Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Eur J Pharm Sci ; 198: 106789, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38710335

RESUMO

BACKGROUND: Cytoplasmic epidermal growth factor receptor (EGFR) is overexpressed in both nasopharyngeal carcinoma (NPC) and triple-negative breast cancer (TNBC), while clinical outcome and prognosis vary greatly among patients treated with gefitinib, and all patients eventually develop resistance to this agent. Therefore, we propose a new concept for synthesizing multitarget compounds and reveal new therapeutic strategies for NPC and TNBC expressing EGFR. METHODS: Compound H was synthesized in our previous study. Molecular docking, and cell thermal shift assays (CETSAs) and drug affinity responsive target stability(DARTS) were used to confirm the binding of compound H to EGFR and GLUT1. Methylthiazolyldiphenyl-tetrazolium bromide(MTT), annexin V-PE assays, mitochondrial membrane potential (MMP) assays, and animal models were used to evaluate the inhibitory effect of compound H on TNBC cell lines. Energy metabolism tests, Western blotting, and immunofluorescence staining were performed to evaluate the synergistic effects on EGFR- and glucose transporter type 1(GLUT1)-mediated energy metabolism. RESULTS: Compound H can simultaneously act on the EGFR tyrosine kinase ATP-binding site and inhibit GLUT1-mediated energy metabolism, resulting in reductions in ATP, MMP, intra-cellular lactic acid, and EGFR nuclear transfer. The anti-tumor activity of compound H is significantly superior to the combination of GLUT1 inhibitor BAY876 and EGFR inhibitor gefitinib. Compound H has remarkable anti-proliferative effects on TNBC MDA-MB231 cells, and importantly, no obvious toxicity effects of compound H were found in vivo. CONCLUSIONS: Synergistic effects of inhibition of EGFR- and GLUT1-mediated energy metabolism by compound H may present a new strategy for the treatment of TNBC and NPC.


Assuntos
Antineoplásicos , Receptores ErbB , Transportador de Glucose Tipo 1 , Carcinoma Nasofaríngeo , Neoplasias de Mama Triplo Negativas , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Transportador de Glucose Tipo 1/antagonistas & inibidores , Transportador de Glucose Tipo 1/metabolismo , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo , Humanos , Animais , Linhagem Celular Tumoral , Feminino , Carcinoma Nasofaríngeo/tratamento farmacológico , Carcinoma Nasofaríngeo/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Simulação de Acoplamento Molecular , Neoplasias Nasofaríngeas/tratamento farmacológico , Neoplasias Nasofaríngeas/metabolismo , Camundongos Nus , Camundongos Endogâmicos BALB C , Gefitinibe/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos
2.
Future Med Chem ; 15(2): 129-145, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36799271

RESUMO

Background: Endoplasmic reticulum (ER) stress is a therapeutic target in cancer given its regulation of bioenergetics and cell death. Methodology & results: We synthesized 14 ER stress-triggered anthraquinone derivatives by introducing an amino group at the 3-position side chain of the lead compound obtained previously. Most of the anthraquinone derivatives exhibited good antitumor activity due to their ability to induce ER damage through cytoplasmic vacuoles. The mechanisms of ER stress caused by compound KA-4c were related to increasing the expression levels of the ATF6 and Bip proteins and upregulating CHOP and cleaved PARP. Conclusion: Compound KA-4c triggers ER stress response and induces apoptosis via the ATF6-CHOP signaling pathway.


Assuntos
Apoptose , Estresse do Retículo Endoplasmático , Fator de Transcrição CHOP/metabolismo , Chaperona BiP do Retículo Endoplasmático , Transdução de Sinais
3.
Planta Med ; 88(13): 1233-1244, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35104905

RESUMO

The aim of the present study was to investigate the cytotoxic effects and underlying molecular mechanisms of nitidine chloride (NC) in hepatocellular carcinoma cells via quantitative proteomics. MTT assays were used to detect the inhibitory effects of NC in Bel-7402 liver cancer cells, and the number of apoptotic cells was measured by flow cytometry. Quantitative proteomics technology based on iTRAQ was used to discover differential expressed proteins after NC treatment, and bioinformatic techniques were further used to screen potential targets of NC. Molecular docking was applied to evaluate the docking activity of NC with possible upstream proteins, and their expression was detected at the mRNA and protein levels by quantitative reverse transcription PCR and western blotting. NC inhibited the proliferation of Bel-7402 cells after 24 h of treatment and stimulated apoptosis in vitro. The proteomics experiment showed that NC triggers mitochondrial damage in HCC cells and transcription factor AP-1 (c-Jun) may be a potential target of NC (fold change = 4.36 ± 0.23). Molecular docking results revealed the highest docking score of NC with c-Jun N-terminal kinase (JNK), one of the upstream proteins of c-Jun. Moreover, the mRNA and protein expression of c-Jun and JNK were significantly increased after NC treatment (p < 0.05). These findings indicate that NC significantly induced mitochondrial damage in HCC cells, and induced apoptosis by activating JNK/c-Jun signaling.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Fator de Transcrição AP-1/metabolismo , Proteômica , Simulação de Acoplamento Molecular , Linhagem Celular Tumoral , Apoptose , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , RNA Mensageiro
4.
Int J Oncol ; 58(5)2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33760108

RESUMO

At present, effective therapeutic drugs for triple­negative breast cancer (TNBC) are lacking due to the absence of identified or available targets. Therefore, the present study aimed to identify key molecular targets and a specific targeted therapeutic drug to aid with the development of novel therapeutic strategies for TNBC. Based on the high expression of EGFR and Rac1 in TNBC and inspired by a novel antitumor strategy termed combi­targeting, novel anthraquinone­quinazoline hybrid 7B was synthesized to simultaneously target EGFR and Rac1. It was hypothesized that hybrid 7B may possess enhanced potency compared with its parent compounds. Breast cancer cell viability was detected by performing MTT assays. Flow cytometry was conducted to detect the effects of hybrid 7B on the cell cycle, apoptosis and the mitochondrial outer membrane potential. Ultrastructural alterations were observed by transmission electron microscopy. Cell invasion and migration were assessed by performing Transwell and wound­healing assays, respectively. The expression levels of epithelial­mesenchymal transition (EMT) markers and metastasis­related proteins were detected by western blotting. Compared with Rhein and gefitinib, hybrid 7B displayed superior antiproliferative activity in MDA­MB­231 cells with an IC50 value of 2.31 µM, which was 14­fold higher compared with the EGFR tyrosine kinase inhibitor gefitinib. Further experiments demonstrated that hybrid 7B significantly reduced the mitochondrial membrane potential, enhanced MDA­MB­231 cell apoptosis and induced cell cycle arrest at the G2/M phase compared with the control group. Typical morphological alterations of apoptotic cells were observed in hybrid 7B­treated MDA­MB­231 and MCF­7 cells. Compared with the control group, hybrid 7B significantly inhibited MDA­MB­231 cell invasion and migration by downregulating Rac1, EGFR, matrix metalloproteinases, snail family transcriptional repressor 1, Vimentin and ß­catenin protein expression levels, and upregulating E­cadherin protein expression levels. The present study demonstrated that hybrid 7B inhibited TNBC cell migration and invasion by reversing EMT and targeting EGFR and Rac1; therefore, hybrid 7B may serve as a promising therapeutic agent for TNBC.


Assuntos
Antraquinonas/farmacologia , Quinazolinas/farmacologia , Neoplasias de Mama Triplo Negativas/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Antraquinonas/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Receptores ErbB/química , Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Modelos Moleculares , Simulação de Acoplamento Molecular , Metástase Neoplásica , Quinazolinas/química , Transdução de Sinais/efeitos dos fármacos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Proteínas rac1 de Ligação ao GTP/química
5.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 36(10): 918-923, 2020 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-33148387

RESUMO

Objective To establish a triple negative breast cancer cell line stably expressing human epidermal growth factor receptor (EGFR) promoter and luciferase (Luc) reporter gene and to preliminarily verify its application. Methods Using genetic recombination technology, the lentiviral vector carrying Luc reporter and EGFR promoter sequence was designed and constructed to infect MDA-MB231 cells and obtain MDA-MB231-EGFR-Luc2 cell lines by the selection with puromycin. The Luc luminescence value after stimulating with EGFR activator EGF or inhibitor gefitinib regulating the EGFR promoter activities was detected. Results Gene sequencing and enzyme digestion verified that the lentiviral expression vector carrying Luc reporter vector recombined with EGFR promoter was successfully constructed. Lentivirus-infected MDA-MB231 cells were screened by puromycin, the MDA-MB231-EGFR-Luc2 cells stably expressing firefly Luc was obtained. EGF increased the Luc luminescence value of MDA-MB231-EGFR-Luc2 cells in a dose-dependent manner, while gefitinib did the opposite. Conclusion The cell line of MDA-MB231-EGFR-Luc2 containing EGFR promoter and Luc reporter gene has been successfully constructed, which provides a new cell model for high throughput screening of EGFR-targeting drugs.


Assuntos
Genes Reporter , Regiões Promotoras Genéticas , Neoplasias de Mama Triplo Negativas/genética , Linhagem Celular Tumoral , Receptores ErbB/genética , Humanos , Luciferases/genética
6.
Eur J Pharmacol ; 886: 173526, 2020 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-32890460

RESUMO

Ovarian cancer is the leading cause of death among gynecologic cancer patients. Although platinum-based chemotherapy as a frontline treatment for ovarian cancer has been widely used in clinical settings, its clinical efficacy is not satisfactory due to the resistance of ovarian cancer cells to apoptosis. Therefore, it is of great significance to induce non-apoptotic programed cell death patterns, such as paraptosis, in ovarian cancer. In this study, we aimed to explore the potential anticancer mechanisms of novel rhein derivative 4a, which was modified with rhein as a lead compound. The results showed that a wide range of vacuoles from the endoplasmic reticulum and mitochondria appeared in ovarian SKOV3, SKOV3-PM4, and A2780 cells treated with derivative 4a, and the cell death caused by derivative 4a is a type of non-apoptotic and non-autophagic death, which is caused by expansion and damage of the endoplasmic reticulum or mitochondria, showing the characteristics of para-apoptotic death. Furthermore, derivative 4a stimulated the unfolded protein reaction of ovarian cancer cells by upregulating the expression of Bip78 and activating the PERK-eIF2α-ATF4 pathways. Notably, rhein derivative 4a-induced cell death was positively correlated with activation of p38, ERK, and JNK, and negatively correlated with Alix, a known protein that inhibits paraptosis. In addition, derivative 4a treatment also induced G2/M phase arrest in ovarian cancer cells. Taken together, our study reveals that derivative 4a induces paraptosis, and this finding can serve as a basis in developing a new strategy for the treatment of antiapoptotic ovarian cancer.


Assuntos
Antraquinonas/farmacologia , Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Neoplasias Ovarianas/tratamento farmacológico , Autofagia/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Feminino , Humanos , Mitocôndrias/efeitos dos fármacos , Neoplasias Ovarianas/patologia , Transdução de Sinais/efeitos dos fármacos , Resposta a Proteínas não Dobradas/efeitos dos fármacos
7.
Front Pharmacol ; 11: 754, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32547389

RESUMO

BACKGROUND: Triple-negative breast cancer is a common malignant tumor with unfavorable prognosis affecting women worldwide; thus, there is an urgent need for novel therapeutic drugs with improved anti-tumor activity. Rac family small GTPase 1 (Rac1) plays an important role in malignant behavior and is a promising therapeutic target. We reported an anthraquinone compound, Rhein, and its derivative, 4F, and investigated their downregulation effects on Rac1 in breast cancer cells in vitro. METHODS: The inhibition of cell proliferation by derivative 4F was investigated in two breast cancer (MDA-MB-231 and MCF-7) and normal breast (MCF-10A) cell lines by cell counting kit-8 assay and growth curves. The role of 4F in cell migration and invasion and cytoskeletal change were assessed by Transwell chamber assay and F-actin staining, respectively. The affinity of Rhein and its derivative for Rac1 protein and the regulation of Rac1 promoter activity were evaluated by molecular docking software and luciferase reporter gene assay, respectively. Rac1 protein expression was determined by western blot assay. RESULTS: Compared to Rhein, derivative 4F more strongly inhibited breast cancer cell proliferation, migration, and invasion and also cause cytoskeletal changes like those in paclitaxel. Derivative 4F not only bound more stably to Rac1 but also inhibited Rac1 promoter activity in cells and downregulated Rac1 protein expression. CONCLUSIONS: Rhein derivative 4F is a new anthraquinone compound with better anti-tumor activity than that of the lead compound Rhein in breast cancer. It down-regulated Rac1 expression and may be a small molecule inhibitor of Rac1.

8.
Eur J Pharm Sci ; 151: 105378, 2020 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-32454130

RESUMO

Radiotherapy has a high cure rate for early nasopharyngeal carcinoma(NPC). However, the radiation resistance of poorly differentiated NPC cells impacts the effectiveness of treatment of early-stage NPC patients. Here, we explored the relationship between Ras-related C3 botulinum toxin substrate 1(Rac1) expression and NPC radiosensitivity. In vitro and in vivo studies revealed that upregulation of Rac1, when combined with X-ray treatment, increased growth inhibition and induced remarkable morphological changes and apoptosis in CNE2 cells. Furthermore, rupturing of the cell and nuclear membranes, degeneration of the cristae and significant swelling of the mitochondria were observed, which were consistent with the high apoptotic rate. The Rac1(+) cells exhibited approximately 50% more migration compared with that of the NC and Rac1(-) cells. The overexpression of Rac1 can increase the radiation sensitivity of NPC CNE2 cells, and the mechanism may be closely related to the oxidative damage of mitochondria. Rac1 might be a potential target for radiosensitization in poorly differentiated NPC.


Assuntos
Carcinoma , Neoplasias Nasofaríngeas , Carcinoma/radioterapia , Linhagem Celular Tumoral , Humanos , Carcinoma Nasofaríngeo/radioterapia , Neoplasias Nasofaríngeas/radioterapia , Tolerância a Radiação , Proteínas rac1 de Ligação ao GTP
9.
Future Med Chem ; 12(7): 627-644, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32175770

RESUMO

Anthraquinones exhibit a unique anticancer activity. Since their discovery, medicinal chemists have made several structural modifications, resulting in the design and synthesis of a large number of novel anthraquinone compounds with different biological activities. In general, anthraquinone compounds have been considered to have anticancer activity mainly through DNA damage, cycle arrest and apoptosis. However, recent studies have shown that novel anthraquinone compounds may also inhibit cancer through paraptosis, autophagy, radiosensitising, overcoming chemoresistance and other methods. This Review article provides an overview of novel anthraquinone compounds that have been developed as anticancer agents in recent years and focuses on their anticancer mechanism.


Assuntos
Antraquinonas/farmacologia , Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Animais , Antraquinonas/química , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Humanos , Estrutura Molecular , Neoplasias/metabolismo , Neoplasias/patologia
10.
Future Med Chem ; 12(2): 111-126, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31718309

RESUMO

Aim: The EGF receptor (EGFR) is overexpressed in multiple epithelial-derived cancers and is considered to be a vital target closely associated with cancer therapy. In this study, a series of novel anthraquinone-quinazoline hybrids targeting several vital sites for cancer therapy were designed and synthesized. Methodology & results: Most of the synthesized hybrids demonstrated excellent antiproliferative activity and downregulation of the expression of EGFR. The most promising compound 7d showed the strongest antiproliferation activity; this compound significantly downregulated the expression of p-EGFR protein, induced a remarkable apoptosis effect, promoted the rearrangement of F-actin filaments and destruction of cytoskeleton, induced DNA damage and enhanced radiosensitivity of A549 cells. Conclusion: The novel anthraquinone-quinazoline hybrid 7d emerges as an anticancer drug candidate with promising multitargeted biological activities.


Assuntos
Antraquinonas/farmacologia , Antineoplásicos/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Antraquinonas/química , Antineoplásicos/síntese química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dano ao DNA , Avaliação Pré-Clínica de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Humanos , Estrutura Molecular , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Quinazolinas/química , Células Tumorais Cultivadas
11.
Anticancer Drugs ; 30(10): 1038-1047, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31274517

RESUMO

Triple negative breast cancer (TNBC) has very poor prognosis and no efficacious therapeutic options due to the absence of a validated molecular target. Therefore, novel therapeutic strategies against TNBC are urgently needed. Our team synthesized and screened a series of compounds derived from Rhein, of which 4F was selected for further analysis based on its ability to produce the vacuolated appearance of cells. Using Cell counting kit-8 assay, colony-formation assay, cell apoptosis and cell cycle assay, we compared the antitumor effects of 4F, Rhein and Cisplatin on a TNBC cell line MDA-MB-231 in vitro. The vacuoles in MDA-MB-231 cells were observed and analyzed by hematoxylin-eosin staining and transmission electron microscopy. Autophagy and apoptosis-related proteins including p62, Microtubule Light Chain 3 (LC3), Beclin-1 and Caspase-3 were determined by western blot. The tandem mRFP-GFP-LC3 Lentivirus was used for monitoring the maturation step of autophagosomes. Our data revealed that 4F had lower cytotoxicity to normal breast cell line MCF-10A as compared with positive drug Doxorubicin. Although 4F had better cytotoxicity than Rhein, it had no influence on cells apoptosis in 4F-treated cells. Accumulation of autolysosomes and autophagosomes was observed in 4F-treated MDA-MB-231 cells, accompanied by increased level of Beclin-1 protein. Enhanced autophagic flux was verified by higher ratio of LC3-II/LC3-I, the degradation of p62 protein and alteration in red and green fluorescence puncta. These findings suggested that the process of MDA-MB-231 cell death induced by 4F seemed rely mainly on autophagy rather than apoptosis. 4F may be an alternative drug candidate against TNBC and merits more exploration.


Assuntos
Antraquinonas/química , Antraquinonas/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacologia , Morte Celular Autofágica/efeitos dos fármacos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Estrutura Molecular , Neoplasias de Mama Triplo Negativas/patologia , Vacúolos/efeitos dos fármacos
12.
ACS Med Chem Lett ; 10(5): 732-736, 2019 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-31097991

RESUMO

Novel anthraquinone compounds that induce ER stress and paraptosis-like cell death were designed and synthesized. Compound 4a is the first organic micromolecule to kill tumor cells by only paraptosis, and its mechanism of action has been further explored. Paraptosis does not appear to involve either phosphatidylserine translocation associated with apoptosis or cell cycle arrest. The bisbenzyloxy and N-(2-hydroxyethyl)formamide structures may be two critical pharmacophores for paraptosis. Bisbenzyloxy can induce ER stress, and the N-(2-hydroxyethyl)formamide structure can increase the ratio of LC3II/I and cytoplasmic vacuolization and facilitates paraptosis. Some antitumor drugs fail to eradicate malignant cell lines with impaired apoptotic pathways; paraptosis may be a route to kill such cells and provides a new potential strategy for cancer chemotherapy.

13.
Cell Signal ; 54: 35-45, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30463023

RESUMO

Radiation resistance and recurrent have become the major factors resulting in poor prognosis in the clinical treatment of patients with nasopharyngeal carcinoma (NPC). New strategies to enhance the efficacy of radiotherapy have been focused on the development of radiosensitizers and searching for directly targets that modulated tumor radiosensitivity. A novel potential radiosensitizer 1,8-Dihydroxy -3-(2'-(4″-methylpiperazin-1″-yl) ethyl-9,10-anthraquinone -3-carboxylate (RP-4) was designed and synthesized based on molecular docking technology, which was expected to regulate the radiosensitivity of tumor cells through targeting Rac1. In order to assess the radiosensitization activity of RP-4 on NPC cells, the highly differentiated CNE1 and poorly differentiated CNE2 cells NPC lines were employed. According to the results, RP-4 showed higher binding affinity toward the interaction with Rac1 than lead compounds. We found that RP-4 could inhibit cell viability and proliferation in CNE1 and CNE2 cells and significantly induced apoptosis after non-toxic concentration of RP-4 combined with 2Gy irradiation. RP-4 could effectively modulated the radiosensitivity both CNE1 cells and CNE2 cells through activating Rac1/NADPH signaling pathway and its downstream JNK/AP-1 pathway. What's more, Rac1/NADPH signaling pathway were significantly activated in Rac1-overexpressed CNE1 and CNE2 cells after treated with RP-4. Taken together, Rac1 and its downstream pathway may probably be the direct targets of RP-4 in regulating radiosensitivity of NPC cells, our finding provided a novel strategy for the development of therapeutic agents in response to tumorous radiation resistance.


Assuntos
Antraquinonas , Carcinoma Nasofaríngeo/radioterapia , Neoplasias Nasofaríngeas/radioterapia , Tolerância a Radiação/efeitos dos fármacos , Radiossensibilizantes/farmacologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Antraquinonas/síntese química , Antraquinonas/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Humanos , NADP/metabolismo
14.
Phytother Res ; 31(12): 1962-1970, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29044868

RESUMO

Prismatomeris connata was a kind of Rubiaceae plant for treatment of hepatitis, hepatic fibrosis and silicosis. Whereas, the effective components of Prismatomeris connata remains unexplored. The aim of this study was to investigate the inhibitory effects and mechanisms of Rubiadin isolated from Prismatomeris connata against HBV using HepG2.2.15 cells. The levels of hepatitis B surface antigen (HBsAg), hepatitis B e antigen (HBeAg), and hepatitis B core antigen (HBcAg) in the supernatants or cytoplasm were examined using by enzyme-linked immunosorbent assay. HBV DNA was qualified q-PCR. Rubiadin was isolated by silica gel column. The structure of the compound was elucidated by HPLC, FT-IR, 1 H-NMR, 13 C-NMR and identified as 1,3-Dihydroxy-2-methyl-9, 10-anthraquinone. Rubiadin significantly decreased HBeAg,HBcAg secretion level and inhibit HBV DNA replication. Rubiadin inhibits the proliferation of the cells and HBx protein expression in a dose-dependent manner. The intracellular calcium concentration was significantly reduced. These results demonstrated that Rubiadin could inhibit HepG2.2.15 cells proliferation, reduce the level of HBx expression, and intracellular free calcium, which might become a novel anti-HBV drug candidate.


Assuntos
Antraquinonas/química , Vírus da Hepatite B/efeitos dos fármacos , Anticorpos Anti-Hepatite C/metabolismo , Raízes de Plantas/química , Rubiaceae/química , Humanos
15.
J Cell Biochem ; 118(10): 3150-3157, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28004426

RESUMO

The aim of this study was to investigate the possibility of APC/CCdh1 as a potential therapeutic target in the radiosensitivity of nasopharyngeal carcinoma (NPC) cell CNE-1, and explain the role of APC subunits after silence of Cdh1 combined with radiotherapy. Transfection with Cdh1 shRNA significantly increased the radiosensitivity of CNE-1 cells and the radiation enhancement ratio (RER) of sh-Cdh1 cells was 1.76. Knockdown of Cdh1 in CNE-1 cells increased irradiation induced apoptosis and G2/M phase cell cycle arrest. The levels of CDC20 and CylinB1 increased and the levels of Ku70 and APC3 decreased after irradiation. APC/CCdh1 is involved in regulation of radiosensitivity in human NPC CNE-1 cells. Our study may provide a promising therapeutic strategy for NPC by targeting Cdh1. J. Cell. Biochem. 118: 3150-3157, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Subunidade Apc3 do Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Apoptose , Caderinas/metabolismo , Carcinoma/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Tolerância a Radiação , Antígenos CD , Subunidade Apc3 do Ciclossomo-Complexo Promotor de Anáfase/genética , Caderinas/genética , Carcinoma/genética , Carcinoma/radioterapia , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/radioterapia
16.
Radiat Res ; 185(6): 638-46, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27243897

RESUMO

We reported in an earlier study that using mass spectrometry and bioinformatic analysis demonstrated Rac1 protein might be mostly mitochondrial target in the radiosensitization process of nasopharyngeal carcinoma CNE-1 cells. The goal of our current study was to reveal the relationship between Rac1/NADPH pathway and radiosensitization in CNE-1 cells using Rac1 activator, phorbol 12-myristate 13-acetate (PMA) and Rac1 inhibitor NSC23766. The Rac1-GTP expression was determined using a pulldown assay, the Rac1 location using a immunofluorescence with a laser scanning confocal microscope, the NADPH oxidase activity with NBT assay and the reactive oxygen species with DCFH-DA probe. The apoptosis rate was analyzed by flow cytometry, and the expressions of p67(phox) and NFκB-p105/p50 were analyzed by Western blot. After treatment with PMA and 2 Gy radiation (compared to the control), Rac1-GTP was activated and translocated to the cell membrane. NADPH oxidase activity, reactive oxygen species of intracellular concentration and the apoptosis rate increased significantly. The expression of p67(phox) and NFκB-p50 protein also increased. However, in the cells treated with NSC23766 alone or NSC23766 combined with 2 Gy irradiation, the results were just the opposite. Overall, these results indicate that the Rac1 protein may be the key target involved in the radiosensitization of nasopharyngeal carcinoma cells. The activated Rac1/NADPH pathway combined with radiation can increase the radiosensitivity of nasopharyngeal carcinoma cells, and the Rac1/NADPH pathway may be the signaling pathway involved in the radiosensitization process.


Assuntos
NADP/metabolismo , Neoplasias Nasofaríngeas/patologia , Tolerância a Radiação , Transdução de Sinais/efeitos da radiação , Proteínas rac1 de Ligação ao GTP/metabolismo , Aminoquinolinas/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Carcinoma , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/efeitos da radiação , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Espaço Intracelular/efeitos da radiação , NADPH Oxidases/metabolismo , NF-kappa B/metabolismo , Carcinoma Nasofaríngeo , Fosfoproteínas/metabolismo , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/efeitos da radiação , Pirimidinas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Acetato de Tetradecanoilforbol/farmacologia , Raios X
17.
Eur J Pharmacol ; 738: 133-41, 2014 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-24877689

RESUMO

In our preliminary study, 1,8-dihydroxy-3-acetyl-6-methyl-9,10 anthraquinone (GXHSWAQ-1), synthesised according to the basic structure of emodin, exhibited a 1.58-fold radiosensitisation on nasopharyngeal carcinoma CNE-1 cells. This study demonstrated that its radiosensitisation activity was achieved by altering the mitochondrial structure: swollen volume, fragmented crista, and decreasing transmembrane potential (P<0.01). Using isobaric tag for relative and absolute quantitation (iTRAQ) technology, 1396 proteins were identified, and the differentially expressed proteins were involved in metabolism, cell proliferation, angiogenesis, DNA repair process according to the biological process clustering results. Bioinformatic analysis showed that CDH1, RAC1, CDC42 proteins might be mostly mitochondrial targets in the radiosensitisation process. Western blotting analyses verified the differential expression of these proteins.


Assuntos
Antraquinonas/farmacologia , Proteínas Mitocondriais/metabolismo , Neoplasias Nasofaríngeas/patologia , Tolerância a Radiação/efeitos dos fármacos , Carcinoma , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos da radiação , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos da radiação , Carcinoma Nasofaríngeo , Proteômica , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação
18.
Mol Med Rep ; 10(2): 965-70, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24912934

RESUMO

1,8­dihydroxy­3­acetyl­6­methyl­9,10 anthraquinone (DAMA) was synthesized from emodin. In the present study, the activity and the oncosis­like mechanism of DAMA­enhanced nasopharyngeal carcinoma (NPC) cell sensitivity to ionizing radiation was examined. The results demonstrated that DAMA has a 1.46­fold radiosensitisation activity for nasopharyngeal carcinoma CNE 1 cells with a non­cytotoxic concentration of 10 µg/ml DAMA combined with 2 Gy. Following treatment of DAMA combined with radiation, CNE­1 cells revealed severe cytoplasmic swelling and vacuolization, swollen mitochondria and dilation of the nuclei without chromatin condensation, yielding a typical morphology of oncosis. Oncosis­related gene expression of ATP synthase F0 subunit 6, chromatin modifying protein 6 and cyclophilin D mRNA increased significantly in the 8 Gy radiation group and the 2 Gy radiation combined with DAMA group. A significant decrease of ATP synthase protein 8 mRNA was observed and the levels of intracellular ATP were also reduced. In addition, the levels of intracellular Ca2+ were increased. In conclusion, DAMA is a potent radiation sensitizer in nasopharyngeal carcinoma cells and mediates its radiosensitisation via oncosis.


Assuntos
Antraquinonas/toxicidade , Apoptose/efeitos dos fármacos , Radiossensibilizantes/toxicidade , Trifosfato de Adenosina/metabolismo , Antraquinonas/química , Apoptose/efeitos da radiação , Cálcio/metabolismo , Carcinoma , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/ultraestrutura , Peptidil-Prolil Isomerase F , Ciclofilinas/genética , Ciclofilinas/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Raios gama , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos da radiação , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , ATPases Mitocondriais Próton-Translocadoras/genética , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patologia , Organelas/efeitos dos fármacos , Organelas/ultraestrutura , Radiossensibilizantes/química
19.
J Pharm (Cairo) ; 2013: 218297, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-26555969

RESUMO

Here, we report that regulation of cellular redox status is required for radiosensitization of nasopharyngeal carcinoma (NPC) cells by emodin. We evaluated emodin's radiosensitivity-enhancing ability by using NPC cells in vitro and xenografts in vivo. A clonogenic assay was performed to evaluate NPC cell survival and to determine dose modification factors. Flow cytometry, western blot analysis, and in vivo radiation-induced tumor regrowth delay assays were performed to characterize emodin's effects. Exposure of CNE-1 NPC cells to emodin enhanced their radiosensitivity. HIF-1α expression significantly increased under hypoxic conditions but did not change after treatment with emodin alone. Emodin downregulated mRNA and protein expression of HIF-1α. Cells exposed to radiation and emodin underwent significant cell cycle arrest at the G2/M phase. The percentage of apoptotic cells and reactive oxygen species (ROS) levels were significantly higher in the group exposed to emodin and radiation hypoxic group than in the other groups. Compared to the CNE-1 xenografts exposed to radiation alone, CNE-1 xenografts exposed to radiation with emodin showed significantly enhanced radiation effects. Our data suggest that emodin effectively enhanced the radiosensitivity of CNE-1 cells in vitro and in vivo. The mechanism appears to involve ROS generation and ROS-mediated inhibition of HIF-1α expression.

20.
Ai Zheng ; 22(8): 801-5, 2003 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-12917023

RESUMO

BACKGROUND & OBJECTIVE: Inducing apoptosis and inhibiting the telomerase activity of tumor cells became a new therapeutic means for tumor. In vivo and in vitro experiments showed that wogonin possesses antioxidant activities and inhibitory effect on tumor cells growth. This study was designed to evaluate the effect of wogonin on telomerase activity and apoptosis of human ovarian carcinoma cell line A2780. METHODS: MTT assay,fluorescent microscopy,and DNA agarose gel electrophoresis were used to determine the role of wogonin on apoptosis of A2780 cells. The telomerase activity of A2780 cells were observed by using TRAP-ELASA method. RESULTS: A2780 cell growth was significantly inhibited by wogonin. The inhibiting effect showed concentration-dependent and time-dependent manners with IC(50) of 85 microg/ml. After treatment with 50 microg/ml and 100 microg/ml wogonin for 48 hours, A2780 cells showed morphological changes associated with the characters of apoptosis under fluorescent microscope. Typical DNA ladder was found using agarose gel electrophoresis. Telomerase activity of A2780 cells was gradually decreased with the increasing of wogonin concentration. When the concentration of wogonin was higher than 200 microg/ml, telomerase activity of A2780 cells was inhibited markedly. CONCLUSION: Wogonin can inhibit proliferation and induce apoptosis of A2780 cells within a certain concentration range(50-250 microg/ml). Anticancer effects of wogonin were associated with the induction of apoptosis and partly with the suppression of telomerase activity.


Assuntos
Apoptose/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Flavanonas/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Telomerase/antagonistas & inibidores , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA