Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Pathol Res Pract ; 216(8): 153031, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32703495

RESUMO

Oridonin has been found to be a potential anti-angiogenesis agent. However, its functional targets and the underlying mechanisms are still vague. In vitro studies we found that oridonin not only inhibited VEGF-induced cell proliferation, migration and tube formation but also caused G2/M phase arrest and triggered cellular apoptosis in HUVECs. In mechanistic studies revealed that oridonin exhibited the anti-angiogenic potency, at least in part, through the down-regulation of VEGFR2-mediated FAK/MMPs, mTOR/PI3K/Akt and ERK/p38 signaling pathways which led to reduced invasion, migration, and tube formation in HUVECs. Our results could provide evidence that oridonin exerts strong anti-angiogenesis activities via specifically targeting VEGFR2 and its signaling pathway.


Assuntos
Inibidores da Angiogênese/farmacologia , Diterpenos do Tipo Caurano/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
2.
J Cell Biochem ; 120(3): 3736-3746, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30229997

RESUMO

Oridonin, an active diterpenoid isolated from Rabdosia rubescens, has been reported for its antitumor activity on several cancers. However, its effect on human esophageal cancer remains unclear. In this study, we demonstrated that oridonin could inhibit the growth of human esophageal cancer cells both in vitro and in vivo. Oridonin not only suppressed the proliferation, but also induced cell cycle arrest and mitochondrial-mediated apoptosis in KYSE-30, KYSE-150, and EC9706 cells with dose-dependent manner. Further mechanism studies revealed that oridonin led cell cycle arrest in esophageal cancer cells via downregulating cell cycle-related proteins, such as cyclin B1 and CDK2, while upregulating p53 and p21. Oridonin also increased proapoptotic protein Bax and reduced antiapoptotic protein Bcl-2, as well as the increased expression of cleaved caspase-3, -8, and -9. In addition, oridonin treatment could significantly inhibit the PI3K/Akt/mTOR and Ras/Raf signaling pathway. In vivo results further demonstrated that oridonin treatment markedly inhibited tumor growth in the esophageal cancer xenograft mice model. Taken together, these results suggest that oridonin may be a potential anticancer agent for the treatment of esophageal cancer.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Diterpenos do Tipo Caurano/farmacologia , Neoplasias Esofágicas/tratamento farmacológico , Regulação Neoplásica da Expressão Gênica , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Serina-Treonina Quinases TOR/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ciclina B1/antagonistas & inibidores , Ciclina B1/genética , Ciclina B1/metabolismo , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Quinase 2 Dependente de Ciclina/genética , Quinase 2 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/agonistas , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Feminino , Humanos , Camundongos , Camundongos Nus , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Carga Tumoral/efeitos dos fármacos , Proteína Supressora de Tumor p53/agonistas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína X Associada a bcl-2/agonistas , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
3.
Artif Cells Nanomed Biotechnol ; 46(sup3): S297-S307, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30183382

RESUMO

Graphene oxides (GO) is a promising building material to fabricate desired drug delivery system due to its excellent physicochemical properties. In this study, an innovative nano-drug (Ori@GE11-GO) was constructed based on GE11 peptide functionalized GO for targeted delivery of oridonin to realize the specific recognition of tumour cells and enhance anticancer efficiency. GE11 surface modification onto GO significantly increased the cellular uptake of GO in EGFR overexpressed oesophageal cancer cells (KYSE-30 and EC109 cells) than that of normal cells, indicating the EGFR targeting effects of Ori@GE11-GO. The internalized Ori@GE11-GO could accumulate into lysosomes and significantly inhibit the viability of cancer cells. Moreover, Ori@GE11-GO could effectively induce KYSE-30 and EC109 cells cycle arrest, apoptosis, mitochondrial membrane potential (△Ψm) disruption through the activation of apoptotic signalling pathways and the inhibition of EGFR/Ras/Raf/MEK/ERK signalling pathway, showing potential use of Ori@GE11-GO for cancer treatment. Taken together, this study demonstrates a good strategy for the construction of bio-functionalized GO drug delivery nanosystem to improve the cancer targeting efficiency of anticancer medicines.


Assuntos
Antineoplásicos , Apoptose/efeitos dos fármacos , Portadores de Fármacos , Neoplasias Esofágicas , Grafite , Nanopartículas , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/farmacologia , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Grafite/química , Grafite/farmacocinética , Grafite/farmacologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Nanopartículas/química , Nanopartículas/uso terapêutico
4.
Pathol Res Pract ; 214(5): 691-699, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29567333

RESUMO

Matrine, as a natural alkaloid isolated from the traditional herb medicine sophora flavescens, has been proved to possess excellent biological activities, including anticancer effects. Now, this research aims to assess the anticancer activities and the mechanism of matrine against esophageal cancer cells, we investigated the proliferative inhibition, apoptosis induction, as well as the underlying mechanism of matrine on esophageal cancer KYSE-150 cells. It was found that matrine could suppress KYSE-150 cell proliferation and significantly mediate cell apoptosis in a dose-dependent relation by increasing intracellular reactive oxygen species level and triggering mitochondrial membrane potential disruption. More precise mechanism studies demonstrated that matrine could up-regulate the expression of Bax proteins and down-regulate the expression of Bcl-2 proteins, as well as the activation about caspase-3, 8 and 9 in KYSE-150 cells. The morphological analysis of KYSE-150 cells exhibited that matrine could destroy the F-actin and nuclei structures and induce morphological damage with increased surface height distribution and roughness of cell membrane. These results not only demonstrated the potential anticancer activity mechanism of matrine at nanoscale, but also provide preliminary guidance for the treatment of esophageal cancer using matrine.


Assuntos
Proliferação de Células/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Mitocôndrias/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Alcaloides , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Quinolizinas , Proteína X Associada a bcl-2/metabolismo , Matrinas
5.
Bioorg Med Chem Lett ; 26(11): 2730-4, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27080177

RESUMO

According to the previous studies, the anticancer activity of flavonoids could be enhanced when they are coordinated with transition metal ions. In this work, kaempferol-zinc(II) complex (kaempferol-Zn) was synthesized and its chemical properties were characterized by UV-VIS, FT-IR, (1)H NMR, elemental analysis, electrospray mass spectrometry (ES-MS) and fluorescence spectroscopy, which showed that the synthesized complex was coordinated with a Zn(II) ion via the 3-OH and 4-oxo groups. The anticancer effects of kaempferol-Zn and free kaempferol on human oesophageal cancer cell line (EC9706) were compared. MTT results demonstrated that the killing effect of kaempferol-Zn was two times higher than that of free kaempferol. Atomic force microscopy (AFM) showed the morphological and ultrastructural changes of cellular membrane induced by kaempferol-Zn at subcellular or nanometer level. Moreover, flow cytometric analysis indicated that kaempferol-Zn could induce apoptosis in EC9706 cells by regulating intracellular calcium ions. Collectively, all the data showed that kaempferol-Zn might be served as a kind of potential anticancer agent.


Assuntos
Antineoplásicos/farmacologia , Quempferóis/farmacologia , Compostos Organometálicos/farmacologia , Zinco/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Quempferóis/química , Estrutura Molecular , Compostos Organometálicos/síntese química , Compostos Organometálicos/química , Espectrometria de Fluorescência , Relação Estrutura-Atividade , Zinco/química
6.
Scanning ; 38(6): 644-653, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26890985

RESUMO

Kaempferol has been identified as a potential cancer therapeutic agent by an increasing amount of evidences. However, the changes in the topography of cell membrane induced by kaempferol at subcellular- or nanometer-level were still unclear. In this work, the topographical changes of cytomembrane in human cervical cancer cell (SiHa) induced by kaempferol, as well as the role of kaempferol in apoptosis induction and its possible mechanisms, were investigated. At the macro level, MTT assays showed that kaempferol inhibited the proliferation of SiHa cells in a time- and dose-dependent manner. Flow cytometry analysis demonstrated that kaempferol could induce SiHa cell apoptosis, mitochondrial membrane potential disruption, and intracellular free calcium elevation. At the micro level, fluorescence imaging by laser scanning confocal microscopy (LSCM) indicated that kaempferol could also destroy the networks of microtubules. Using high resolution atomic force microscopy (AFM), we determined the precise changes of cellular membrane induced by kaempferol at subcellular or nanometer level. The spindle-shaped SiHa cells shrank after kaempferol treatment, with significantly increased cell surface roughness. These data showed structural characterizations of cellular topography in kaempferol-induced SiHa cell apoptosis and might provide novel integrated information from macro to nano level to assess the impact of kaempferol on cancer cells, which might be important for the understanding of the anti-cancer mechanisms of drugs. SCANNING 38:644-653, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Apoptose/efeitos dos fármacos , Quempferóis/farmacologia , Neoplasias do Colo do Útero/tratamento farmacológico , Cálcio/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Feminino , Humanos , Microscopia de Força Atômica , Neoplasias do Colo do Útero/patologia , Neoplasias do Colo do Útero/ultraestrutura
7.
Oncotarget ; 6(36): 38952-66, 2015 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-26517515

RESUMO

Previously we reported that valproic acid (VPA) acts in synergy with GOS to enhance cell death in human DU145 cells. However, the underlying mechanism remains elusive. In this study, we observed that such synergistic cytotoxicity of GOS and VPA could be extended to human A375, HeLa, and PC-3 cancer cells. GOS and VPA co-treatment induced robust apoptosis as evidenced by caspase-8/-9/-3 activation, PARP cleavage, and nuclear fragmentation. GOS and VPA also markedly decreased cyclin A2 protein expression. Owing to the reduction of cyclin A2, Akt signaling was suppressed, leading to dephosphorylation of FOXO3a. Consequently, FOXO3a was activated and the expression of its target genes, including pro-apoptotic FasL and Bim, was upregulated. Supporting this, FOXO3a knockdown attenuated FasL and Bim upregulation and apoptosis induction in GOS+VPA-treated cells. Furthermore, blocking proteasome activity by MG132 prevented the downregulation of cyclin A2, dephosphorylation of Akt and FOXO3a, and induction of apoptosis in cells co-treated with GOS and VPA. In mouse model, GOS and VPA combination significantly inhibited the growth of A375 melanoma xenografts. Our findings indicate that GOS and VPA co-treatment induces apoptosis in human cancer cells by suppressing the cyclin-A2/Akt/FOXO3a pathway.


Assuntos
Ciclina A2/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Gossipol/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ácido Valproico/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Proteína Forkhead Box O3 , Células HeLa , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Fragmentos de Peptídeos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais
8.
Chem Asian J ; 9(8): 2272-80, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24958675

RESUMO

A simple method for preparing quercetin surface-functionalized germanium nanoparticles (Qu-GeNPs) with enhanced antioxidant and anticancer activity is reported. Spherical germanium nanoparticles (GeNPs) were capped by quercetin (Qu) with a mean particle size of approximately 33 nm and were characterized by TEM, AFM, UV-visible absorption spectroscopy, FTIR, and XRD measurements. The in vitro drug release of Qu from the Qu-GeNPs indicated that Qu could principally be distributed around tumor tissues rather than in the normal section and Qu-GeNPs were internalized by MCF-7 cells. Their biological activity test results indicated that these Qu-GeNPs possessed stronger hydroxyl-scavenging effects and proliferative inhibition effect on MCF-7 cancer cells than quercetin, thus suggesting that the strategy to use GeNPs as a carrier of Qu could be an efficient way to achieve enhanced antioxidant and anticancer activity. In addition, Qu-GeNPs possessed a high apoptotic induction effect in cancer cells, especially in high dosages, and could arrest MCF-7 cells in the S phase.


Assuntos
Antineoplásicos/administração & dosagem , Germânio/química , Nanopartículas Metálicas , Quercetina/administração & dosagem , Antineoplásicos/farmacologia , Antioxidantes/administração & dosagem , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Humanos , Concentração de Íons de Hidrogênio , Células MCF-7 , Microscopia de Força Atômica , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Quercetina/farmacologia , Espectrofotometria Ultravioleta
9.
Biosens Bioelectron ; 56: 271-7, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24514079

RESUMO

A new method based on Atomic Force Microscopy (AFM) was developed to real-time and in-situ detect epidermal growth factor receptor (EGFR) expression levels on living MCF-7 cells for evaluating the anticancer activity of resveratrol. Here, the inhibition effect of resveratrol on EGFR expression levels on MCF-7 cells was probed by epidermal growth factor (EGF)-functionalized tips for the first time. Changes in morphology and stiffness of single cell stimulated by resveratrol at different concentrations were detected by AFM. The consequences showed that resveratrol influenced the cellular state and reduced expression of EGFR on the cell surface, which were also interpreted by MTT assay and confocal microscopy assay. AFM, which was used to investigate potential targets for anti-tumor drug on living cells and realize a better understanding of drug action mechanism, was expected to be developed into a promising tool for screening of drugs.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos Fitogênicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/análise , Estilbenos/farmacologia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Sobrevivência Celular/efeitos dos fármacos , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Feminino , Humanos , Proteínas Imobilizadas/metabolismo , Células MCF-7 , Microscopia de Força Atômica/métodos , Imagem Óptica , Resveratrol
10.
Artigo em Inglês | MEDLINE | ID: mdl-24496671

RESUMO

Oval cells have a potential to differentiate into a variety of cell lineages including hepatocytes and biliary epithelia. Several models have been established to activate the oval cells by incorporating a variety of toxins and carcinogens, alone or combined with surgical treatment. Those models are obviously not suitable for the study on human hepatic oval cells. It is necessary to establish a new and efficient model to study the human hepatic oval cells. In this study, the hepatocyte growth factor (HGF) and epidermal growth factor (EGF) were used to induce differentiation of mouse embryonic stem (ES) cells into hepatic oval cells. We first confirmed that hepatic oval cells derived from ES cells, which are bipotential, do exist during the course of mouse ES cells' differentiation into hepatic parenchymal cells. RT-PCR and transmission electron microscopy were applied in this study. The ratio of Sca-1+/CD34+ cells sorted by FACS in the induction group was increased from day 4 and reached the maximum on the day 8, whereas that in the control group remained at a low level. The differentiation ratio of Sca-1+/CD34+ cells in the induction group was significantly higher than that in the control group. About 92.48% of the sorted Sca-1+/CD34+ cells on the day 8 were A6 positive. Highly purified A6+/Sca-1+/CD34+ hepatic oval cells derived from ES cells could be obtained by FACS. The differentiation ratio of hepatic oval cells in the induction group (up to 4.46%) was significantly higher than that in the control group. The number of hepatic oval cells could be increased significantly by HGF and EGF. The study also examined the ultrastructures of ES-derived hepatic oval cells' membrane surface by atomic force microscopy. The ES-derived hepatic oval cells cultured and sorted by our protocols may be available for the future clinical application.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Fígado/citologia , Células-Tronco/citologia , Animais , Antígenos CD34/genética , Antígenos CD34/metabolismo , Antígenos Ly/genética , Antígenos Ly/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/ultraestrutura , Fator de Crescimento Epidérmico/farmacologia , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Fígado/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Proteínas dos Microfilamentos/metabolismo , Microscopia de Força Atômica , Microscopia Eletrônica de Transmissão , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/metabolismo , Células-Tronco/ultraestrutura , Fatores de Tempo
11.
Bioorg Med Chem Lett ; 23(20): 5544-51, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24007917

RESUMO

Chrysin (5,7-dihydroxylflavone, Chry) is a natural product extracted from plants, honey, and propolis. In this work, a novel chrysin-organogermanium(IV) complex (Chry-Ge) with enhanced anticancer activities was synthesized, and its potential anticancer effects against cancer cells were measured using various methods. MTT results showed that Chry-Ge had significant inhibition effects on the proliferation of MCF-7, HepG2 and Colo205 human cancer cell lines in a dose-dependent manner while had little cytotoxic effects on MCF-10A human normal cells (MCF-10A cells) with the same treatment of Chry-Ge. These results suggested that Chry-Ge possessed enhanced anticancer effects and high selectivity between cancer cells and normal cells. The immuno-staining results showed that the nuclei of MCF-7 cells represented a total fragmented morphology and a disorganized cytoskeletal network in MCF-7 cells after Chry-Ge treatment. Besides, atomic force microscopy (AFM) was applied to detect the changes of ultrastructural and biomechanical properties of MCF-7 cellular membrane induced by Chry-Ge. The AFM data indicated that Chry-Ge treatment directly caused the decrease of cell rigidity and adhesion force of MCF-7 cells, suggesting that membrane toxicity might be one of the targets for Chry-Ge in MCF-7 cells. Moreover, the fluorescence-based flow cytometric analysis demonstrated that Chry-Ge could induce apoptosis in MCF-7 cells in ROS-dependent mitochondrial pathway. All results collectively showed that Chry-Ge could be as a promising anticancer drug for cancer therapy.


Assuntos
Antineoplásicos/química , Complexos de Coordenação/química , Flavonoides/química , Germânio/química , Antineoplásicos/síntese química , Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Complexos de Coordenação/síntese química , Complexos de Coordenação/toxicidade , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Células Hep G2 , Humanos , Células MCF-7 , Metaloproteinases da Matriz/metabolismo , Microscopia de Força Atômica , Espécies Reativas de Oxigênio/metabolismo
12.
Biosens Bioelectron ; 49: 46-52, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23708817

RESUMO

A novel electrochemiluminescence (ECL) cytosensors was developed for the detection of early apoptotic cells by the specific interaction between Annexin V and phosphatidylserine(PS) based on ECL signal of CdS-QDs. Immobilization of Annexin V on a L-cysteine-capped CdS-QDs/Polyaniline nanofibers (PANI-NF) resulted in the stable and high loading of Annexin V on the sensor surface and the possibility of sensitivity enhancement. Early apoptotic cells showed an increased exposure of PS on the cell membrane caused by physiological and pathological response reactions, leading to a strong interaction between the apoptotic cells and the sensor surface, which could be probed by the ECL. Using a real of early apoptotic HepG2 cell induced by resveratrol (RVL), the proposed novel strategy has demonstrated its simplicity, high sensitivity, good selectivity and high reproducibility and label-free capability which might hold a great potential for rapid detection of cell apoptosis and drug screening. The results from this approach have showed good agreement with those obtained using inverted microscope, flow cytometry(FCM) and Atomic force microscopy(AFM). The linear range for early apoptotic cells detection ranged from 500 to 1.0 × 10(6) cells mL(-1) with a detection limit of 500 cells mL(-1). The reported strategy has provided a promising platform for highly sensitive cytosensing and convenient screening of some clinically anticancer drugs.


Assuntos
Compostos de Anilina/química , Anexina A5/metabolismo , Apoptose , Técnicas Biossensoriais/métodos , Nanofibras/química , Fosfatidilserinas/metabolismo , Pontos Quânticos , Anexina A5/química , Antineoplásicos Fitogênicos/farmacologia , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Compostos de Cádmio/química , Células Hep G2 , Humanos , Proteínas Imobilizadas/química , Proteínas Imobilizadas/metabolismo , Limite de Detecção , Medições Luminescentes/métodos , Resveratrol , Estilbenos/farmacologia , Sulfetos/química
13.
Bioorg Med Chem Lett ; 23(10): 2902-8, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23570787

RESUMO

Germanium (Ge) is considered to play a key role in the pharmacological effects of some medicinal plants. Here, two new Ge(IV)-polyphenol complexes were synthesized and measured for their potential biological activities. The results indicated that these Ge(IV)-polyphenol complexes possessed great anti-oxidative activities, both showing stronger hydroxyl scavenging effects than their corresponding ligands. We also demonstrated the strong intercalating abilities of Ge(IV)-polyphenol complexes into calf thymus-DNA molecules. In addition, these two Ge(IV)-polyphenol complexes showed strong proliferative inhibition effect on HepG2 cancer cells. Moreover, the morphological changes in HepG2 cells induced by Ge(IV)-polyphenol complexes were detected by atomic force microscopy. All these results collectively suggested that Ge(IV)-polyphenol complexes could be served as promising pharmacologically active substances against cancer treatment.


Assuntos
Antineoplásicos/farmacologia , Germânio/química , Compostos Organometálicos/farmacologia , Polifenóis/química , Antineoplásicos/síntese química , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Células Hep G2 , Humanos , Estrutura Molecular , Compostos Organometálicos/síntese química , Compostos Organometálicos/química , Relação Estrutura-Atividade
14.
Autophagy ; 9(1): 20-32, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23075929

RESUMO

Autophagic responses to chemotherapeutic agents may vary greatly among different prostate cancer cells and have not been well characterized. In this study, we showed that valproic acid (VPA) induced conversion of LC3-I to LC3-II and formation of LC3 puncta, the typical markers of autophagy, in LNCaP and PC-3 cells. However, these markers were undetectable in DU145 cells upon autophagic stimulation, indicating a defect of autophagy in this cell line. Among several critical autophagy-related proteins, ATG5 and ATG12-ATG5 conjugates, which are essential for autophagy induction, were absent in DU145 cells. No canonical transcripts for full-length ATG5 but only two alternatively spliced ATG5 transcripts were identified in DU145 cells. These alternative transcripts lack one or two exons, leading to premature termination of ATG5 translation. Transfection of the wild-type ATG5 gene into DU145 cells rescued the production of ATG5 and ATG12-ATG5 conjugates, resulting in formation of LC3-II conjugates and LC3 puncta. Moreover, the levels of the SQSTM1 protein, which should be degradable as an autophagy adaptor, were much higher in DU145 than in LNCaP and PC-3 cells, but were significantly decreased after ATG5 restoration in DU145 cells. However, expression of wild-type ATG5 in DU145 or knockdown of ATG5 in LNCaP and PC-3 cells did not change the inhibitory effects of VPA on these cells. Collectively, these results indicated that VPA-induced autophagy in prostate cancer cells depended on ATG5 and more importantly, that the autophagy pathway was genetically impaired in DU145 cells, suggesting caution in interpreting autophagic responses in this cell line.


Assuntos
Processamento Alternativo , Autofagia/genética , Autofagia/fisiologia , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Processamento Alternativo/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Proteína 12 Relacionada à Autofagia , Proteína 5 Relacionada à Autofagia , Sequência de Bases , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Masculino , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Proteína Sequestossoma-1 , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Transfecção , Ácido Valproico/farmacologia
15.
Cancer Chemother Pharmacol ; 70(3): 415-24, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22814677

RESUMO

PURPOSE: The cucurbitacins are a class of triterpenoid molecules that possess cytotoxic characteristics for plant defense against herbivore feeding. 23,24-dihydrocucurbitacin F (DHCF), a derivative of the cucurbitacin family, has been isolated as an active component from the root of Hemsleya amabilis (Cucurbitaceae), an ancient Chinese remedy for bacillary dysentery, gastroenteritis, and cancers. While the toxicity of other cucurbitacins has been explored in several cancers, little data exist on the effect of DHCF on human cancers, including prostate cancer (PCa). In this study, we explore the level and mechanisms of DHCF toxicity on human PCa cell lines. METHODS: Human PCa DU145, PC3, and LNCaP cells were treated with graded doses of DHCF in vitro, and anti-proliferative, cytotoxic, and proteomic effects were determined using MTS assay, cell cycle analysis, immunofluorescent staining, and western blotting. RESULTS: DHCF inhibited cell growth and induced cell cycle arrest at G(2)/M phase, formation of binucleated cells, and increased levels of apoptosis in all PCa cell lines tested. G-actin depletion, actin aggregation, and rod-like actin fibers, with little effect on microtubule structure, were observed after DHCF treatment. Actin aggregation and cofilin-actin rod formation were highly correlated with rapid and persistent dephosphorylation of cofilin-1 (cofilin). DHCF treatment resulted in upregulation of p21(Cip1) and downregulation of cyclin A in all three PCa cell lines. CONCLUSIONS: The anti-proliferative activity of DHCF on human PCa cells may be brought about by inducing actin aggregation and cofilin-actin rod formation, leading to cell cycle arrest, cytokinesis failure, and apoptosis.


Assuntos
Actinas/metabolismo , Proliferação de Células/efeitos dos fármacos , Cucurbitacinas/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Fatores de Despolimerização de Actina/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Cucurbitaceae/química , Cucurbitacinas/isolamento & purificação , Ciclina A/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Citocinese/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Humanos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Masculino , Neoplasias da Próstata/patologia , Regulação para Cima/efeitos dos fármacos
16.
J Proteomics ; 74(10): 2180-93, 2011 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-21726675

RESUMO

Gossypol (GOS), a BH3 mimetic, has been investigated as a sensitizing co-therapy to radiation and chemotherapy in treatment of metastatic prostate cancer. In this study, we found that valproic acid (VPA), a histone deacetylase inhibitor (HDACI), counteracted the suppressive effect of GOS on histone H3 acetylation and enhanced the cytotoxicity of GOS to DU145 prostate cancer cells. Significant synergistic effects were observed in combined GOS and VPA treatment, culminating in more DNA damage and cell death. The iTRAQ-based quantitative proteomic analysis revealed differential proteomic profiles in cells treated with VPA, GOS or their combination. In GOS-treated cells, oxidative phosphorylation-related proteins were depressed and endoplasmic reticulum stress markers were upregulated. In the presence of VPA, the GOS-induced mitochondrial stress was further enhanced since glycolysis- and hypoxia-associated proteins were upregulated, suggesting a disruption of energy metabolism in these cells. Furthermore, the DNA damage repair ability of cells co-treated with GOS and VPA was also decreased, as evidenced by the downregulation of DNA damage repair proteins and the enhancement of DNA fragmentation and cell death. These findings suggest that GOS in combination with an HDACI has the potential to increase its clinical efficacy in the treatment of prostate cancer.


Assuntos
Antineoplásicos/uso terapêutico , Gossipol/uso terapêutico , Inibidores de Histona Desacetilases/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Ácido Valproico/uso terapêutico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Dano ao DNA/efeitos dos fármacos , Sinergismo Farmacológico , Metabolismo Energético/efeitos dos fármacos , Humanos , Masculino , Mitocôndrias/efeitos dos fármacos , Proteômica
17.
Guang Pu Xue Yu Guang Pu Fen Xi ; 29(5): 1398-401, 2009 May.
Artigo em Chinês | MEDLINE | ID: mdl-19650499

RESUMO

A novel strategy based on antigen-antibody interaction was developed by means of backfilling transferrin on antibody functionalized gold nanoparticles surfaces in the present study. Nano gold particles were immobilized with cysteamine layer by self-assembly, whose surfaces were chemically coupled with anti-transferrin antibodies by using EDCX, forming the anti-transferrin-Au immuno-probes. The particles and the nano-probes were characterized by the integrated tools of resonance Rayleigh scattering (RRS), UV-Vis absorption spectra, transmission electron microscopy (TEM) and laser light scattering. The result showed that the nano-probe with the ability of specific recognition of transferrin had good immune activity. The RRS peak at 470 nm was amplified obviously by using this signal amplification, and the antigen recognition was monitored via the enhancement of 470 nm RRS intensity when this binding event occurred. The result showed that the amplification strategy led to a dramatic improvement of the detection sensitivity of transferrin, and the detection of transferrin featured a linear range of 0.85 to 33.9) x 10(-10) mol x L(-1) with the detection limit of 8.5 x 10(-11) mol x L(-1).

18.
Zhonghua Yi Xue Za Zhi ; 89(12): 800-4, 2009 Mar 31.
Artigo em Chinês | MEDLINE | ID: mdl-19595116

RESUMO

OBJECTIVE: To investigate the effects of nanogold in inhibition of angiogenesis and growth of liver cancer cells. METHODS: Nanogold was co-incubated with VEGF165 and VDGF121 respectively. Atomic force microscopy (AFM) was used to observe the changes of the form of the particles. Human umbilical vascular endothelial cells (HUVEC) were serum-starved for 24 hours, then co-cultured with VEGF165 + nanogold or VEGF121 + nanogold for 24 h. ATM was used to observe the ultrastructure of the cells. Another HUVEC were serum-starved for 24 hours and then cultured with VEGF165 (10 microg/L) 100 microl + nanogold 125, 250, and 500 nmol/L 100 microl respectively for 5 min. Then Western blotting was used to detect the phosphorylation protein of phospholipase C (PLC)-gamma1 on VEGFR-2. Hepatocellular cancer cells of the line H22 were injected subcutaneously into the right armpit of 20 Balb/c nude mice. When the size of transplanted tumor reached about 8 mm, the mice were divided into 2 equal groups: experimental group undergoing injection of nanogold into the tumor once a day for 8 days, and control group injected with normal saline. On day 14 the mice were sacrificed with the liver tumors taken out to measure the size and weight. The microvascular density (MVD) of tumor was determined by immunohistochemical staining. RESULTS: ATM showed that acted with VEGF165, the size of nanogold became over 30 nm. Treated with VEGF165 the HUVEC became larger with obvious pseudopodium. However, such changes were obviously milder in those HUVEC treated with nanogold + VEGF165. The PLC-gamma1 phosphorylation level VEGF receptor-2 was decreased along with the increase of the concentration of nanogold. The MVD of liver cancer tissue in the experimental group was 14.27 +/- 1.08, significantly lower than that of the control group [(23.52 +/- 1.36), P < 0.01]. The mean weight and volume of tumor of the experimental group were (1.39 +/- 0.08) g and (1.37 +/- 0.34) cm(3) respectively, both significantly lower than those of the control group [(2.47 +/- 0.15) g and (2.49 +/- 0.78) cm(3) respectively, both P < 0.05] with a tumor growth inhibition rate of 43.72%. CONCLUSION: Nanogold significantly inhibits the angiogenesis and growth of liver cancer cells with the possible mechanism that nanogold inhibits the VEGF165-induced signaling.


Assuntos
Ouro/farmacologia , Neoplasias Hepáticas Experimentais/sangue , Neoplasias Hepáticas Experimentais/patologia , Nanopartículas , Neovascularização Patológica/prevenção & controle , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Ouro/administração & dosagem , Ouro/uso terapêutico , Humanos , Neoplasias Hepáticas Experimentais/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
19.
Micron ; 39(8): 1216-21, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18514532

RESUMO

Taxol (paclitaxel), one of the most active cancer chemotherapeutic agents, can cause programmed cell death (PCD) and cytoplasmic vacuolization. The objective of this study was to analyze the morphological characteristics induced by taxol. Human lung adenocarcinoma (ASTC-a-1) cells were exposed to various concentration of taxol. CCK-8 was used to assay the cell viability. Atomic force microscopy (AFM), plasmid transfection and confocal fluorescence microscopy were performed to image the cells morphological change induced by taxol. Fluorescence resonance energy transfer (FRET) was used to monitor the caspase-3 activation in living cells during taxol-induced cell death. Cells treated with taxol exhibited significant swelling and cytoplasmic vacuolization which may be due to endoplasmic reticulum (ER) vacuolization. Caspase-3 was not activated during taxol-induced cytoplasmic vacuolization and cell death. These findings suggest that taxol induces caspase-3-independent cytoplasmic vacuolization, cell swelling and cell death through ER vacuolization.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos Fitogênicos/farmacologia , Citoplasma/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Paclitaxel/farmacologia , Vacúolos/efeitos dos fármacos , Adenocarcinoma/ultraestrutura , Caspase 3/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citoplasma/ultraestrutura , Retículo Endoplasmático/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/ultraestrutura , Vacúolos/ultraestrutura
20.
Appl Biochem Biotechnol ; 141(2-3): 203-13, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18025552

RESUMO

A novel strategy for constructing multiple joined genes of acidic partner-mediated antimicrobial peptide is described. This strategy allows the expression of antimicrobial peptide by Escherichia coli in a stable form and with high yield. Cecropin A (1-8)-melittin (1-10) (CAME) hybrid peptide was selected as a model of antimicrobial peptide. An acidic fragment from magainin intervening sequence was fused to the antimicrobial peptide as a partner to neutralize the lethal effects on the host cells. Multiple copies of the fusion peptide gene were tandemly linked and cloned into the expression vector pET21a. Multimers were expressed at high levels, reaching up to 36% of total cell proteins, and expression levels were proportional to the degree of multimerization. The fusion proteins were mainly expressed as inclusion bodies, probably owing to cysteine residues in the multimers. The target CAME peptide was obtained by cleaving the multimers with cyanogen bromide and purified by cation-exchange chromatography. Recombinant CAME peptide showed strong antimicrobial activities against both Gram-negative and -positive bacteria. These results might provide an efficient solution for high-level expression of various kinds of antimicrobial peptides that are toxic to the host.


Assuntos
Escherichia coli/genética , Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Peptídeos/química , Peptídeos Catiônicos Antimicrobianos/química , Cátions , Cromatografia por Troca Iônica/métodos , Clonagem Molecular , Cisteína/química , Expressão Gênica , Vetores Genéticos , Concentração de Íons de Hidrogênio , Plasmídeos/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA