Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Clin Transl Oncol ; 26(9): 2250-2261, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38554191

RESUMO

BACKGROUND: The objective of this research was to investigate how the combination of semen coicis extract and PD-1 inhibitors can potentially work together to enhance the anti-tumor effects, with a focus on understanding the underlying mechanism. METHODS: We obtained the active components and specific targets of semen coicis in the treatment of NSCLC from various databases, namely TCMSP, GeneCard, and OMIM. By utilizing the STRING database and Cytoscape software, we established a protein interaction network (PPI) for the active ingredient of semen coicis and the target genes related to NSCLC. To explore the potential pathways involved, we conducted gene ontology (GO) and biological pathway (KEGG) enrichment analyses, which were further supported by molecular docking technology. Additionally, we conducted cyto-inhibition experiments to verify the inhibitory effects of semen coicis alone or in combination with a PD-1 inhibitor on A549 cells, along with examining the associated pathways. Furthermore, we investigated the synergistic mechanism of these two drugs through cytokine release experiments and the PD-L1 expression study on A549 cells. RESULTS: Semen coicis contains two main active components, Omaine and (S)-4-Nonanolide. Its primary targets include PIK3R1, PIK3CD, PIK3CA, AKT2, and mTOR. Molecular docking experiments confirmed that these ingredients and targets form stable bonds. In vitro experiments showed that semen coicis demonstrates inhibitory effects against A549 cells, and this effect was further enhanced when combined with PD-1 inhibitors. PCR and WB analysis confirmed that the inhibition of the PI3K-AKT-mTOR pathway may contribute to this effect. Additionally, semen coicis was observed to decrease the levels of IFN-γ, IL-6, and TNF-α, promoting the recovery of the human anti-tumor immune response. And semen coicis could inhibit the induced expression of PD­L1 of A549 cells stimulated by IFN­Î³ as well. CONCLUSION: Semen coicis not only has the ability to kill tumor cells directly but also alleviates the immunosuppression found in the tumor microenvironment. Additionally, it collaboratively enhances the effectiveness of PD-1 inhibitors against tumors by blocking the activation of PI3K-AKT-mTOR.


Assuntos
Antineoplásicos , Coix , Neoplasias Pulmonares , Receptor de Morte Celular Programada 1 , Transdução de Sinais , Humanos , Células A549 , Antígeno B7-H1/metabolismo , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Sinergismo Farmacológico , Inibidores de Checkpoint Imunológico/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Simulação de Acoplamento Molecular , Fosfatidilinositol 3-Quinases/metabolismo , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/metabolismo , Mapas de Interação de Proteínas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Coix/química , Antineoplásicos/farmacologia
2.
Acta Pharmacol Sin ; 45(6): 1287-1304, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38360930

RESUMO

HER2-positive (HER2+) metastatic breast cancer (mBC) is highly aggressive and a major threat to human health. Despite the significant improvement in patients' prognosis given the drug development efforts during the past several decades, many clinical questions still remain to be addressed such as efficacy when combining different therapeutic modalities, best treatment sequences, interindividual variability as well as resistance and potential coping strategies. To better answer these questions, we developed a mechanistic quantitative systems pharmacology model of the pathophysiology of HER2+ mBC that was extensively calibrated and validated against multiscale data to quantitatively predict and characterize the signal transduction and preclinical tumor growth kinetics under different therapeutic interventions. Focusing on the second-line treatment for HER2+ mBC, e.g., antibody-drug conjugates (ADC), small molecule inhibitors/TKI and chemotherapy, the model accurately predicted the efficacy of various drug combinations and dosing regimens at the in vitro and in vivo levels. Sensitivity analyses and subsequent heterogeneous phenotype simulations revealed important insights into the design of new drug combinations to effectively overcome various resistance scenarios in HER2+ mBC treatments. In addition, the model predicted a better efficacy of the new TKI plus ADC combination which can potentially reduce drug dosage and toxicity, while it also shed light on the optimal treatment ordering of ADC versus TKI plus capecitabine regimens, and these findings were validated by new in vivo experiments. Our model is the first that mechanistically integrates multiple key drug modalities in HER2+ mBC research and it can serve as a high-throughput computational platform to guide future model-informed drug development and clinical translation.


Assuntos
Neoplasias da Mama , Receptor ErbB-2 , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Neoplasias da Mama/metabolismo , Humanos , Feminino , Receptor ErbB-2/metabolismo , Receptor ErbB-2/antagonistas & inibidores , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Inibidores de Proteínas Quinases/farmacologia , Imunoconjugados/uso terapêutico , Imunoconjugados/farmacologia , Farmacologia em Rede , Modelos Biológicos , Antineoplásicos/uso terapêutico , Antineoplásicos/administração & dosagem , Camundongos , Linhagem Celular Tumoral , Metástase Neoplásica
3.
Mol Med Rep ; 23(1)2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33179089

RESUMO

In recent years, obesity has become a major public health concern. Obesity has been previously associated with low­grade inflammation and TNF­α induction in adipose tissue, which subsequently disrupts adipocyte metabolism. MicroRNAs (miRNAs/miRs) are important metabolic factors and their dysregulation has been associated with obesity­related metabolic syndromes. In fact, it has been directly suggested that miR­424 may be functionally associated with adipogenesis, although its exact role in this process remains unclear. The present study aimed to identify the function of miR­424 in adipogenesis. In the present study, miR­424 expression levels were analyzed during adipogenesis and the differential expression of this miRNA in the adipose tissue of obese and non­obese children was also assessed. Furthermore, the interaction between miR­424 and the adipocytokine TNF­α was determined. Finally, miR­424 target genes and downstream signaling pathways were predicted via bioinformatics and analyzed by performing a luciferase reporter assay to elucidate the functional mechanisms of miR­424 in adipogenesis of visceral adipocytes. The results revealed that the expression levels of miR­424 upregulated in the adipose tissue biopsies from obese children compared with the biopsies of non­obese children. However, in cultured adipocytes, the expression levels of miR­424 were discovered to be gradually downregulated during the adipogenesis process. TNF­α treatment significantly downregulated the expression levels of miR­424 via binding to its promoter region and reducing its transcriptional activity. Through bioinformatic prediction analysis, miR­424 target genes were analyzed, of which several were identified to be involved in signaling pathways that are known to regulate adipogenesis, such as the Wnt signaling pathway. In conclusion, the present study indicated that miR­424 was regulated by TNF­α and served an important role in adipogenesis.


Assuntos
Adipócitos/citologia , MicroRNAs/genética , Obesidade/genética , Fator de Necrose Tumoral alfa/metabolismo , Adipócitos/química , Adipogenia , Estudos de Casos e Controles , Diferenciação Celular , Células Cultivadas , Criança , Humanos , Masculino , Obesidade/metabolismo , Regiões Promotoras Genéticas , Regulação para Cima , Via de Sinalização Wnt
4.
Med Sci Monit ; 24: 2983-2991, 2018 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-29735973

RESUMO

BACKGROUND Baicalein, one of the major flavonoids in the plant [i]Scutellaria baicalensis[/i], can regulate the invasive ability of cancer cells. The invasion of trophoblasts is similar to the invasion of tumor cells into host tissues. The appropriate invasion of trophoblast cells into the endometrium is an important factor for successful embryo implantation. In this research, we investigated the effect of baicalein on the invasion and migration of trophoblast cells and its possible molecular mechanism. MATERIAL AND METHODS We treated HTR-8/SVneo cells with different concentrations (0, 0.05, 0.1, and 0.5 µM) of baicalein. The invasion and migration abilities of HTR-8/SVneo cells were studied. Protein levels and gene expression related to invasion and migration were analyzed by Western blot analysis and reverse transcription-quantitative polymerase chain reaction, respectively. RESULTS Baicalein enhanced the migration and invasion of HTR-8/SVneo cells. In addition, gene expression and protein levels of MMP-9 in HTR-8/SVneo cells changed in the presence of baicalein. Moreover, the data show that baicalein activated the NF-κB pathway. Baicalein was also able to rescue effects of an NF-κB-specific inhibitor (JSH-23) on the migration and invasion of HTR-8/SVneo cells. CONCLUSIONS In conclusion, our results indicate that baicalein enhances migration and invasion of HTR-8/SVneo cells, which is important for successful pregnancy.


Assuntos
Movimento Celular/efeitos dos fármacos , Flavanonas/farmacologia , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Trofoblastos/citologia , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Fenilenodiaminas/farmacologia , Transporte Proteico/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo , Trofoblastos/efeitos dos fármacos , Trofoblastos/enzimologia
5.
Mol Med Rep ; 16(5): 5965-5971, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28901460

RESUMO

Obesity is a confirmed risk factor for hyperlipidemia, type­II diabetes, hypertension, and cardiovascular disease. MicroRNAs (miRs) have emerged as an important field of study within energy metabolism and obesity. A previous study demonstrated miR­1275 to be markedly down­regulated during maturation of human preadipocytes. It has been reported that miR­1275 dysregulates expression in several types of cancer and infections. Little is currently known about the regulation of miR­1275 transcription. The aim of the current study was to explore the mechanism underlying the expression of miR­1275 in mature human adipocytes. After differentiation, human adipocytes were incubated with tumor necrosis factor (TNF)­α and interleukin­6. The results of reverse transcription­quantitative polymerase chain reaction demonstrated that miR­1275 can be down­regulated by TNF­α and IL­6, in human mature adipocytes. Bioinformatic analysis was used to predict nuclear factor (NF)­κB binding sites of miR­1275's promoter region. Luciferase assay and rescue experiments were performed in HEK293T cells. NF­κB was involved in regulating miR­1275 transcription by binding to its promoter. In response to TNF­α, NF­κB was bound to the promoter of miR­1275 and inhibited its transcription. These results indicated that inflammatory factors could regulate miR­1275 transcription through NF­κB and influencing miR­1275 effects on obesity.


Assuntos
Adipócitos Brancos/efeitos dos fármacos , Interleucina-6/farmacologia , MicroRNAs/genética , NF-kappa B/genética , Transcrição Gênica , Fator de Necrose Tumoral alfa/farmacologia , Adipócitos Brancos/citologia , Adipócitos Brancos/metabolismo , Sequência de Bases , Sítios de Ligação , Diferenciação Celular , Linhagem Celular , Regulação da Expressão Gênica , Genes Reporter , Células HEK293 , Humanos , Gordura Intra-Abdominal/citologia , Gordura Intra-Abdominal/efeitos dos fármacos , Gordura Intra-Abdominal/metabolismo , Luciferases/genética , Luciferases/metabolismo , MicroRNAs/metabolismo , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , Transdução de Sinais
6.
Cell Biochem Biophys ; 66(3): 489-97, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23274913

RESUMO

NYGGF4, also known as phosphotyrosine interaction domain containing 1(PID1), is a recently discovered gene which is involved in obesity-related insulin resistance (IR) and mitochondrial dysfunction. We aimed to further elucidate the effects and mechanisms underlying NYGGF4-induced IR by investigating the effect of overexpressing mitochondrial transcription factor A (TFAM), which is essential for mitochondrial DNA transcription and replication, on NYGGF4-induced IR and mitochondrial abnormalities in 3T3-L1 adipocytes. Overexpression of TFAM increased the mitochondrial copy number and ATP content in both control 3T3-L1 adipocytes and NYGGF4-overexpressing adipocytes. Reactive oxygen species (ROS) production was enhanced in NYGGF4-overexpressing adipocytes and reduced in TFAM-overexpressing adipocytes; co-overexpression of TFAM significantly attenuated ROS production in NYGGF4-overexpressing adipocytes. However, overexpression of TFAM did not affect the mitochondrial transmembrane potential (ΔΨm) in control 3T3-L1 adipocytes or NYGGF4-overexpressing adipocytes. In addition, co-overexpression of TFAM-enhanced insulin-stimulated glucose uptake by increasing Glucose transporter type 4 (GLUT4) translocation to the PM in NYGGF4-overexpressing adipocytes. Overexpression of NYGGF4 significantly inhibited tyrosine phosphorylation of Insulin receptor substrate 1 (IRS-1) and serine phosphorylation of Akt, whereas overexpression of TFAM strongly induced phosphorylation of IRS-1 and Akt in NYGGF4-overexpressing adipocytes. This study demonstrates that NYGGF4 plays a role in IR by impairing mitochondrial function, and that overexpression of TFAM can restore mitochondrial function to normal levels in NYGGF4-overexpressing adipocytes via activation of the IRS-1/PI3K/Akt signaling pathway.


Assuntos
Adipócitos/metabolismo , Adipócitos/patologia , Proteínas de Transporte/genética , Proteínas de Ligação a DNA/genética , Resistência à Insulina , Mitocôndrias/patologia , Proteínas Mitocondriais/genética , Fatores de Transcrição/genética , Células 3T3-L1 , Trifosfato de Adenosina/metabolismo , Adipócitos/efeitos dos fármacos , Animais , Proteínas de Transporte/metabolismo , Proteínas de Ligação a DNA/metabolismo , Expressão Gênica , Glucose/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Insulina/metabolismo , Insulina/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Tamanho Mitocondrial/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA