Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(3)2022 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-35163790

RESUMEN

Aurora A kinase (Aurora A) is a serine/threonine kinase regulating control of multiple events during cell-cycle progression. Playing roles in promoting proliferation and inhibiting cell death in cancer cells leads Aurora A to become a target for cancer therapy. It is overexpressed and associated with a poor prognosis in ovarian cancer. Improving cisplatin therapy outcomes remains an important issue for advanced-stage ovarian cancer treatment, and Aurora A inhibitors may improve it. In the present study, we identified natural compounds with higher docking scores than the known Aurora A ligand through structure-based virtual screening, including the natural compound fangchinoline, which has been associated with anticancer activities but not yet investigated in ovarian cancer. The binding and inhibition of Aurora A by fangchinoline were verified using cellular thermal shift and enzyme activity assays. Fangchinoline reduced viability and proliferation in ovarian cancer cell lines. Combination fangchinoline and cisplatin treatment enhanced cisplatin-DNA adduct levels, and the combination index revealed synergistic effects on cell viability. An in vivo study showed that fangchinoline significantly enhanced cisplatin therapeutic effects in OVCAR-3 ovarian cancer-bearing mice. Fangchinoline may inhibit tumor growth and enhance cisplatin therapy in ovarian cancer. This study reveals a novel Aurora A inhibitor, fangchinoline, as a potentially viable adjuvant for ovarian cancer therapy.


Asunto(s)
Aurora Quinasa A/metabolismo , Bencilisoquinolinas/administración & dosificación , Cisplatino/administración & dosificación , Aductos de ADN/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Animales , Aurora Quinasa A/química , Bencilisoquinolinas/química , Bencilisoquinolinas/farmacología , Línea Celular Tumoral , Cisplatino/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Modelos Moleculares , Simulación del Acoplamiento Molecular , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Conformación Proteica , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Pharm Biol ; 60(1): 75-86, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34962429

RESUMEN

CONTEXT: The mechanism of tetrandrine (TET) in hepatocellular carcinoma (HCC) progression and sorafenib (Sora) chemosensitivity deserves investigation. OBJECTIVE: Using network pharmacology approaches to elucidate the mechanisms of TET in HCC. MATERIALS AND METHODS: CCK-8, colony formation, and flow cytometry assays were used to measure cell phenotypes. BALB/c nude mice were divided into Control, Sora (10 mg/kg), TET (50 mg/kg), and TET + Sora (10 mg/kg Sora plus 50 mg/kg TET) groups to evaluate the antitumor effects of TET for 21 days. Sora and TET were given by intraperitoneal injection or oral gavage. RESULTS: For SMMC7721 (IC50 = 22.5 µM) and PLC8024 (IC50 = 18.4 µM), TET (10, 20 µM) reduced colony number (0.68 ± 0.04- and 0.50 ± 0.04-fold, 0.56 ± 0.04- and 0.42 ± 0.02-fold), induced cell cycle arrest at G0/G1 stage (1.22 ± 0.03- and 1.39 ± 0.07-fold, 1.37 ± 0.06- and 1.55 ± 0.05-fold), promoted apoptosis (2.49 ± 0.26- and 3.63 ± 0.33-fold, 2.74 ± 0.42- and 3.73 ± 0.61-fold), and inactivated PI3K/AKT/mTOR signalling. Sora (10 µM) decreased cell proliferation, enhanced apoptosis, and inhibited PI3K/AKT/mTOR signalling, and these effects were further aggravated in the combination group. Activating PI3K/AKT/mTOR reversed the effects of TET on cell proliferation and Sora sensitivity. In the combination group, tumour volumes and weights were decreased to 202.3 ± 17.4 mm3 and 151.5 ± 25.8 mg compared with Sora (510.6 ± 48.2 mm3 and 396.7 ± 33.5 mg). DISCUSSION AND CONCLUSIONS: TET enhances Sora sensitivity by inactivating PI3K/AKT/mTOR, suggesting the potential of TET as a chemosensitizer in HCC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Bencilisoquinolinas/administración & dosificación , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Humanos , Concentración 50 Inhibidora , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Farmacología en Red , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sorafenib/administración & dosificación , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Immunopharmacol Immunotoxicol ; 43(6): 749-757, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34591732

RESUMEN

OBJECTIVE: Cerebral infarction (CI) is the mayor reason of death in China. Reperfusion is the only immediate treatment for acute cerebral infarction. However, blood reperfusion recovery may cause ischemia-reperfusion (I/R) injuries. The purpose of this study was to investigate the effects of Tetrandrine (TTD) and 3-n-Butylphthalide (NBP) on cerebral I/R injury. MATERIALS AND METHODS: I/R was used to establish CI model in vivo. TTD was performed to analyze cerebral infarction volume. OGD was applied to establish CI model in vitro. Flow cytometry and TUNEL assays were utilized to determine the cell death. ELISA was conducted to determine the release of cytokines. mRNA and protein expressions were detected using qRT-PCR and western blot. RESULTS: We found that NBP + TTD treatment significantly reduced cerebral infarction volume and inhibited the death of neurons in vivo. Moreover, NBP + TTD treatment suppressed the apoptosis and inflammatory response of neurons in vitro. Additionally, NBP + TTD suppressed the expression of activator transcription factor 2 (ATF2). However, overexpression of ATF2 contributed to the degeneration of neurons. Moreover, ATF2 transcriptionally activated Toll-like receptor 4 (TLR4). NBP + TTD inactivated ATF2/TLR4 signaling. CONCLUSIONS: Taken together, TTD combined with NBP protected against cerebral infarction by inhibiting the inflammatory response and neuronal cell apoptosis via inactivating ATF2/TLR4 signaling pathways. This may provide an alternative for I/R injury.


Asunto(s)
Factor de Transcripción Activador 2/antagonistas & inhibidores , Benzofuranos/administración & dosificación , Bencilisoquinolinas/administración & dosificación , Isquemia Encefálica/prevención & control , Daño por Reperfusión/prevención & control , Receptor Toll-Like 4/antagonistas & inhibidores , Factor de Transcripción Activador 2/metabolismo , Animales , Isquemia Encefálica/metabolismo , Células Cultivadas , Quimioterapia Combinada , Masculino , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/administración & dosificación , Daño por Reperfusión/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Receptor Toll-Like 4/metabolismo
4.
Environ Toxicol ; 36(4): 460-471, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33156559

RESUMEN

BACKGROUND: Neferine (NEF) is nontoxic, bisbenzylisoquinoline alkaloid is derived from the seed embryo of lotus, a familiar medicinal plant. Although several mechanisms have been planned, an evident antitumor action pathway of NEF on the oral tumor is still not known. In the current study, we aimed at investigating the protecting effect of NEF against experimental oral carcinoma and clarify its possible mechanism through the induction of apoptosis, proliferation, and inflammatory signaling pathways. METHODS: The experimental hamsters were divided into four groups (I-IV) containing six hamsters each. The group I was control group, group II and III hamsters treated with 7,12-dimethylbenz(a)anthracene (DMBA) (0.5%) alone, thrice in a week for 10 weeks, and group III and IV hamsters received oral supplementation of NEF at a concentration of 15 mg/kg bw. All the hamsters were sacrificed after 16 weeks. RESULTS: Our results revealed that DMBA treated hamsters exhibited 100% oral tumor cell formation with high-tumor incidence (TI), tumor number (TN), tumor volume (TV), decreased levels of antioxidants, increased status of lipid peroxidation (LPO), and modulated the activities of liver marker agents as well as NF-kB, cell proliferation (PCNA), and p53 proteins. NEF supplementation in DMBA treated hamsters, resulted in delayed lesion synthesis, and brought back the levels of the biochemical parameters. In addition, immunostaining of NF-kB, PCNA, and p53 showed that they were inhibited by NEF. CONCLUSION: Thus, NEF might be considered a better chemopreventive drug in an experimental model of home-based primary care (HBPC). More research is necessary to study other pathways implicated in oral carcinomas and their modulation by NEF.


Asunto(s)
Anticarcinógenos/farmacología , Bencilisoquinolinas/farmacología , Carcinogénesis/efectos de los fármacos , Carcinoma de Células Escamosas/prevención & control , Medicamentos Herbarios Chinos/farmacología , Neoplasias de la Boca/prevención & control , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , Anticarcinógenos/administración & dosificación , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Bencilisoquinolinas/administración & dosificación , Carcinoma de Células Escamosas/patología , Proliferación Celular/efectos de los fármacos , Cricetinae , Medicamentos Herbarios Chinos/administración & dosificación , Células Epiteliales/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Masculino , Neoplasias de la Boca/patología , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo
5.
Biochem Biophys Res Commun ; 531(4): 573-580, 2020 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-32811645

RESUMEN

Taxol resistance led to the poor survival prognosis in advanced nasopharyngeal carcinoma (NPC). Epithelial-mesenchymal transition (EMT) plays an important role in tumor chemoresistance. Neferine (NEF) is found to sensitize the cancer cells to chemotherapeutic agents, but its effects and mechanisms on NPC Taxol resistance is unclear. In this study, we discovered that Taxol-resistant cell lines 5-8F/Taxol and CNE-1/Taxol had the greater ability to metastasis and the higher expression of EMT markers. Then we found that NEF could inhibit the viability and EMT process in the Taxol-resistant cell lines. Furthermore, we confirmed that NEF could augment therapeutic efficacy of Taxol on NPC Taxol-resistant cell lines. Further through Microarray based analysis, we found that miR-130b-5p was stably down-regulated after treating 5-8F/Taxol with NEF. Later we verified that up-regulation of miR-130b-5p could not only promote the EMT-related migration/invasion, but also impair the inhibition effects of NEF on the EMT-associated metastatic ability and the chemotherapy resistance to Taxol. In conclusion, our results firstly suggested that NEF may enhanced Taxol sensitivity in NPC Taxol-resistant cell lines through inhibition of EMT which mediated by miR-130b-5p downregulation in vitro and in vivo. NEF may be used as a Taxol sensitizer in chemotherapy of NPC.


Asunto(s)
Bencilisoquinolinas/farmacología , Resistencia a Antineoplásicos/genética , MicroARNs/genética , Carcinoma Nasofaríngeo/tratamiento farmacológico , Neoplasias Nasofaríngeas/tratamiento farmacológico , Paclitaxel/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Bencilisoquinolinas/administración & dosificación , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones Endogámicos BALB C , Carcinoma Nasofaríngeo/genética , Neoplasias Nasofaríngeas/genética , Paclitaxel/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Mol Pharm ; 17(5): 1596-1607, 2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32142292

RESUMEN

Pulmonary fibrosis (PF) is a kind of interstitial lung disease with the features of progressive and often fatal dyspnea. Tetrandrine (TET) is the major active constituent of Chinese herbal Stephania tetrandra S. Moore, which has already applied clinically to treat rheumatism, lung cancer, and silicosis. In this work, a tetrandrine-hydroxypropyl-ß-cyclodextrin inclusion compound (TET-HP-ß-CD) was developed for the treatment of pulmonary fibrosis via inhalation administration. TET-HP-ß-CD was prepared by the freeze-drying method and identified using the cascade impactor, differential scanning calorimetry (DSC), X-ray diffraction (XRD), and Fourier transform infrared spectrum (FT-IR). A bleomycin-induced pulmonary fibrosis rat model was used to assess the effects of inhaled TET and TET-HP-ß-CD. Animal survival, hydroxyproline content in the lungs, and lung histology were detected. The results showed that inhalation of TET-HP-ß-CD alleviated inflammation and fibrosis, limited the accumulation of hydroxyproline in the lungs, regulated protein expression in PF development, and improved postoperative survival. Moreover, nebulized delivery of TET-HP-ß-CD accumulated chiefly in the lungs and limited systemic distribution compared with intravenous administration. The present results indicated that inhalation of TET-HP-ß-CD is an attractive candidate for the treatment of pulmonary fibrosis.


Asunto(s)
2-Hidroxipropil-beta-Ciclodextrina/química , Bencilisoquinolinas/química , Fibrosis Pulmonar/tratamiento farmacológico , 2-Hidroxipropil-beta-Ciclodextrina/administración & dosificación , 2-Hidroxipropil-beta-Ciclodextrina/farmacocinética , Administración por Inhalación , Animales , Bencilisoquinolinas/administración & dosificación , Bencilisoquinolinas/farmacocinética , Modelos Animales de Enfermedad , Pulmón/metabolismo , Pulmón/patología , Masculino , Fibrosis Pulmonar/mortalidad , Fibrosis Pulmonar/patología , Ratas , Ratas Sprague-Dawley , Solubilidad , Espectroscopía Infrarroja por Transformada de Fourier , Factor de Necrosis Tumoral alfa/análisis
7.
J Biochem Mol Toxicol ; 34(2): e22429, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31860774

RESUMEN

Alzheimer's disease (AD) is an age-associated neurodegenerative disease, which is developed by oxidative stress and acetylcholine contraction in the synaptic cleft of the neurons. This leads to dementia, memory loss, and decrease in learning ability and orientation. In this research work, we aimed to explore the neuroprotective effect of neferine on AlCl3 -induced AD in rats. The results of our study revealed that the increased reactive oxygen species (ROS) and nitric oxide in the hippocampus leads to the development of AD in the rats. The oral treatment of neferine done the following occurrences such as; it potentially inhibited the ROS formation and acts as a scavenging molecule by preventing the neurodegeneration. It also improved the memory and learning ability to complete the maze activity in the AD rats and significantly increased the antioxidants superoxide dismutase, catalase, and reduced glutathione in neferine treated AD rats. It aggressively declined the activity of acetylcholine esterase and Na+ K+ ATPase in the neurodegenerative rat models. The gene expression pattern of neuroinflammatory cytokines such as tumor necrosis factor α (TNF-α), interleukin-6 (IL-6), and interleukin-1ß (IL-1ß) were decreased in the neferine-treated rats. The neuroinflammatory proteins such as inducible nitric oxide (iNOS), cyclooxygenase-2 (COX-2), and nuclear factor kappa ß (Nf-κß) were decreased and Nf-κß inhibitor IKBα was increased in the neferine-treated AD rats. Finally, the histology study proved that the neferine treatment possibly prevents neurodegeneration in the hippocampus tissue of the AD models. Hence, these all findings concluded that the neferine could be a potential neuropreventive as well as neurodegenerative therapeutic compound in neurological and cognitive dysfunction.


Asunto(s)
Cloruro de Aluminio/farmacología , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/tratamiento farmacológico , Bencilisoquinolinas/farmacología , Medicamentos Herbarios Chinos/farmacología , Fármacos Neuroprotectores/farmacología , Nootrópicos/farmacología , Administración Oral , Cloruro de Aluminio/efectos adversos , Animales , Conducta Animal/efectos de los fármacos , Bencilisoquinolinas/administración & dosificación , Ciclooxigenasa 2/metabolismo , Citocinas/genética , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/administración & dosificación , Femenino , Expresión Génica/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Peroxidación de Lípido/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , FN-kappa B/metabolismo , Fármacos Neuroprotectores/administración & dosificación , Óxido Nítrico Sintasa de Tipo II/metabolismo , Nootrópicos/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar
8.
J Nanobiotechnology ; 18(1): 61, 2020 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-32306970

RESUMEN

Primary intracerebral hemorrhage (ICH) is a leading cause of long-term disability and death worldwide. Drug delivery vehicles to treat ICH are less than satisfactory because of their short circulation lives, lack of specific targeting to the hemorrhagic site, and poor control of drug release. To exploit the fact that metal ions such as Fe2+ are more abundant in peri-hematomal tissue than in healthy tissue because of red blood cell lysis, we developed a metal ion-responsive nanocarrier based on a phosphonated calix[4]arene derivative in order to deliver the neuroprotective agent dauricine (DRC) specifically to sites of primary and secondary brain injury. The potential of the dauricine-loaded nanocarriers for ICH therapy was systematically evaluated in vitro and in mouse models of autologous whole blood double infusion. The nanocarriers significantly reduced brain water content, restored blood-brain barrier integrity and attenuated neurological deficits by inhibiting the activation of glial cells, infiltration by neutrophils as well as production of pro-inflammatory factors (IL-1ß, IL-6, TNF-α) and matrix-metalloprotease-9. These results suggest that our dauricine-loaded nanocarriers can improve neurological outcomes in an animal model of ICH by reducing inflammatory injury and inhibiting apoptosis and ferroptosis.


Asunto(s)
Bencilisoquinolinas/química , Calixarenos/química , Hemorragia Cerebral/patología , Portadores de Fármacos/química , Metales/química , Nanoestructuras/química , Fármacos Neuroprotectores/química , Fenoles/química , Tetrahidroisoquinolinas/química , Animales , Bencilisoquinolinas/administración & dosificación , Bencilisoquinolinas/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Hemorragia Cerebral/tratamiento farmacológico , Modelos Animales de Enfermedad , Humanos , Interleucina-1beta/metabolismo , Iones/química , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Organofosfonatos/química , Especies Reactivas de Oxígeno/metabolismo , Tetrahidroisoquinolinas/administración & dosificación , Tetrahidroisoquinolinas/farmacología
9.
J Microencapsul ; 37(5): 384-393, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32349635

RESUMEN

Aim: The aim of this study was to improve solubility and antitumour ability in vitro of tetrandrine (Tet) via preparing nanosuspensions (NSs).Methods: The Tet-NSs were prepared by wet media milling. The Tet-CCS-NS was prepared with croscarmellose sodium (CCS) as single stabiliser. The Tet-HACC-TPGS-NS was manufactured with D-α-tocopheryl polyethylene glycol 1,000 succinate (TPGS) and hydroponically trimethyl ammonium chloride chitosan (HACC) as combined stabilisers. Physicochemical properties of the NSs such as particle size, surface morphologies, crystallinity and molecular interactions were investigated. In addition, the in vitro dissolution and antitumour activities using A549 human lung cancer cells were evaluated.Results: The mean particle sizes and Zeta potential of freshly prepared Tet-CCS-NS, Tet-HACC-TPGS-NS were 469.1 ± 14nm and 157.3 ± 5nm, -29.4 ± 0.26 mV and 23.3 ± 0.36 mV, respectively. In comparison to pure Tet, the cumulative dissolution of Tet-NSs were increased by 4 ∼ 5 times in 2 h. In vitro antitumour studies on Tet- NSs in A549 cells, the cell survival rate of the Tet-NSs at high concentration (30-50µg/ml) were less than 10% within 48 h. Meanwhile, Tet-NSs were revealed to induce A549 cells apoptosis and promote cell uptake.Conclusion: The present study has proved that the Tet-NSs can increase Tet solubility as well as improve Tet antitumour activity in vitro.


Asunto(s)
Antineoplásicos/farmacología , Bencilisoquinolinas/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Células A549 , Apoptosis , Rastreo Diferencial de Calorimetría , Carboximetilcelulosa de Sodio/química , Línea Celular Tumoral , Humanos , Nanomedicina/métodos , Tamaño de la Partícula , Compuestos de Amonio Cuaternario/química , Solubilidad , Espectroscopía Infrarroja por Transformada de Fourier , Vitamina E/química , Difracción de Rayos X
10.
J Cell Biochem ; 120(3): 3101-3113, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30485494

RESUMEN

Berbamine (BBM), a bisbenzylisoquinoline alkaloid from roots, bark, and stem of Berberis plant such as Berberis aristata has a wide range of pharmacological activities. However, the evidence for the cardioprotective effect of BBM is inadequate and the molecular mechanism of BBM remains unclear. This study investigated the underlying molecular mechanism of BBM-mediated cardioprotection on isoproterenol (ISO)-induced mitochondrial dysfunction and apoptosis in rats. The assays of mitochondria antioxidant status, mitochondrial marker enzymes, and electron microscopic analysis of mitochondria revealed BBM significantly prevented the mitochondrial dysfunction induced by ISO. The ISO-induced elevation of mitochondrial oxidative stress was also curbed by BBM. Furthermore, pretreatment with BBM protected the heart tissue from ISO-induced apoptosis as evident from decreased terminal dUTP nickend-labeling positive cells and decreased expression of Bax, cytochrome c, cleaved caspase-9, and caspase-3, and poly (ADP-ribose) polymerase and increased expression of Bcl-2 in ISO-induced rats. These current findings suggest that BBM exerts a significant cardioprotective effect on ISO-induced myocardial infarction in rats.


Asunto(s)
Bencilisoquinolinas/administración & dosificación , Isoproterenol/efectos adversos , Mitocondrias/efectos de los fármacos , Infarto del Miocardio/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Bencilisoquinolinas/farmacología , Masculino , Mitocondrias/metabolismo , Infarto del Miocardio/inducido químicamente , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar
11.
J Liposome Res ; 29(1): 21-34, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29166813

RESUMEN

Brain glioma is one of the most common and devastating intracranial malignancies with a high mortality. Chemotherapy for brain glioma is not ideal due to blood brain barrier (BBB) and multidrug resistance (MDR). The objectives of the present study were to develop a kind of RGD (Arg-Gly-Asp) tripeptide modified vinorelbine plus tetrandrine liposomes to achieve BBB transportation, MDR reversion and glioma cell targeting simultaneously. The studies were performed on glioma cells, resistant glioma cells and glioma-bearing mice. Results showed that the constructed liposomes with suitable physicochemical properties could significantly enhance the transport across BBB, obviously accumulate in glioma cells, and exhibit evident capabilities in diminishing brain glioma in mice. Action mechanism studies indicated that the enhanced anticancer efficacy could be attribute to the follows: prolonged elimination half-life (7.093 ± 1.311 h); increased AUC0-24 h (28.92 ± 2.66 mg/L*h); transporting across BBB; enhanced cellular uptake; down-regulation on P-gp (0.49 ± 0.06 fold); inducing apoptosis via activating caspase 8, 9, and 3 (2.40 ± 0.22, 3.57 ± 0.29, and 4.33 ± 0.30 folds, respectively). In conclusion, the RGD modified vinorelbine plus tetrandrine liposomes may offer a promising therapeutic strategy for treatment of brain glioma.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Bencilisoquinolinas/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Resistencia a Antineoplásicos/efectos de los fármacos , Glioma/tratamiento farmacológico , Liposomas , Oligopéptidos , Vinorelbina/administración & dosificación , Animales , Barrera Hematoencefálica/efectos de los fármacos , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Humanos , Liposomas/química , Ratones
12.
J Microencapsul ; 36(4): 356-370, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31190597

RESUMEN

Here, we have successfully synthesised and purified multifunctional PLGA-based nanoparticles by the co-encapsulation of an anticancer drug (tetrandrine) and a magnetic material (Fe3O4). The obtained Tet-Fe3O4-PLGA NPs had a uniform spherical shape with a particle size of approximately 199 nm and a negative surface charge of -18.0 mV, displaying a high encapsulation efficiency. Furthermore, TEM studies provided representative images of the purification process of the magnetic nanoparticles with MACS® technology. The MFM and VSM results indicated that both the Fe3O4 NPs and Tet-Fe3O4-PLGA NPs were superparamagnetic. The DSC spectrum demonstrated that Tet was successfully encapsulated within the PLGA-based nanoparticles. Significantly, the release studies revealed NPs had a relatively slower release rate than free Tet after 8 h's initial burst release, which had decreased from 98% to 65% after 24 h. In vitro cellular studies revealed that NPs could effectively penetrate into A549 cells and A549 multicellular spheroids to exert cytotoxicity, displaying a significantly high anti-proliferation effect. Moreover, western blot demonstrated that the co-loaded NPs had a higher anticancer activity by injuring lysosomes to activate the mitochondria pathway and induce A549 cell apoptosis. The magnetic characteristics and high anticancer activity support the use of Tet/Fe3O4 co-loaded PLGA-based nanoparticles as a promising strategy in the treatment of lung cancer.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Bencilisoquinolinas/administración & dosificación , Portadores de Fármacos/química , Nanopartículas de Magnetita/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Células A549 , Antineoplásicos Fitogénicos/farmacocinética , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Bencilisoquinolinas/farmacocinética , Bencilisoquinolinas/farmacología , Liberación de Fármacos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico
13.
Biochem Biophys Res Commun ; 495(1): 1516-1521, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29197576

RESUMEN

Neferine, an alkaloid isolated from Lotus seeds, displays multiple pharmacological effects that counter cancer, oxidants, and arrhythmia. It was initially identified as a strong inducer for macroautophagy in cancer cells by suppressing AMPK/mTOR signaling. In this study, we found that autophagy signaling was inhibited in the condition of neferine treatment. Exposure to neferine resulted in the accumulation of LC3-II and an associated adaptor protein, p62/SQSTM1. Knockdown of ATG5 failed to reduce the accumulation of LC3-II induced by neferine. The electron microscopy (EM) images showed that neferine induce accumulation of multi-vesicle bodies (MVB) and failure of lysosome maturation. Moreover, exposure to neferine reduced maturation of cathepsin D and impaired the degradation of autophagic and phagocytic cargos. Rather than stimulate autophagic flux, the data indicate that neferine impaired lysosomes to block degradation within phagolysosomes.


Asunto(s)
Autofagia/efectos de los fármacos , Autofagia/fisiología , Bencilisoquinolinas/administración & dosificación , Lisosomas/fisiología , Lisosomas/ultraestructura , Proteínas Asociadas a Microtúbulos/metabolismo , Antioxidantes/administración & dosificación , Relación Dosis-Respuesta a Droga , Células HEK293 , Células HeLa , Humanos , Lisosomas/efectos de los fármacos , Análisis de Flujos Metabólicos
14.
Pharmazie ; 73(9): 541-544, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30223939

RESUMEN

This study aimed to evaluate the anti-inflammatory mechanism of neferine, a bisbenzylisoquinoline alkaloid, on the lipopolysaccharide (LPS)-induced inflammation in Human Umbilical Endothelial Cells (HUVECs) and pulmonary aorta cells. Production of pro-inflammatory cytokines and nitric oxide (NO) was determined using Griess reaction in human endothelial cells. The levels of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and mitogen-activated protein kinases (MAPKs) were analyzed using real time PCR and western blotting. Neferine significantly prevented the NO, TNF-α, COX-2, iNOS, IL-1B, and other inflammatory mediators formation in increasing dose as compared to LPS-induced human endothelial cells. The expressions of NADPH oxidase subunits p22phox, p47phox, and gp91phox were increased in LPS-induced HUVECs but neferein was able to reverse the effect in a dose-dependent manner. The anti-inflammatory effects of neferine in LPS-induced endothelial cells are attributed through the modulation of MAPK and NF-κß pathways. Collectively, these results suggest that neferine could be beneficial in the early treatment of atherosclerosis to prevent stroke and heart disease.


Asunto(s)
Antiinflamatorios/farmacología , Bencilisoquinolinas/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Inflamación/tratamiento farmacológico , Animales , Antiinflamatorios/administración & dosificación , Aorta/citología , Aorta/patología , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/patología , Bencilisoquinolinas/administración & dosificación , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Inflamación/patología , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/toxicidad , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ratas , Factor de Necrosis Tumoral alfa/metabolismo
15.
Zhongguo Zhong Yao Za Zhi ; 43(12): 2531-2536, 2018 Jun.
Artículo en Zh | MEDLINE | ID: mdl-29950071

RESUMEN

In order to optimize the prescription and preparation process of norcantharidin/tetrandrine dual loaded liposomes, the dual drug loaded liposomes were prepared by film dispersion-ultrasonic method using norcantharidin-mesoporous silica nanoparticles(MSN-NCTD)and tetrandrine(Tet). With particle size and encapsulation efficiency as comprehensive indexes, the influences of phospholipid cholesterol amount, ultrasonic time and ultrasonic power on prescription process were investigated by orthogonal test; the in vitro release characteristics of liposomes were investigated by dialysis method. The results indicated that the best prescription process of prepared norcantharidin/tetrandrine dual loaded liposomes was as follows: phospholipid-cholesterol ratio 2.5:1, ultrasonic time 4 min, ultrasonic power 40%; the encapsulation efficiency was 86.62% and 79.19%respectively for NCTD and Tet;liposomes were well-shaped under the transmission microscope, with average particle size of (207.5±3.6) nm, Zeta potential of (1.345±0.173) mV; and the 48 h cumulative release rates of NCTD and Tet were 85.14% and 85.00% respectively. The experiment results proved that the dual drug loaded liposomes prepared by film dispersion-ultrasonic method had uniform particle size, high encapsulation efficiency and in vitro sustained release characteristics.


Asunto(s)
Bencilisoquinolinas/administración & dosificación , Compuestos Bicíclicos Heterocíclicos con Puentes/administración & dosificación , Portadores de Fármacos/química , Liposomas/química , Tamaño de la Partícula
16.
Biochem Biophys Res Commun ; 484(3): 694-701, 2017 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-28161641

RESUMEN

Cisplatin is one of the most effective chemotherapeutic agents; however, its clinical use is limited by serious side effects of which nephrotoxicity is the most important. Nephrotoxicity induced by cisplatin is closely associated with autophagy reduction and caspase activation. In this study, we investigated whether neferine, an autophagy inducer, had a protective effect against cisplatin-induced nephrotoxicity. In an in vitro cisplatin-induced nephrotoxicity model, we determined that neferine was able to induce autophagy and that pretreatment with neferine not only attenuated cisplatin-induced cell apoptosis but further activated cell autophagy. This pro-survival effect was abolished by the autophagic flux inhibitor chloroquine. Furthermore, neferine pretreatment activated the AMPK/mTOR pathway; however, pharmacological inhibition of AMPK abolished neferine-mediated autophagy and nephroprotection against cisplatin-induced apoptosis. Collectively, our findings suggest for the first time the possible protective mechanism of neferine, which is crucial for its further development as a potential therapeutic agent for cisplatin-induced nephrotoxicity.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia/efectos de los fármacos , Bencilisoquinolinas/administración & dosificación , Cisplatino/efectos adversos , Enfermedades Renales/inducido químicamente , Enfermedades Renales/metabolismo , Animales , Antineoplásicos/efectos adversos , Línea Celular , Citoprotección/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Enfermedades Renales/prevención & control , Ratas , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
17.
Pharm Biol ; 55(1): 1775-1779, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28521597

RESUMEN

CONTEXT: Cepharanthine (CPA) has been reported to possess a wide range of pharmacological activities. OBJECTIVE: This study investigates the pharmacokinetic characteristics after oral or intravenous administration of CPA by using a sensitive and rapid LC-MS/MS method. MATERIALS AND METHODS: A sensitive and rapid LC-MS/MS method was developed for the determination of CPA in Sprague-Dawley rat plasma. Twelve rats were equally randomized into two groups, including the intravenous group (1 mg/kg) and the oral group (10 mg/kg). Blood samples (250 µL) were collected at designated time points and determined using this method. The pharmacokinetic parameters were calculated. RESULTS: The calibration curve was linear within the range of 0.1-200 ng/mL (r = 0.999) with the lower limit of quantification at 0.1 ng/mL. After 1 mg/kg intravenous injection, the concentration of CPA reached a maximum of 153.17 ± 16.18 ng/mL and the t1/2 was 6.76 ± 1.21 h. After oral administration of 10 mg/kg of CPA, CPA was not readily absorbed and reached Cmax 46.89 ± 5.25 ng/mL at approximately 2.67 h. The t1/2 was 11.02 ± 1.32 h. The absolute bioavailability of CPA by oral route was 5.65 ± 0.35%, and the bioavailability was poor. DISCUSSION AND CONCLUSIONS: The results indicate that the bioavailability of CPA was poor in rats, and further research should be conducted to investigate the reason for its poor bioavailability and address this problem.


Asunto(s)
Bencilisoquinolinas/sangre , Bencilisoquinolinas/farmacocinética , Cromatografía Liquida , Espectrometría de Masas en Tándem , Administración Oral , Animales , Bencilisoquinolinas/administración & dosificación , Disponibilidad Biológica , Calibración , Cromatografía Liquida/normas , Semivida , Inyecciones Intravenosas , Modelos Lineales , Masculino , Modelos Biológicos , Ratas Sprague-Dawley , Estándares de Referencia , Reproducibilidad de los Resultados , Espectrometría de Masas en Tándem/normas
18.
J Microencapsul ; 33(3): 249-56, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26961245

RESUMEN

The objective of this study was to improve the efficacy of a natural compound tetrandrine against cancer by designing surfactant-free poly(lactic-co-glycolic acid) (PLGA) nanoparticles as drug carriers for tetrandrine. Nanoparticles were prepared from PLGA via the nano-precipitation method with or without the presence of surfactant poly(vinyl alcohol) (PVA) to encapsulate tetrandrine. Tetrandrine-loaded surfactant-free PLGA nanoparticles had an average particle size of 169.3 nm and morphology similar to the PLGA nanoparticles prepared using PVA as the surfactant. Tetrandrine-loaded surfactant-free PLGA nanoparticles could retard drug release in phosphate buffered saline (PBS) at pH 7.4 and the cumulative release of tetrandrine reached up to 68.33% over a period of 120 h. A549 cell line was used as the model cancer cells to investigate anticancer capability of tetrandrine-loaded surfactant-free PLGA nanoparticles via apoptosis assay, cytotoxicity and lysosome injury studies. The results showed that tetrandrine-loaded surfactant-free PLGA nanoparticles could effectively reduce cell viability and synergistically enhance tetrandrine-induced cell apoptosis.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacología , Bencilisoquinolinas/administración & dosificación , Bencilisoquinolinas/farmacología , Preparaciones de Acción Retardada/química , Poliglactina 910/química , Células A549 , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Liberación de Fármacos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Nanopartículas/química
19.
J BUON ; 21(1): 125-34, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27061540

RESUMEN

PURPOSE: Response surface methodology (RSM) using the central composite rotatable design (CCRD) model was used to optimize the formulation of paclitaxel (PTX)-cepharanthine (CEP) nanoparticles for gastric cancer. METHODS: Nanoparticles were prepared using nanoprecipitation technique and optimized using central composite rotatable design response surface methodology (CCRD-RSM). Further the optimized nanoparticles were characterised for particle size (PS), zeta potential, entrapment efficiency (EE), drug loading efficiency (DL), anticancer potential against MKN45 (human gastric cancer) cells, in vivo tumor inhibition and survival analysis. RESULTS: Significant findings were the optimal formulation of polymer concentration of 48 mg, surfactant concentration of 45% and EE of 98.12%, DL of 15.61% and mean diameter of 198±4.7 nm. The encapsulation of PTX/CEP into nanoparticles retained the synergistic anticancer efficiency against MKN45 cells. In the in vivo evaluation, PTXsCEP nanoparticles delivered into mice by intravenous injection significantly improved the antitumor efficacy of PTX/CEP. Moreover, PTX/CEP co-loaded nanoparticles substantially increased the overall survival in an established MKN45-transplanted mouse model. CONCLUSION: These data are the first to demonstrate that PTX/CEP co-loaded nanoparticles increased the anticancer efficacy in cell lines and xenograft mouse model. Our results suggest that PTX/CEP coloaded nanoparticles could be a potential useful chemotherapeutic formulation for gastric cancer.


Asunto(s)
Bencilisoquinolinas/administración & dosificación , Nanopartículas , Paclitaxel/administración & dosificación , Neoplasias Gástricas/tratamiento farmacológico , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones , Tamaño de la Partícula , Polímeros/administración & dosificación , Neoplasias Gástricas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Drug Metab Dispos ; 43(4): 534-52, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25609220

RESUMEN

The disposition and metabolism of hydrastine was investigated in 11 healthy subjects following an oral dose of 2.7 g of goldenseal supplement containing 78 mg of hydrastine. Serial blood samples were collected for 48 hours, and urine was collected for 24 hours. Hydrastine serum and urine concentrations were determined by Liquid Chromatography-tandem mass spectrometry (LC-MS/MS). Pharmacokinetic parameters for hydrastine were calculated using noncompartmental methods. The maximal serum concentration (Cmax) was 225 ± 100 ng/ml, Tmax was 1.5 ± 0.3 hours, and area under the curve was 6.4 ± 4.1 ng ⋅ h/ml ⋅ kg. The elimination half-life was 4.8 ± 1.4 hours. Metabolites of hydrastine were identified in serum and urine by using liquid chromatography coupled to high-resolution mass spectrometry. Hydrastine metabolites were identified by various mass spectrometric techniques, such as accurate mass measurement, neutral loss scanning, and product ion scanning using Quadrupole-Time of Flight (Q-ToF) and triple quadrupole instruments. The identity of phase II metabolites was further confirmed by hydrolysis of glucuronide and sulfate conjugates using bovine ß-glucuronidase and a Helix pomatia sulfatase/glucuronidase enzyme preparation. Hydrastine was found to undergo rapid and extensive phase I and phase II metabolism. Reduction, O-demethylation, N-demethylation, hydroxylation, aromatization, lactone hydrolysis, and dehydrogenation of the alcohol group formed by lactone hydrolysis to the ketone group were observed during phase I biotransformation of hydrastine. Phase II metabolites were primarily glucuronide and sulfate conjugates. Hydrastine undergoes extensive biotransformation, and some metabolites may have pharmacological activity. Further study is needed in this area.


Asunto(s)
Bencilisoquinolinas/sangre , Bencilisoquinolinas/orina , Suplementos Dietéticos , Hydrastis/química , Administración Oral , Bencilisoquinolinas/administración & dosificación , Bencilisoquinolinas/metabolismo , Cromatografía Liquida , Estabilidad de Medicamentos , Femenino , Voluntarios Sanos , Humanos , Masculino , Fase I de la Desintoxicación Metabólica , Fase II de la Desintoxicación Metabólica , Proyectos Piloto , Espectrometría de Masas en Tándem , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA