Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Oncol Lett ; 26(2): 327, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37415631

RESUMO

Gastric cancer is one of the most lethal cancers worldwide. Research has focused on exploring natural medicines to improve the systematic chemotherapy for gastric cancer. Luteolin, a natural flavonoid, possesses anticancer activities. Nevertheless, the mechanism of the anticancer effects of luteolin is still not clear. The present study aimed to verify the inhibitory effect of luteolin on gastric cancer HGC-27, MFC and MKN-45 cells and to explore the underlying mechanism. A Cell Counting Kit-8 cell viability assay, flow cytometry, western blot, an ATP content assay and an enzyme activity testing assay were used. Luteolin inhibited the proliferation of gastric cancer HGC-27, MFC and MKN-45 cells. Further, it impaired mitochondrial integrity and function by destroying the mitochondrial membrane potential, downregulating the activities of mitochondrial electron transport chain complexes (mainly complexes I, III and V), and unbalancing the expression of B cell lymphoma-2 family member proteins, eventually leading to apoptosis of gastric cancer HGC-27, MFC and MKN-45 cells. The intrinsic apoptosis pathway was involved in luteolin's anti-gastric cancer effects. Furthermore, mitochondria were the main target in luteolin-induced gastric cancer apoptosis. The present study may provide a theoretical basis for the research on the effect of luteolin on the mitochondrial metabolism in cancer cells, and pave the way for its practical application in the future.

2.
Biomed Res Int ; 2022: 3768880, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36033571

RESUMO

Dihuang Yinzi, as a classical Chinese medicine prescription, plays an important role for the treatment of ischemic stroke. Gut microbiota play a functional role for the expression of proinflammatory cytokines and anti-inflammatory cytokines, which further affect central nervous system and change brain function. Our research confirmed that Dihuang Yinzi can exert brain protection by inhibiting inflammatory reaction. Dihuang Yinzi can significantly decrease the contents of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and interleukin-17 (IL-17) in brain, serum, and colon tissues and increase the contents of transforming growth factor-ß (TGF-ß) and interleukin-10 (IL-10) in cerebral ischemia-reperfusion model rats. The results of 16s rRNA high-throughput sequencing showed that Dihuang Yinzi had a significant effect on microbiome in rats. The firmicutes, bacteroidetes, and proteobacteria were dominant in Dihuang Yinzi group. The content of firmicutes increased with the increase of dosage of Dihuang Yinzi. Especially, the content of actinomycetes in the high-dose group was higher than other groups. At the genus level, the number of bacteroides in the antibiotic groups was significantly higher than that in the other treatment groups. The results suggest that Dihuang Yinzi may play important roles in treatment of ischemic stroke by regulating the gut microbiota and the inflammatory reaction in the colon tissues, serum, and brain of the model rats, to verify the scientific nature of this prescription in relieving brain inflammatory reaction and brain injury by this way and to reveal the brain-gut related mechanism of Dihuang Yinzi in treating ischemic stroke.


Assuntos
Isquemia Encefálica , Microbioma Gastrointestinal , AVC Isquêmico , Animais , Infarto Cerebral , Citocinas , Inflamação , RNA Ribossômico 16S , Ratos , Ratos Sprague-Dawley , Reperfusão
3.
Oncol Lett ; 23(1): 16, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34820015

RESUMO

Although the reduction of oxaliplatin doses may alleviate deleterious side effects of gastrointestinal and gynecological cancer treatment, it also limits the anticancer therapeutic effects. As a high-efficient and low-priced herbal medicine ingredient, luteolin is an agent with a broad spectrum of anticancer activities and acts as a potential enhancer of therapeutic effects of chemotherapy agents in cancer treatment. This study focused on the antitumor effects and mechanism of combined treatment with luteolin and oxaliplatin on a mouse forestomach carcinoma (MFC) cell line. The study used CCK-8 assay, flow cytometry, Annexin V-FITC/PI double staining assay, reactive oxygen species testing assay, mitochondrial membrane potential testing assay, and western blot assay. The results showed that luteolin and oxaliplatin exerted synergistic effects on inhibiting MFC cell proliferation by inducing G2/M cell cycle arrest and apoptosis. Inhibiting the tumor necrosis factor receptor-associated protein 1/phosphorylated-extracellular-regulated protein kinases1/2/cell division cycle 25 homolog C/cyclin-dependent kinase-1/cyclin B1 pathway was indispensable to the combined treatment with luteolin and oxaliplatin to induce G2/M cell cycle arrest. In addition, luteolin increased oxidative stress in MFC cells treated with a low dose of oxaliplatin. The combined therapy damaged mitochondrial membrane potential and regulated BCL-2-associated X protein and B-cell lymphoma 2 protein expression, leading to apoptosis. Findings of the present study suggest that luteolin may be a qualified chemotherapy enhancer to potentiate the anticancer effects of low-dose oxaliplatin in MFC cells. This work provides a theoretical foundation for future research on applications of luteolin in clinical chemotherapy.

4.
Oncol Lett ; 21(6): 453, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33907563

RESUMO

Research suggests that daphnoretin exhibits a diverse array of antitumor mechanisms and pharmacological activities. However, there is no definitive explanation for the antitumor mechanisms of daphnoretin in malignant melanoma. In the present study, MTT and colony formation assays demonstrated that daphnoretin significantly inhibited the proliferation of melanoma A375 and B16 cells. Following treatment with daphnoretin, apoptotic bodies were observed in A375 and B16 cells via Hoechst 33258 staining. Furthermore, western blot analysis revealed that the apoptosis-related proteins cleaved caspase-3, cleaved caspase-9, Bax, cytochrome c and apoptotic protease-activating factor 1 were significantly upregulated, while the expression levels of caspase-3, caspase-9 and Bcl-2 were downregulated in A375 and B16 cells. Flow cytometry and fluorescence microscopy revealed that daphnoretin induced higher levels of reactive oxygen species (ROS). Therefore, the results of the present study indicated that daphnoretin induced ROS-mediated mitochondria apoptosis in human (A375) and murine (B16) malignant melanoma cells.

5.
Biomed Pharmacother ; 133: 111042, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33378950

RESUMO

The application of photodynamic therapy (PDT) for the treatment of skin diseases has been receiving much attention. Here, we examined the anti-tumor effect of a novel porphyrin-based photosensitizer TBPoS-2OH in the malignant melanoma A375 and B16 cells. TBPoS-2OH has obvious cell photo-cytotoxicity, but it has low cell dark-cytotoxicity. Further research showed that TBPoS-2OH is enriched in lysosomes after being taken up by cells. Subsequently, the apoptotic rates were significantly increased in TBPoS-2OH-treated A375 and B16 cells. The specific mechanism may be that after receiving light stimulation, TBPoS-2OH could effectively increase the level of intracellular reactive oxygen species (ROS), thereby activating mitochondrial apoptosis pathway-related proteins in A375 and B16 cells. We found an increase in the content of cytochrome C in the cytoplasm, and the levels of related proteins, such as cleaved caspase-3, cleaved caspase-9, and cleaved PARP1, were significantly increased in TBPoS-2OH-treated cells. These results indicated that the new compound TBPoS-2OH could be developed and become an alternative drug for the treatment of melanoma. Some reference ideas for the development of new photosensitizers are also provided.


Assuntos
Melanoma/tratamento farmacológico , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Neoplasias Cutâneas/tratamento farmacológico , Sulfonamidas/farmacologia , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia
6.
Eur J Med Chem ; 209: 112867, 2021 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-33010634

RESUMO

Photodynamic therapy (PDT) is an emerging alternative cancer treatment modality that utilizes photo-sensitivity to cause cell death upon photo-irradiation. However, PDT efficiency has been hampered by tumor hypoxia, blue-shifted excitation wavelengths, and the high dark toxicity of photo-sensitizers. We designed and synthesized two novel porphycene-based photosensitizers (TBPoS-OH and TBPoS-2OH) with potent photo-cytotoxicity and a LD50 in the nM range under both normoxic and hypoxic conditions in a variety of cell types after photo-irradiation (λ = 640 ± 15 nm). Further studies showed fast-cellular uptake for TBPoS-OH that localized lysosomes and subsequently induced cell apoptosis via the lysosomal-mitochondrial pathway. Moreover, TBPoS-OH significantly reduced tumor growth in two xenografted mouse models bearing melanoma A375 and B16 cells. Finally, TBPoS-OH exhibited no obvious immunogenicity and toxicity to blood cells and major organs in mice. These data demonstrated that these two porphycene-based photosensitizers, especially TBPoS-OH, could be developed as a potential PDT modality.


Assuntos
Melanoma/tratamento farmacológico , Fármacos Fotossensibilizantes/uso terapêutico , Porfirinas/uso terapêutico , Sulfonamidas/uso terapêutico , Hipóxia Tumoral/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Cristalografia por Raios X , Humanos , Luz , Masculino , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Fotoquimioterapia , Fármacos Fotossensibilizantes/química , Porfirinas/química , Sulfonamidas/química
7.
Front Oncol ; 10: 558414, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33178586

RESUMO

Background: Existing research shows that p-coumaric acid (p-CA) can inhibit the proliferation of a variety of tumor cells in vitro. However, there are no reports on the anti-tumor effects of p-CA on melanoma cells. In this study, the inhibitory effects of p-CA on mouse melanoma B16 and human melanoma A375 cells are reported, and the related mechanisms are investigated. Methods: CCK-8 assay was used to detect the effects of p-CA on cell vitality, colony formation assay was used to observe the effects on cell proliferation, Hoechst 33,258 staining was used to observe the morphology of apoptotic cells, flow cytometry was used to detect the effects on apoptosis and the cell cycle, and western blot was used to measure the levels of cell cycle- and apoptosis-related signaling pathway proteins. Results: p-CA significantly inhibits cell proliferation of A375 and B16 cells in a dose-dependent manner and obviously induced cell morphological changes. p-CA arrested A375 cells in the S phase by downregulating the cell cycle-related proteins Cyclin A and CDK2, and arrested B16 cells in the G0-G1 phase through downregulating the cell cycle-related proteins Cyclin E and CDK2. In addition, p-CA significantly promoted apoptosis of A375 and B16 cells. Furthermore, p-CA significantly upregulated the levels of Apaf1 and Bax and downregulated the levels of Bcl-2, and subsequently increased the levels of cytoplasmic cytochrome c (Cyto-c), cleaved caspase-3, and cleaved caspase-9, leading to apoptosis in A375 and B16 cells. Conclusion: p-CA can significantly inhibit the proliferation of human and mouse melanoma cells in vitro. Our research is a step in the development of anti-melanoma drugs.

8.
Oxid Med Cell Longev ; 2020: 3237150, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32411325

RESUMO

The botanical constituents of Stellera chamaejasme Linn. exhibit various pharmacological and medicinal activities. Neochamaejasmin A (NCA), one main active constituent of S. chamaejasme, inhibits cell proliferation and induces cell apoptosis in several types of tumor cells. However, the antitumor effect of NCA on hepatocellular carcinoma cells is still unclear. In this study, NCA (36.9, 73.7, and 147.5 µM) significantly inhibited hepatoblastoma-derived HepG2 cell proliferation in a concentration-dependent manner. Hoechst 33258 staining and flow cytometry showed that apoptotic morphological changes were observed and the apoptotic rate was significantly increased in NCA-treated HepG2 cells, respectively. Additionally, the levels of Bax, cleaved caspase-3, and cytoplasmic cytochrome c were increased, while the level of Bcl-2 was decreased in NCA-treated HepG2 cells when compared with the control group. Moreover, we found that the reactive oxygen species (ROS) level was significantly higher and the mitochondrial membrane potential was remarkably lower in NCA-treated HepG2 cells than in the control group. Further studies demonstrated that the levels of p-JNK and p-ERK1/2 were significantly upregulated in NCA-treated HepG2 cells, and pretreatment with JNK and ERK1/2 inhibitors, SP600125 and PD0325901, respectively, suppressed NCA-induced cell apoptosis of HepG2 cells. In addition, NCA also significantly inhibited human hepatoma BEL-7402 cell proliferation and induced cell apoptosis through the ROS-mediated mitochondrial apoptotic pathway. These results implied that NCA induced mitochondrial-mediated cell apoptosis via ROS-dependent activation of the ERK1/2/JNK signaling pathway in HepG2 cells.


Assuntos
Apoptose/efeitos dos fármacos , Biflavonoides/farmacologia , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Sistema de Sinalização das MAP Quinases , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Antioxidantes/metabolismo , Biflavonoides/química , Carcinoma Hepatocelular/enzimologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neoplasias Hepáticas/enzimologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos
9.
Biomed Pharmacother ; 122: 109763, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31918288

RESUMO

Emerging evidence suggests that cinobufagin, an active ingredient in Venenum Bufonis, inhibits cell proliferation in several tumor cells. However, the anti-tumor effect of cinobufagin on nasopharyngeal carcinoma and the underlying molecular mechanisms are still unclear. In this study, we found that cinobufagin significantly inhibits the proliferation of nasopharyngeal carcinoma HK-1 cells. Further analyses demonstrated that cinobufagin induces cell cycle arrest at the S phase in HK-1 cells through downregulating the levels of CDK2 and cyclin E. Moreover, cinobufagin significantly downregulates the protein level of Bcl-2 and upregulates the levels of Bax, subsequently increasing the levels of cytoplasmic cytochrome c, Apaf-1, cleaved PARP1, cleaved caspase-3, and cleaved caspase-9, leading to HK-1 apoptosis. Furthermore, we found that cinobufagin significantly increases ROS levels and decreases the mitochondrial membrane potential in HK-1 cells. Collectively, these data imply that cinobufagin induces cell cycle arrest at the S phase and induces apoptosis through increasing ROS levels, thereby inhibiting cell proliferation in HK-1 cells. Therefore, cinobufagin is a promising bioactive agent that may contribute to the development of treatment strategies of nasopharyngeal carcinoma.


Assuntos
Apoptose/efeitos dos fármacos , Bufanolídeos/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Carcinoma Nasofaríngeo/tratamento farmacológico , Neoplasias Nasofaríngeas/tratamento farmacológico , Fase S/efeitos dos fármacos , Antineoplásicos/farmacologia , Caspase 3/metabolismo , Caspase 9/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Carcinoma Nasofaríngeo/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Regulação para Cima/efeitos dos fármacos
10.
Oncol Lett ; 18(4): 3880-3886, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31516599

RESUMO

Melanoma is the most malignant type of skin cancer and is resistant to numerous chemotherapeutic and radiotherapy-based treatment approaches due to the activation of rapid and reversible pro-survival signaling pathways. As a result, patients will often present with a poor prognosis. Therefore, novel preventive methods and treatments are urgently required for patients with melanoma. Vitamin C (also known as L-ascorbic acid) is a water-soluble vitamin that is widely used as a dietary additive and has been demonstrated to exhibit anti-cancer properties. In the present study, the effects of vitamin C in human melanoma A375 cells, and the mechanisms underlying these effects were investigated. Vitamin C potently suppressed human melanoma A375 cell proliferation by inducing apoptosis in A375 cells. Induction of apoptosis was related to caspase-9 and caspase-3 activation and the mitochondrial membrane potential of A375 cells significantly decreased in the presence of vitamin C. Furthermore, vitamin C induced apoptosis in A375 cells by activating the Bax- and Bcl-2-mediated mitochondrial pathway. These results indicate that vitamin C may be a potentially useful clinical anti-tumor drug for treating patients with melanoma.

11.
Front Oncol ; 9: 853, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31552178

RESUMO

Emerging evidence has shown that cinobufagin, as an active ingredient of Venenum Bufonis, inhibits tumor development. The aim of this study was to investigate the inhibitory effects of cinobufagin on A375 human malignant melanoma cells. MTT and colony formation assays showed that cinobufagin significantly inhibited A375 cell proliferation and cell colony formation. Additional studies demonstrated that cinobufagin markedly increased the levels of ATM serine/threonine kinase (ATM) and checkpoint kinase 2 (Chk2) and decreased the levels of cell division cycle 25C (CDC25C), cyclin-dependent kinase 1 (CDK1), and cyclin B, subsequently inducing G2/M cell cycle arrest in A375 cells. Moreover, cinobufagin clearly inhibited the levels of phosphoinositide 3-kinase (PI3K), phosphorylated PI3K (p-PI3K), AKT, p-AKT, and B-cell lymphoma 2 (Bcl-2). By contrast, it increased the levels of Bcl-2-associated death promoter, Bcl-2-associated X, cytoplasmic cytochrome C, and apoptotic protease activating factor 1, leading to increased levels of cleaved caspase-9 and cleaved caspase-3, resulting in the apoptosis of A375 cells. Together, these results indicate that cinobufagin can induce cell cycle arrest at the G2/M phase and apoptosis, leading to inhibition of A375/B16 cell proliferation. Thus, cinobufagin may be useful for melanoma treatment.

12.
Biomolecules ; 9(7)2019 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-31336757

RESUMO

Malignant melanoma is the most lethal type of skin cancer. Previous studies have shown that ailanthone has potent antitumor activity in a variety of cell lines. However, the anti-tumor effect of ailanthone on malignant melanoma remains unclear. To investigate the anti-tumor mechanisms of ailanthone in human melanoma B16 and mouse melanoma A375 cells, the cell counting kit-8 assay, colony formation assay, DNA content analysis, Hoechst 33258, and Annexin V-FITC/PI staining were used to assess cell proliferation, cell cycle distribution, and cell apoptosis, respectively. Western blotting was performed to evaluate the expression of cell cycle- and apoptosis-related proteins and regulatory molecules. The results showed that ailanthone significantly inhibited melanoma B16 and A375 cell proliferation as well as remarkably induced cell cycle arrest at the G0-G1 phase in B16 cells and the G2-M phase in A375 cells in a dose-dependent manner. Further investigation revealed that ailanthone promoted the expression of p21 and suppressed the expression of cyclin E in B16 cells or cyclin B in A375 cells through the PI3K-Akt signaling pathway. In addition, ailanthone induced B16 and A375 cell apoptosis via a caspase-dependent mechanism. Further studies showed that ailanthone remarkably downregulated Bcl-2 and upregulated Apaf-1 and Bax, and subsequently increased mitochondrial membrane permeabilization and released cytochrome c from the mitochondria in B16 cells and A375 cells. Taken together, ailanthone induces cell cycle arrest via the PI3K-Akt signaling pathway as well as cell apoptosis via the mitochondria-mediated apoptotic signaling pathway. Ailanthone may be potentially utilized as an anti-tumor agent in the management of malignant melanoma.


Assuntos
Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Quassinas/farmacologia , Animais , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/genética , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética
13.
Biomolecules ; 9(8)2019 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-31349651

RESUMO

The main chemical component of cannabis, cannabidiol (CBD), has been shown to have antitumor properties. The present study examined the in vitro effects of CBD on human gastric cancer SGC-7901 cells. We found that CBD significantly inhibited the proliferation and colony formation of SGC-7901 cells. Further investigation showed that CBD significantly upregulated ataxia telangiectasia-mutated gene (ATM) and p53 protein expression and downregulated p21 protein expression in SGC-7901 cells, which subsequently inhibited the levels of CDK2 and cyclin E, thereby resulting in cell cycle arrest at the G0-G1 phase. In addition, CBD significantly increased Bax expression levels, decreased Bcl-2 expression levels and mitochondrial membrane potential, and then upregulated the levels of cleaved caspase-3 and cleaved caspase-9, thereby inducing apoptosis in SGC-7901 cells. Finally, we found that intracellular reactive oxygen species (ROS) increased after CBD treatment. These results indicated that CBD could induce G0-G1 phase cell cycle arrest and apoptosis by increasing ROS production, leading to the inhibition of SGC-7901 cell proliferation, thereby suggesting that CBD may have therapeutic effects on gastric cancer.


Assuntos
Canabidiol/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Redes Reguladoras de Genes/efeitos dos fármacos , Neoplasias Gástricas/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ciclina E/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Neoplasias Gástricas/tratamento farmacológico , Proteína Supressora de Tumor p53/metabolismo
14.
Recent Pat Anticancer Drug Discov ; 14(2): 187-197, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31096897

RESUMO

BACKGROUND: A recent patent has been issued for hydroxysafflor yellow A (HSYA) as a drug to prevent blood circulation disorders. Hydroxysafflor yellow B (HSYB), an isomer of HSYA with antioxidative effects, has been isolated from the florets of Carthamus tinctorius. The effects of HSYB on the proliferation of cancer cells and its mechanism of action have not been investigated. OBJECTIVE: The aims of this study were to investigate the anti-cancer effects and the molecular mechanism of HSYB for breast cancer MCF-7 cells. METHODS: MTT assays and colony formation assays were used to assess the survival and proliferation of MCF-7 cells, respectively. Hoechst 33258 and flow cytometry were used to measure cell apoptosis and flow cytometry to determine effects on the cell cycle. Western blots were used to measure protein levels. RESULTS: Treatment with HSYB reduced survival and proliferation of human breast cancer MCF-7 cells in a dose-dependent manner. Furthermore, HSYB arrested the MCF-7 cell cycle at the S phase and downregulated cyclin D1, cyclin E, and CDK2. Compared with a control group, HSYB suppressed the protein levels of p-PI3K, PI3K, AKT, and p-AKT in MCF-7 cells. In addition, HSYB decreased the levels of Bcl- 2, increased the levels of Bax, cleaved caspase-3 and caspase-9, and subsequently induced MCF-7 cell apoptosis. CONCLUSION: These data demonstrate that HSYB arrests the MCF-7 cell cycle at the S phase and induces cell apoptosis. Patent US20170246228 indicates that HSYB can be potentially used for the prevention and treatment of human breast cancer.


Assuntos
Adenocarcinoma/patologia , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Pigmentos Biológicos/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Chalcona/análogos & derivados , Chalcona/química , Chalcona/farmacologia , Feminino , Humanos , Células MCF-7 , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Pigmentos Biológicos/química , Quinonas/química , Quinonas/farmacologia , Transdução de Sinais/efeitos dos fármacos
15.
Oncol Rep ; 41(4): 2409-2417, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30816542

RESUMO

Venenum bufonis has been used as an antitumor drug in China for many years. Bufotalin, as an active component of Venenum bufonis, has been proven to exhibit antitumor effects in cancer types. In the present study, the effect of bufotalin on the human melanoma skin cancer cell line A375 was analyzed using MTT and colony formation assays. Bufotalin significantly inhibited the proliferation and colony formation of A375 cells. Further studies demonstrated that bufotalin significantly upregulated the protein levels of ATM serine/threonine kinase and Chk2, downregulated CDC25C protein expression, and subsequently inhibited CDK1 expression, leading to cell cycle arrest at the G2/M phase of the A375 cells. Furthermore, bufotalin significantly increased BAX expression levels, decreased BCL­2 expression, and then upregulated apoptosis­related proteins, caspase­3/-9, followed by A375 cell apoptosis. Taken together, these results show that bufotalin induces cell cycle arrest at the G2/M phase and cell apoptosis, resulting in the inhibition of A375 cell proliferation, thereby suggesting that bufotalin may be utilized in melanoma treatment.


Assuntos
Apoptose/efeitos dos fármacos , Bufanolídeos/farmacologia , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Melanoma/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Bufanolídeos/química , Bufanolídeos/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Queratinócitos , Melanoma/patologia , Neoplasias Cutâneas/patologia , Melanoma Maligno Cutâneo
16.
Oxid Med Cell Longev ; 2019: 9817576, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30805086

RESUMO

The mitochondrial protein mitoNEET is a type of iron-sulfur protein localized to the outer membrane of mitochondria and is involved in a variety of human pathologies including cystic fibrosis, diabetes, muscle atrophy, and neurodegeneration. In the current study, we found that isoliquiritigenin (ISL), one of the components of the root of Glycyrrhiza glabra L., could decrease the expression of mitoNEET in A375 melanoma cells. We also demonstrated that mitoNEET could regulate the content of reactive oxygen species (ROS), by showing that the ISL-mediated increase in the cellular ROS content could be mitigated by the mitoNEET overexpression. We also confirmed the important role of ROS in ISL-treated A375 cells. The increased apoptosis rate and the decreased mitochondrial membrane potential were mitigated by the overexpression of mitoNEET in A375 cells. These findings indicated that ISL could decrease the expression of mitoNEET, which regulated ROS content and subsequently induced mitochondrial dysfunction and apoptosis in A375 cells. Our findings also highlight mitoNEET as a promising mitochondrial target for cancer therapy.


Assuntos
Apoptose/efeitos dos fármacos , Chalconas/farmacologia , Melanoma/metabolismo , Melanoma/patologia , Mitocôndrias/patologia , Proteínas Mitocondriais/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo
17.
Oncotarget ; 8(21): 34565-34575, 2017 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-28410220

RESUMO

Isoliquiritigenin (ISL), a member of the flavonoids, is known to have anti-tumor activity in vitro and in vivo. The effect of ISL on reprogramming in cancer cells, however, remains elusive. In this study, we investigated the effect of ISL on reprogramming in human melanoma A375 cells. ISL (15 µg/ml) significantly inhibited A375 cell proliferation, anchorage independent cell proliferation and G2/M cell cycle arrest after ISL exposure for 24 h. However, there were no significant changes in apoptosis rate. Terminal differentiation indicators (melanin content, melanogenesis mRNA expression, tyrosinase (TYR) activity) were all up-regulated by ISL treatment. In ISL-treated cells, glucose uptake, lactate levels and mRNA expression levels of GLUT1 and HK2 were significantly decreased, and accompanied by an increase in O2 consumption rate (OCR) and adenosine triphosphate (ATP) deficiency. Protein expression levels of mTORC2-AKT-GSK3ß signaling pathway components (mTOR, p-mTOR, RICTOR, p-AKT, p-GSK3ß) decreased significantly after ISL treatment. Co-treatment of ISL and the mTOR-specific inhibitor Ku-0063794 had a synergistic effect on the inhibition of proliferation, and increased melanin content and TYR activity. Glucose uptake and lactate levels decreased more significantly than treatment with ISL alone. These findings indicate that ISL induced reprogramming in A375 melanoma cells by activating mTORC2-AKT-GSK3ß signaling.


Assuntos
Chalconas/farmacologia , Inibidores Enzimáticos/farmacologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Melanoma/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , Humanos , Ácido Láctico/metabolismo , Melanoma/tratamento farmacológico , Morfolinas/farmacologia , Oxigênio/metabolismo , Pirimidinas/farmacologia , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA