdR (Aza) and 4-phenylbutyric acid (PBA) resulted in miR128-1 upregulation in both GBM cells and GSCs. Either forced expression of miR128-1 or Aza/PBA treatment inhibited tumor cell proliferation, migration and invasion in vitro. Moreover, overexpression of miR128-1 inhibited the growth of transplant tumor in vivo. BMI1 and E2F3 were found to be direct targets of miR128-1 and downregulated by miR128-1 in vitro and in vivo. Our results revealed a mechanism of methylation that controls miR128-1 expression in GBM cells and GSCs and indicate miR128-1 could function as a tumor suppressor in GBM by negatively regulating tumor cell proliferation, invasion and self-renewal through direct targeting BMI1 and E2F3. Our findings suggest that DNA methylation inhibitors are potential agents for GBM treatment by upregulating miR-128-1.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Proliferación Celular , Factor de Transcripción E2F3/metabolismo , Glioblastoma/metabolismo , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Animales , Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Autorrenovación de las Células , Metilación de ADN , Metilasas de Modificación del ADN/antagonistas & inhibidores , Metilasas de Modificación del ADN/metabolismo , Factor de Transcripción E2F3/genética , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/patología , Humanos , Masculino , Ratones Desnudos , MicroARNs/genética , Invasividad Neoplásica , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Complejo Represivo Polycomb 1/genética , Transducción de Señal , Factores de Tiempo , Transcripción Genética , Transfección
4.
Oncol Rep ; 26(5): 1305-13, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21769436

RESUMEN

In the present study, we investigated the prognostic roles of the O6-methylguanine-DNA methyltransferase (MGMT) gene methylation status, the protein profiles of MGMT, and the glioma stem cell (GSC) marker CD133 in malignant glioma resistance to radiotherapy. The proliferation of glioma cells was assessed using a clonogenic survival assay and flow cytometry. CD133 expression was assessed in SHG-44-GSCs using RT-PCR and flow cytometry. MGMT exhibited resistance to radiation in the SHG-44-GSCs using siRNA transfection. The effects of the siRNA on mRNA and protein expression of MGMT in SHG-44-GSCs were detected using semi-quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blotting. MGMT methylation status, MGMT and CD133 expression profiles were assessed in 59 malignant glioma patients using methylation-specific polymerase chain reaction (MSP), and immunohistochemistry. In vitro, SHG-44-GSCs exhibited a characteristic resistance to radiation that was not observed in SHG-44 cells. This resistance was attributed to the unmethylated status of the MGMT promoter and to high expression levels of MGMT mRNA in the glioma cells. In these patients, the CD133 marker, but not MGMT promoter methylation or MGMT protein level, was associated with resistance to radiotherapy (n=59; hazard ratio=2.838; 95% CI, 1.725-7.597; p=0.001). The median progression-free survival (PFS) among patients with the CD133 marker was 14 months, whereas it was 35 months in patients without CD133 (p=0.001). Notably, co-expression of the methylated MGMT promoter and the CD133 marker was associated with the poorest outcome in patients with gliomas treated by radiotherapy; in these patients, PFS was 7 months. These results suggest that assessment of GSC MGMT and CD133 levels will guide future clinical targeted therapies and stratify glioma patient treatment regimens. High expression levels of the CD133 protein could be used as a predictor for poor survival in patients treated with radiotherapy.


Asunto(s)
Antígenos CD/biosíntesis , Biomarcadores de Tumor/biosíntesis , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/radioterapia , Metilasas de Modificación del ADN/biosíntesis , Enzimas Reparadoras del ADN/biosíntesis , Glioblastoma/metabolismo , Glioblastoma/radioterapia , Glicoproteínas/biosíntesis , Proteínas Supresoras de Tumor/biosíntesis , Antígeno AC133 , Adulto , Anciano , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Quimioradioterapia Adyuvante , Metilación de ADN , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Femenino , Glioblastoma/genética , Glioblastoma/patología , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Péptidos , Pronóstico , Regiones Promotoras Genéticas , Tolerancia a Radiación , Transfección , Proteínas Supresoras de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...