Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Front Oncol ; 13: 1254026, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38023194

RESUMO

Background: Cholangiocarcinoma (CCA) is an aggressive disease with limited treatment options. Despite substantial efforts to explore better regimens, gemcitabine-based chemotherapy has been the standard first-line treatment for decades. With the growing field of precision medicine, biomarker-guided treatments are gaining popularity. MET alteration is a frequent occurrence in various cancer types, making it a promising target. Case presentation: A 53-year-old man visited our hospital with a complaint of upper abdominal pain. Advanced CCA was diagnosed based on the biopsy of the metastatic lymph nodes and immunohistochemistry. Next-generation sequencing revealed MET amplification. As the patient was intolerant to traditional chemotherapy, savolitinib (a c-MET inhibitor) was administered. Partial response was achieved, and the treatment was well tolerated. After 1 year, the patient developed progressive disease, to which the emergence of epidermal growth factor receptor amplification may have contributed. Conclusion: Our study verified the therapeutic value of a c-MET inhibitor in advanced CCA-harboring MET amplification and provides an alternative strategy for patients who are intolerant to chemotherapy.

3.
J Drug Target ; 31(10): 1013-1026, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37869884

RESUMO

Pancreatic cancer (PC) is a common malignant tumour in the digestive system. Due to the lack of sensitive diagnostic markers, strong metastasis ability, and resistance to anti-cancer drugs, the prognosis of PC is inferior. In the past decades, increasing evidence has indicated that the development of PC is closely related to various signalling pathways. With the exploration of RAS-driven, epidermal growth factor receptor, Hedgehog, NF-κB, TGF-ß, and NOTCH signalling pathways, breakthroughs have been made to explore the mechanism of pancreatic carcinogenesis, as well as the novel therapies. In this review, we discussed the signalling pathways involved in PC and summarised current targeted agents in the treatment of PC. Furthermore, opportunities and challenges in the exploration of potential therapies targeting signalling pathways were also highlighted.


Assuntos
Antineoplásicos , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/metabolismo , Transdução de Sinais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Prognóstico , Neoplasias Pancreáticas
4.
Phytomedicine ; 120: 155062, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37683586

RESUMO

BACKGROUND: Icaritin has a wide range of pharmacological activities, including significant an-titumor activity. However, the mechanism of action of icaritin in endometrial cancer (UCEC) remains unknown. FOX proteins are a highly conserved transcription factor superfamily that play important roles in epithelial cell differentiation, tumor metastasis, angiogenesis, and cell cycle regulation. FOXC1 is an important member of the FOX protein family. FOXC1 is aberrantly expressed in endometrial cancer and may play a role in the migration and invasion of endometrial cancer; however, its mechanism of action has not yet been reported. O-GlcNAc glycosylation is a common post-translational modification. In endometrial cancer, high levels of O-GlcNAcylation promote cell proliferation, migration, and invasion. Cancer development is often accompanied by O-GlcNAc modification of proteins; however, O-GlcNAc modification of the transcription factor FOXC1 has not been reported to date. PURPOSE: To investigate the inhibitory effects of icaritin on RL95-2 and Ishikawa endometrial cancer cells in vitro and in vivo and to elucidate the possible molecular mechanisms. METHODS/STUDY DESIGN: CCK8, colony formation, migration, and invasion assays were used to determine the inhibitory effects of icaritin on endometrial cancer cells in vitro. Cell cycle regulation was assayed by flow cytometry. Protein levels were measured based on western blotting. The level of FOXC1 expression in endometrial cancer tissues was determined by immunohistochemistry. To assess whether icaritin also has activity in vivo, its effect on tumor xenografts was evaluated. RESULTS: Immunohistochemical analysis of clinical samples revealed that FOXC1 expression was significantly higher in endometrial cancer tissues than in normal tissues. Downregulation of FOXC1 inhibited the proliferative, colony formation, migration, and invasive abilities of RL95-2 and Ishikawa endometrial cancer cells. Icaritin inhibited the proliferation, colony formation, migration, and invasion of endometrial cancer cells and blocked the cell cycle in S phase. Icaritin affected O-GlcNAc modification of FOXC1 and thus the stability of FOXC1, which subsequently triggered the inhibition of endometrial cancer cell proliferation. CONCLUSION: The anti-endometrial cancer effect of icaritin is related to the inhibition of abnormal O-GlcNAc modification of FOXC1, which may provide an important theoretical foundation for the use of icaritin against endometrial cancer.


Assuntos
Neoplasias do Endométrio , Humanos , Feminino , Neoplasias do Endométrio/tratamento farmacológico , Flavonoides/farmacologia , Divisão Celular , Proliferação de Células , Fatores de Transcrição Forkhead
5.
Front Chem ; 10: 919624, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35692694

RESUMO

ß-Glucosidase (ß-Glc) is an enzyme capable of the selective hydrolysis of the ß-glycosidic bond of glycosides and glycans containing glucose. ß-Glc expressed by intestinal microbiota has attracted increasing levels of interest, due to their important roles for the metabolism of exogenous substances in the gut. Using the 2-((6-hydroxy-2,3-dihydro-1H-xanthen-4-yl)methylene)malononitrile fluorophore (DXM-OH, λem 636 nm) and the recognition group ß-Glucose, an enzymatic activatable turn-on fluorescent probe (DXM-Glc) was developed for the selective and sensitive sensing of ß-Glc. In addition, DXM-Glc could be used to sense endogenous ß-Glc in living fungal cells. Using DXM-Glc, Pichia terricola M2 was identified as a functional intestinal fungus with ß-Glc expression. P. terricola M2 could transform the flavone glycoside Icariin to Icariside Ⅱ efficiently, which confirmed the metabolism of glycosides in the gut mediated by fungi. Furthermore, Icariside Ⅱ could inhibit the proliferation of human endometrial cancer cells (RL 95-2 and ishikawa) significantly, suggesting the metabolic activation of Icariin by intestinal fungi in vivo. Therefore, DXM-Glc as a probe for ß-Glc provided a novel technique for the investigation of the metabolism of bioactive substances by intestinal microbiota.

6.
J Food Biochem ; 44(12): e13502, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33025647

RESUMO

Bovine α-lactalbumin (α-La)/ß-lactoglobulin (ß-Lg) was pretreated through ultrasonic treatment and subsequently binding with oleic acid (OA) by heat treatment. And, the antitumor activity, IgE/IgG-binding ability, and structural modifications were investigated. After α-La/ß-Lg were treated by ultrasonic prior to binding with OA, the treated α-La/ß-Lg showed high antitumor activity and IgE/IgG-binding ability, and significantly affected the structural modifications, which reflected by the reduction in α-helix content, the increase of molecular weight, intrinsic fluorescence intensity, and surface hydrophobicity. Molecular docking studies indicated that OA bound to α-La/ß-Lg by hydrogen bonds and hydrophobic interaction. Therefore, ultrasonic prior to binding with OA could improve antitumor activity and IgE/IgG-binding ability of α-La/ß-Lg as a result of structural modifications. And, ultrasonic prior to binding with fatty acid processing of milk products alone may increase the antitumor activity, this change may enhance the risk of an allergenic reaction in milk allergy patients to some extent. PRACTICAL APPLICATIONS: Fatty acids, natural ligands associated with the bovine milk proteins, and milk protein-fatty acid complex has a variety of functional applications in the food industry. This study revealed that antitumor activity, IgE/IgG-binding ability, and structural modifications of α-La/ß-Lg induced by ultrasonic prior to binding with oleic acid. It will be beneficial to understand the mechanism of the functional changes of protein. Ultrasonic prior to binding with oleic acid will be more likely to develop a practical technology to improve the functional characteristics of milk protein and design the optimal nutritional performance of milk food.


Assuntos
Lactalbumina , Lactoglobulinas , Animais , Bovinos , Humanos , Imunoglobulina E , Imunoglobulina G , Simulação de Acoplamento Molecular , Ácido Oleico
7.
Arch Biochem Biophys ; 692: 108531, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32745464

RESUMO

Adipose-derived stem cell (ADSC) therapy is a promising treatment strategy for wound healing; however, the mechanism underlying this effect remains unclear. In the present study, we aimed to explore the influence of ADSC-derived VEGF on diabetic wounds and its role in modulating endothelial progenitor cells. The effect of ADSCs and ADSC-derived VEGF in vivo was investigated using a diabetic wound healing model, and inflammatory factors, such as IL-6, IL-10, and TNF-α, were detected. RT-qPCR and western blot analysis were used to detect the expression of downstream targets. In addition, the role of ADSC-derived VEGF in modulating endothelial progenitor cells (EPCs) was investigated using EdU assay, CD-31 immunofluorescence, and Transwell assay in vitro. The results show that ADSCs accelerated diabetic wound tissue closure and decreased the expression of inflammatory factors, such as IL-6, IL-10, and TNF-α. Further molecular mechanism studies indicated that coculturing EPCs with ADSC--conditioned medium enhanced the proliferation, mobilization and differentiation of EPCs into endothelial cells. This enhancement was inhibited when the expression of the VEGF downstream signal molecules VEGFR2, PLCγ, and ERK1/ERK2 was blocked, indicating that ADSCs might accelerate diabetic wound healing through the recruitment and differentiation of EPCs mediated by VEGF. Overall, the results of the study revealed that ADSCs could promote diabetic wound healing through the recruitment and differentiation of EPCs via angiogenesis effects regulated by the VEGF-PLCγ-ERK1/ERK2 pathway and suppression of the inflammatory response. In addition, it will be helpful to establish further understanding of ADSC therapy for clinical application.


Assuntos
Tecido Adiposo/metabolismo , Diabetes Mellitus Experimental , Angiopatias Diabéticas , Sistema de Sinalização das MAP Quinases , Fosfolipase C gama/metabolismo , Transplante de Células-Tronco , Células-Tronco/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Cicatrização , Tecido Adiposo/patologia , Aloenxertos , Animais , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/terapia , Angiopatias Diabéticas/metabolismo , Angiopatias Diabéticas/patologia , Angiopatias Diabéticas/terapia , Ratos , Ratos Sprague-Dawley , Células-Tronco/patologia
8.
J Comput Biol ; 27(5): 769-778, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31502860

RESUMO

We aimed to identify differentially expressed genes (DEGs) in epidermal stem cells (epiSCs) in response to high fat diet (HFD). DEGs were identified by time-series analysis of the gene expression profile (GSE84510) in Gene Expression Omnibus (GEO) database. Functions and pathways affected by HFD were identified by functional annotation of DEGs. Key factors responding to HFD was identified by protein-protein interaction (PPI) network analysis. Two groups of genes with the same tendency in response to HFD were identified. ECM-related processes and PI3K pathway were altered in the early stage of obesity. A PPI network was constructed to delineate the interactions among proteins encoded by DEGs and ICAM1 and RELA were key epiSC factors respond to HFD. Our studies may provide valuable insights into the molecular mechanisms underlying how obesity affects the functions of epiSC.


Assuntos
Molécula 1 de Adesão Intercelular/genética , Obesidade/genética , Mapas de Interação de Proteínas/genética , Células-Tronco/metabolismo , Fator de Transcrição RelA/genética , Animais , Biologia Computacional , Dieta Hiperlipídica/efeitos adversos , Células Epidérmicas/metabolismo , Regulação da Expressão Gênica/genética , Ontologia Genética , Redes Reguladoras de Genes/genética , Humanos , Camundongos , Obesidade/patologia , Transcriptoma/genética
9.
Exp Ther Med ; 17(5): 4139-4143, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30988790

RESUMO

This study investigated optimal pathways for preeclampsia (PE) utilizing the network-based guilt by association (GBA) algorithm. The inference method consisted of four steps: preparing differentially expressed genes (DEGs) between PE patients and normal controls from gene expression data; constructing co-expression network (CEN) for DEGs utilizing Spearman's correlation coefficient (SCC) method; and predicting optimal pathways by network-based GBA algorithm of which the area under the receiver operating characteristics curve (AUROC) was gained for each pathway. There were 351 DEGs and 61,425 edges in the CEN for PE. Subsequently, 53 pathways were obtained with a good classification performance (AUROC >0.5). AUROC for 9 was >0.9 and defined as optimal pathways, especially microRNAs in cancer (AUROC=0.9966), gap junction (AUROC=0.9922), and pathogenic Escherichia coli infection (AUROC=0.9888). Nine optimal pathways were identified through comprehensive analysis of data from PE patients, which might shed new light on uncovering molecular and pathological mechanism of PE.

10.
Biomed Pharmacother ; 108: 119-129, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30218856

RESUMO

Colon cancer (CC), one of the most frequently diagnosed malignancies deriving from the digestive system, has greatly threatened human health and life. Fatty acid binding protein 7 (FABP7), an intracellular protein with the tissue-specific expression pattern, has been reported to be implicated in diverse types of human tumors. However, the biological role of FABP7 in CC is still poorly understood. The current study aimed to investigate the role of FABP7 in CC and illuminate the potential molecular mechanisms. In this present study, we found that FABP7 was highly expressed in CC tissues and cell lines, suggesting the possible involvement of FABP7 in CC tumorigenesis. Moreover, functional investigations showed that FABP7-overexpression promoted CC cell proliferation, colony formation, cell cycle progression and inhibited cell apoptosis; on the contrary, FABP7 knockdown produced an inhibitory effects on CC cell proliferation and survival. Notably, FABP7 knockdown inhibited colon tumor growth in vivo. In addition, mechanistic investigations demonstrated that FABP7 exerted its promoting effects on CC cell proliferation and survival through activation of the MEK/ERK signaling pathway. Collectively, our data indicate that FABP7 may be used as a novel diagnostic bio-marker and a potential therapeutic target for CC.


Assuntos
Neoplasias do Colo/enzimologia , Neoplasias do Colo/patologia , Proteína 7 de Ligação a Ácidos Graxos/metabolismo , Sistema de Sinalização das MAP Quinases , Animais , Apoptose/genética , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Sobrevivência Celular/genética , Neoplasias do Colo/genética , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ensaio Tumoral de Célula-Tronco , Regulação para Cima/genética
11.
Oncol Rep ; 40(4): 2215-2223, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30015962

RESUMO

Metadherin (MTDH) is a multifunctional oncogene involved in tumor cell migration and metastasis through regulating a number of oncogenic signaling pathways in various human malignancies. Previous studies have demonstrated that MTDH is overexpressed in human colorectal cancer (CRC) and associated with cancer progression and a poor prognosis. However, the underlying mechanisms remain largely unknown. The present study investigated the expression and role of MTDH in CRC cells as well as the underlying mechanism of this. Western blot analysis and quantitative polymerase chain reaction were conducted to determine protein and mRNA expression of MTDH in three human CRC cell lines. A short hairpin RNA (shRNA) targeting MTDH was introduced into CRC HCT116 cells to stably inhibit MTDH expression. A Cell Counting Kit­8 assay, colony formation assay, Transwell assay and flow cytometry were used to investigate the effect of MTDH­knockdown on cell proliferation, migration, apoptosis and cell cycle arrest. Western blotting was performed to examine the protein expression levels of cell growth­ and apoptosis­associated genes. The results demonstrated that MTDH was commonly expressed in CRC cell lines. MTDH silencing significantly suppressed cell growth, colony forming ability and migration while inducing the apoptosis of HCT116 cells. In addition, MTDH depletion induced S phase cell cycle arrest in HCT116 cells. Mechanistically, knockdown of MTDH markedly downregulated the expression of phosphorylated protein kinase B, c­Myc, proliferating cell nuclear antigen and B­cell lymphoma 2 (Bcl­2) protein in HCT116 cells, and the expression of p53 and Bcl­2­associated X protein was significantly increased compared with the negative control shRNA group (P<0.05), suggesting that MTDH may function through the expression of numerous types of apoptosis­associated and signaling channel proteins in CRC cells. Taken together, these data indicated that MTDH may serve as a biomarker and candidate therapeutic target for CRC.


Assuntos
Biomarcadores Tumorais/metabolismo , Moléculas de Adesão Celular/metabolismo , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Apoptose , Biomarcadores Tumorais/genética , Moléculas de Adesão Celular/antagonistas & inibidores , Moléculas de Adesão Celular/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas de Membrana , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-myc/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA , Transdução de Sinais , Células Tumorais Cultivadas
12.
Biomed Pharmacother ; 105: 599-605, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29890468

RESUMO

Acute pancreatitis (AP) is a common acute abdominal disease accompanied by systemic inflammatory response syndrome, and could even be complicated by multiple-organ damage. This study aimed to examine whether calycosin, an isoflavone isolated from Radix astragali with antioxidant and anti-inflammatory activity, could protect against AP induced by cerulein. To this end, Balb/C mice were injected with cerulein (50 µg/kg) to establish the animal model of AP. Calycosin (25 and 50 mg/kg, p.o.) was administered 1 h prior to the first cerulein injection. After the last injection of cerulein, the mice were sacrificed and blood was obtained for cytokine analysis. The pancreas was removed for morphological examination, myeloperoxidase (MPO) and malondialdehyde (MDA) analyses, immunohistochemistry, and western blot analysis. Calycosin treatment reversed the increased serum levels of amylase and lipase, alleviated the pathological damage in the pancreas, and decreased the levels of tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1ß in mice with AP. Additionally, calycosin significantly reduced cerulein-induced pancreatic edema, inhibited MPO activity and increased superoxide dismutase (SOD) activity, and inhibited the expression of NF-κB/p65 and phosphorylation of the inhibitor of NF-κB (IκBα) and p38 MAPK. These results suggested that calycosin protects against AP by exerting anti-inflammatory and anti-oxidative stress effects via the p38 MAPK and NF-κB signal pathways. Calycosin's benefits for AP patients need to be explored further.


Assuntos
Anti-Inflamatórios/uso terapêutico , Isoflavonas/uso terapêutico , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Pancreatite/tratamento farmacológico , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Doença Aguda , Animais , Anti-Inflamatórios/isolamento & purificação , Astragalus propinquus , Ceruletídeo , Citocinas/imunologia , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/química , Isoflavonas/isolamento & purificação , Masculino , Camundongos Endogâmicos BALB C , Pancreatite/induzido quimicamente , Pancreatite/imunologia , Transdução de Sinais
13.
Biomed Pharmacother ; 104: 204-210, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29772441

RESUMO

This work aims to study the roles and related mechanisms of six2 in 5-FU sensitivity of hepatocellular carcinoma (HCC) cells. KM-Plotter analysis showed that HCC patients with higher six2 expression levels had shorter overall survival. Six2 expression was higher in clinical HCC tissues than in normal tissues, and was negatively correlated with E-cadherin expression. Additionally, six2 overexpression decreased the sensitivity of HCC cells to 5-Fu, characterized as attenuating 5-FU-induced cell apoptosis and downregulation of cell viability, and promoted HCC cells stemness. Mechanistically, six2 overexpression repressed E-cadherin expression via stimulating promoter methylation of the E-cadherin. And E-cadherin overexpression rescued six2-induced decrease of 5-FU sensitivity and promotion on HCC cells stemness. Therefore, our results suggest that Six2 is negatively correlated with good prognosis and decreases 5-FU sensitivity via suppressing E-cadherin expression in HCC cells.


Assuntos
Caderinas/metabolismo , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/metabolismo , Fluoruracila/farmacologia , Proteínas de Homeodomínio/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Antígenos CD , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células Hep G2 , Humanos , Neoplasias Hepáticas/patologia , Prognóstico , Regiões Promotoras Genéticas/efeitos dos fármacos
14.
RSC Adv ; 9(1): 151-165, 2018 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-35521597

RESUMO

Background: EMT has a crucial effect on the progression and metastasis of tumors. This work will elucidate the role of miR-425 in EMT and the development of TNBC. Methods: The differential miRNA expression among non-tumor, para-tumor (adjacent tissue of tumor) and tumor tissues was analyzed. The luciferase activities of TGF-ß1 3'UTR treated with miR-425 were determined. Then human breast cancer cell lines were treated with mimics or inhibitors of miR-425, and then the cell proliferation and migration, and invasion ability were assessed. The expression of TGF-ß1 and markers of epithelial cells and mesenchymal cells were analyzed. The influences of miR-425 on the development of TNBC through inducing EMT by targeting the TGF-ß1/SMAD3 signaling pathway in TNBC cell lines were investigated. Furthermore, xenograft mice were used to explore the potential roles of miR-425 on EMT and the development of TNBC in vivo. Results: Compared with non-tumor tissues, 9 miRNAs were upregulated and 3 miRNAs were down-regulated in tumor tissues. The relative expression of miR-425 in tumor tissues was obviously much lower than that in para-tumor and non-tumor tissues. MiR-425 suppressed TGF-ß1 expression, and further inhibited expression of mesenchymal cell markers, while it exerted effects on cell proliferation and migration of TNBC cell lines. Moreover, the agomir of miR-425 could protect against the development process in a murine TNBC xenograft model. Conclusions: Our results demonstrated that miR-425 targets TGF-ß1, and was a crucial suppressor on EMT and the development of TNBC through inhibiting the TGF-ß1/SMAD3 signaling pathway. This suggests that aiming at the TGF-ß1/SMAD3 signaling pathway by enhancing relative miR-425 expression, is a feasible therapy strategy for TNBC.

16.
Cancer Sci ; 108(3): 469-477, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28004478

RESUMO

Although inhibitors targeting tumor angiogenic pathway have provided improvement for clinical treatment in patients with various solid tumors, the still very limited anti-cancer efficacy and acquired drug resistance demand new agents that may offer better clinical benefits. In the effort to find a small molecule potentially targeting several key pathways for tumor development, we designed, discovered and evaluated a novel multi-kinase inhibitor, CS2164. CS2164 inhibited the angiogenesis-related kinases (VEGFR2, VEGFR1, VEGFR3, PDGFRα and c-Kit), mitosis-related kinase Aurora B and chronic inflammation-related kinase CSF-1R in a high potency manner with the IC50 at a single-digit nanomolar range. Consequently, CS2164 displayed anti-angiogenic activities through suppression of VEGFR/PDGFR phosphorylation, inhibition of ligand-dependent cell proliferation and capillary tube formation, and prevention of vasculature formation in tumor tissues. CS2164 also showed induction of G2/M cell cycle arrest and suppression of cell proliferation in tumor tissues through the inhibition of Aurora B-mediated H3 phosphorylation. Furthermore, CS2164 demonstrated the inhibitory effect on CSF-1R phosphorylation that led to the suppression of ligand-stimulated monocyte-to-macrophage differentiation and reduced CSF-1R+ cells in tumor tissues. The in vivo animal efficacy studies revealed that CS2164 induced remarkable regression or complete inhibition of tumor growth at well-tolerated oral doses in several human tumor xenograft models. Collectively, these results indicate that CS2164 is a highly selective multi-kinase inhibitor with potent anti-tumor activities against tumor angiogenesis, mitosis and chronic inflammation, which may provide the rationale for further clinical assessment of CS2164 as a therapeutic agent in the treatment of cancer.


Assuntos
Adenocarcinoma/tratamento farmacológico , Inibidores da Angiogênese/uso terapêutico , Neoplasias do Colo/tratamento farmacológico , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Mitose/efeitos dos fármacos , Neovascularização Patológica/tratamento farmacológico , Fenilenodiaminas/uso terapêutico , Quinolinas/uso terapêutico , Células 3T3 , Animais , Aurora Quinase B/antagonistas & inibidores , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Histonas/metabolismo , Humanos , Inflamação/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Simulação de Acoplamento Molecular , Naftalenos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-kit/antagonistas & inibidores , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Mol Neurobiol ; 53(3): 1648-1653, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25682969

RESUMO

Transforming growth factor beta (TGF-ß) is suggestive of a molecular target for cancer therapy due to its involvement in cell cycle, differentiation, and morphogenesis. Meanwhile, survivin is identified as an apoptosis inhibitor and involved in tumorgenesis. Here, we aimed to investigate the potential associations between TGF-ß and survivin in glioblastoma U87 cell line. Survivin small interfering RNA (siRNA), Western blotting, and cell cycle analysis were introduced to detect relevant proteins in TGF-ß pathways. In this study, we observed a concentration- and time-dependent increase of survivin expression after treatment with TGF-ß1. However, the kinase inhibitors U0126 and LY294002 inhibited the upregulation of survivin in comparison with DMSO. In addition, survivin siRNA effectively abrogated survivin expression in U87 cells, therefore affected cells' entry into the S phase of cell cycle, and then repressed the expression of epidermal growth factor receptor (EGFR) and matrix metalloproteinase 9 (MMP9) in comparison with non-transfection. In conclusion, the present study shows that TGF-ß upregulates survivin expression via ERK and PI3K/AKT pathway, leading to glioblastoma cell cycle progression. Thus, the blockade of survivin will allow for the treatment of glioblastoma, partially attributing to the inhibition of EGFR and MMP9 expression.


Assuntos
Ciclo Celular/efeitos dos fármacos , Receptores ErbB/metabolismo , Glioblastoma/enzimologia , Glioblastoma/patologia , Proteínas Inibidoras de Apoptose/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Survivina , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos
18.
Exp Ther Med ; 6(3): 727-730, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24137255

RESUMO

The aim of this study was to investigate the application of adenosine stress echocardiography (ASE) in the prognosis of acute myocardial infarction (AMI) following percutaneous coronary intervention (PCI). A total of 79 patients with AMI who underwent PCI were selected for the study. ASE testing was performed within one week following the PCI. Subsequent to the ASE, the patients with ≥5% increases in the left ventricular ejection fraction (LVEF) levels were included in the improved LVEF group, while patients with <5% increases in LVEF levels were included in the unimproved LVEF group. A follow-up study was performed during the 24 months subsequent to the ASE. The incidence of major adverse cardiovascular events (MACEs) was observed and compared between the two groups and logistic regression analysis was applied to identify the risk factors for clinical prognosis. There were no significant differences in Killip classification, LVEF, left ventricular end-diastolic diameter or blood plasma B-type natriuretic peptide concentration between the two groups following PCI. The incidence of MACEs in the improved LVEF group was significantly lower than that in unimproved LVEF group (14.29 versus 43.24%, respectively; P<0.05). Logistic regression analysis identified LVEF increases of <5% and segment improvements of ≤3 as the risk factors for the clinical prognosis of AMI following treatment with PCI. Therefore, ASE is an effective method of assessing the clinical effect of PCI treatment, which may be utilized to predict the incidence of MACEs following PCI.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA