Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biomedicines ; 11(12)2023 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-38137388

RESUMEN

Glioblastoma (GBM) is a representative malignant brain tumor characterized by a dismal prognosis, with survival rates of less than 2 years and high recurrence rates. Despite surgical resection and several alternative treatments, GBM remains a refractory disease due to its aggressive invasiveness and resistance to anticancer therapy. In this report, we explore the role of fibronectin type III domain containing 3B (FNDC3B) and its potential as a prognostic and therapeutic biomarker in GBM. GBM exhibited a significantly higher cancer-to-normal ratio compared to other organs, and patients with high FNDC3B expression had a poor prognosis (p < 0.01). In vitro studies revealed that silencing FNDC3B significantly reduced the expression of Survivin, an apoptosis inhibitor, and also reduced cell migration, invasion, extracellular matrix adhesion ability, and stem cell properties in GBM cells. Furthermore, we identified that FNDC3B regulates PTEN/PI3K/Akt signaling in GBM cells using MetaCore integrated pathway bioinformatics analysis and a proteome profiler phospho-kinase array with sequential western blot analysis. Collectively, our findings suggest FNDC3B as a potential biomarker for predicting GBM patient survival and for the development of treatment strategies for GBM.

2.
Artículo en Inglés | MEDLINE | ID: mdl-37859347

RESUMEN

Objective: Isoflurane, a widely used common inhalational anesthetic agent, can induce brain toxicity. The challenge lies in protecting neurologically compromised patients from neurotoxic anesthetics. Choline alfoscerate (L-α-Glycerophosphorylcholine, α-GPC) is recognized for its neuroprotective properties against oxidative stress and inflammation, but its optimal therapeutic window and indications are still under investigation. This study explores the impact of α-GPC on human astrocytes, the most abundant cells in the brain that protect against oxidative stress, under isoflurane exposure. Methods: This study was designed to examine changes in factors related to isoflurane-induced toxicity following α-GPC administration. Primary human astrocytes were pretreated with varying doses of α-GPC (ranging from 0.1 to 10.0 µM) for 24 hours prior to 2.5% isoflurane exposure. In vitro analysis of cell morphology, water-soluble tetrazolium salt-1 assay, quantitative real-time polymerase chain reaction, proteome profiler array, and transcriptome sequencing were conducted. Results: A significant morphological damage to human astrocytes was observed in the group that had been pretreated with 10.0 mM of α-GPC and exposed to 2.5% isoflurane. A decrease in cell viability was identified in the group pretreated with 10.0 µM of α-GPC and exposed to 2.5% isoflurane compared to the group exposed only to 2.5% isoflurane. Quantitative real-time polymerase chain reaction revealed that mRNA expression of heme-oxygenase 1 and hypoxia-inducible factor-1α, which were reduced by isoflurane, was further suppressed by 10.0 µM α-GPC pretreatment. The proteome profiler array demonstrated that α-GPC pretreatment influenced a variety of factors associated with apoptosis induced by oxidative stress. Additionally, transcriptome sequencing identified pathways significantly related to changes in isoflurane-induced toxicity caused by α-GPC pretreatment. Conclusion: The findings suggest that α-GPC pretreatment could potentially enhance the vulnerability of primary human astrocytes to isoflurane-induced toxicity by diminishing the expression of antioxidant factors, potentially leading to amplified cell damage.

3.
Theranostics ; 11(2): 941-957, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33391514

RESUMEN

Extra-domain B of fibronectin (EDB-FN) is an alternatively spliced form of fibronectin with high expression in the extracellular matrix of neovascularized tissues and malignant cancer cells. In this study, we evaluated the practicality of using EDB-FN as a biomarker and therapeutic target for malignant gliomas (MGs), representative intractable diseases involving brain tumors. Methods: The microarray- and sequence-based patient transcriptomic database 'Oncopression' and tissue microarray of MG patient tissue samples were analyzed. EDB-FN data were extracted and evaluated from 23,344 patient samples of 17 types of cancer to assess its effectiveness and selectivity as a molecular target. To strengthen the results of the patient data analysis, the utility of EDB-FN as a molecular marker and target for MG was verified using active EDB-FN-targeting ultrasmall lipidic micellar nanoparticles (~12 nm), which had a high drug-loading capacity and were efficiently internalized by MG cells in vitro and in vivo. Results: Brain tumors had a 1.42-fold cancer-to-normal ratio (p < 0.0001), the second highest among 17 cancers after head and neck cancer. Patient tissue microarray analysis showed that the EDB-FN high-expression group had a 5.5-fold higher risk of progression than the EDB-FN low-expression group (p < 0.03). By labeling docetaxel-containing ultrasmall micelles with a bipodal aptide targeting EDB-FN (termed APTEDB-DSPE-DTX), we generated micelles that could specifically bind to MG cells, leading to superior antitumor efficacy of EDB-FN-targeting nanoparticles compared to nontargeting controls. Conclusions: Taken together, these results show that EDB-FN can be an effective drug delivery target and biomarker for MG.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Docetaxel/farmacología , Sistemas de Liberación de Medicamentos , Fibronectinas/metabolismo , Glioma/tratamiento farmacológico , Nanopartículas/administración & dosificación , Animales , Antineoplásicos/farmacología , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proliferación Celular , Docetaxel/química , Femenino , Fibronectinas/química , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Glioma/patología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Pronóstico , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Br J Cancer ; 123(7): 1204, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32820222

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

5.
Br J Cancer ; 123(7): 1123-1130, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32546833

RESUMEN

BACKGROUND: Epithelial-mesenchymal transition (EMT) is the most common cause of death in colorectal cancer (CRC). In this study, we investigated the functional roles of miRNA-17-5p in EMT of CRC cells. METHODS: In order to determine if miRNA-17-5p regulated EMT, the precursors and inhibitors of miR-17-5p were transduced into four CRC cells. To evaluate the regulatory mechanism, we performed argonaute 2 (Ago2) immunoprecipitation (IP) and luciferase assay. In addition, we used an intra-splenic injection mouse model of BALB/c nude mice to investigate the metastatic potential of miRNA-17-5p in vivo. RESULTS: The miRNA-17-5p expression was lower in primary CRC tissues with metastasis than in primary CRC tissues without metastasis in our RNA sequencing data of patient tissue. Real-time quantitative PCR revealed that miRNA-17-5p was inversely correlated with that of vimentin in five CRC cell lines. Over-expression of miRNA-17-5p decreased vimentin expression and inhibited cell migration and invasion in both LoVo and HT29 cells. However, inhibition of miRNA-17-5p showed the opposite effect. Ago2 IP and luciferase assay revealed that miRNA-17-5p directly bound to the 3'UTR of VIM mRNA. Furthermore, miRNA-17-5p inhibited the metastasis of CRC into liver in vivo. CONCLUSIONS: Our results demonstrated that miRNA-17-5p regulates vimentin expression, thereby regulating metastasis of CRC.


Asunto(s)
Neoplasias Colorrectales/patología , Transición Epitelial-Mesenquimal , MicroARNs/fisiología , Vimentina/genética , Animales , Línea Celular Tumoral , Movimiento Celular , Neoplasias Colorrectales/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Metástasis de la Neoplasia
6.
Exp Mol Med ; 52(5): 815-831, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32444799

RESUMEN

Colorectal cancer is a devastating disease with a low 5-year survival rate. Recently, many researchers have studied the mechanisms of tumor progression related to the tumor microenvironment. Here, we addressed the prognostic value of tumor-associated macrophages (TAMs) using a total of 232 CRC patient tissue samples and investigated the mechanisms underlying TAM-related colon cancer progression with respect to PI3Kγ regulation using in vitro, in vivo, and ex vivo approaches. Patients with M2/M1 < 3 had significantly improved progression-free survival and overall survival compared with patients with M2/M1 > 3. M1 and M2 macrophages elicited opposite effects on colon cancer progression via the FBW7-MCL-1 axis. Blocking macrophage PI3Kγ had cytotoxic effects on colon cancer cells and inhibited epithelial-mesenchymal transition features by regulating the FBW7-MCL-1 axis. The results of this study suggest that macrophage PI3Kγ may be a promising target for immunotherapy in colon cancer.


Asunto(s)
Plasticidad de la Célula/inmunología , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Animales , Apoptosis , Biomarcadores , Línea Celular Tumoral , Movimiento Celular , Plasticidad de la Célula/genética , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Neoplasias del Colon/etiología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Neoplasias del Colon/terapia , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Transición Epitelial-Mesenquimal/inmunología , Femenino , Humanos , Activación de Macrófagos/genética , Macrófagos/efectos de los fármacos , Macrófagos/patología , Fenotipo , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Pronóstico , Macrófagos Asociados a Tumores/efectos de los fármacos , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo , Macrófagos Asociados a Tumores/patología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Adv Exp Med Biol ; 1231: 33-43, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32060844

RESUMEN

The tumor microenvironment is the primary location in which tumor cells and the host immune system interact. There are many physiological, biochemical, cellular mechanisms in the neighbor of tumor which is composed of various cell types. Interactions of chemokines and chemokine receptors can recruit immune cell subsets into the tumor microenvironment. These interactions can modulate tumor progression and metastasis. In this chapter, we will focus on chemokine (C-C motif) ligand 7 (CCL7) that is highly expressed in the tumor microenvironment of various cancers, including colorectal cancer, breast cancer, oral cancer, renal cancer, and gastric cancer. We reviewed how CCL7 can affect cancer immunity and tumorigenesis by describing its regulation and roles in immune cell recruitment and stromal cell biology.


Asunto(s)
Quimiocina CCL7/inmunología , Transducción de Señal/inmunología , Microambiente Tumoral/inmunología , Animales , Carcinogénesis/inmunología , Humanos , Receptores de Quimiocina/metabolismo
8.
Oncol Rep ; 42(5): 2029-2038, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31432145

RESUMEN

In vitro culture of patient­derived tumor cells offers many advantages in the development of novel therapies for colorectal cancer. Although various culture systems have been developed, the long­term expansion of patient­derived tumor cells remains challenging. The present results suggested that tumor cells isolated from colorectal cancer patient­derived xenografts can be efficiently immortalized in conditioned medium from irradiated feeder cells containing Y­27632, a rho­associated coiled­coil containing protein kinase (ROCK) inhibitor. Patient­derived tumor cells proliferated rapidly, reaching 90­95% confluence in ~6 days. Short tandem repeat analysis suggested that these tumor tissues and cultured cells presented 13 identical short tandem repeat loci, including Amelogenin, Penta E, Penta D, D2S1338 and D19S433. Their epithelial phenotype was confirmed by staining for epithelial cell adhesion molecule and cytokeratin 20, whereas vimentin was used as a mesenchymal marker. When cells were transferred to 3D cultures, they continued to proliferate, forming well­defined tumor spheroids. Expression levels of human telomerase reverse transcriptase and C­Myc mRNA were increased in cultured cells. Finally, immortalized cells were used for the screening of 65 anticancer drugs approved by the Food and Drug Administration, allowing the identification of gene­drug associations. In the present study, primary culture models of colorectal cancer were efficiently established using a ROCK inhibitor and feeder cells, and this approach could be used for personalized treatment strategies for patients with colorectal cancer.


Asunto(s)
Amidas/farmacología , Neoplasias Colorrectales/patología , Células Nutrientes/citología , Cultivo Primario de Células/métodos , Piridinas/farmacología , Células Tumorales Cultivadas/citología , Anciano , Animales , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Medios de Cultivo Condicionados/química , Femenino , Humanos , Masculino , Ratones , Repeticiones de Microsatélite , Persona de Mediana Edad , Trasplante de Neoplasias , Esferoides Celulares/citología , Esferoides Celulares/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos
9.
Curr Drug Deliv ; 16(1): 86-92, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30246640

RESUMEN

BACKGROUND: Chrysin is a strong inhibitor of breast cancer resistance protein (BCRP) but it is practically insoluble in water. Effective solubilization of chrysin is critical for its pharmaceutical application as an absorption enhancer via inhibition of BCRP-mediated drug efflux. OBJECTIVE: This study aimed to develop an effective oral formulation of chrysin to improve its in vivo effect as an absorption enhancer. METHOD: Solid dispersions (SDs) of chrysin were prepared with hydrophilic carriers having surface acting properties and a pH modulator. In vitro and in vivo characterizations were performed to select the optimal SDs of chrysin. RESULTS: SDs with Brij®L4 and aminoclay was most effective in increasing the solubility of chrysin by 13-53 fold at varying drug-carrier ratios. Furthermore, SDs significantly improved the dissolution rate and extent of drug release. SDs (chrysin: Brij®L4: aminoclay=1:3:5) achieved approximately 60% and 83% drug release within 1 h and 8 h, respectively, in aqueous medium, while the dissolution of the untreated chrysin was less than 13%. XRD patterns indicated the amorphous state of chrysin in SDs. The SD formulation was effective in improving the bioavailability of topotecan, a BCRP substrate in rats. Following oral administration of topotecan with the SDs of chrysin, the Cmax and AUC of topotecan was enhanced by approximately 2.6- and 2-fold, respectively, while the untreated chrysin had no effect. CONCLUSION: The SD formulation of chrysin with Brij®L4 and aminoclay appeared to be promising in improving the dissolution of chrysin and enhancing its in vivo effect as an absorption enhancer.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Flavonoides/farmacología , Absorción Intestinal/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Administración Oral , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Disponibilidad Biológica , Arcilla/química , Detergentes/química , Composición de Medicamentos/métodos , Evaluación Preclínica de Medicamentos , Flavonoides/química , Flavonoides/uso terapéutico , Células HT29 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Masculino , Polidocanol/química , Ratas , Ratas Sprague-Dawley , Solubilidad , Topotecan/farmacología , Topotecan/uso terapéutico , Agua/química
10.
Oncotarget ; 8(45): 78365-78378, 2017 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-29108235

RESUMEN

Injection of the Helicobacter pylori cytotoxin-associated gene A (CagA) is closely associated with the development of chronic gastritis and gastric cancer. Individuals infected with H. pylori possessing the CagA protein produce more reactive oxygen species (ROS) and show an increased risk of developing gastric cancer. Sirtuins (SIRTs) are nicotinamide adenine dinucleotide (NAD+)-dependent deacetylases and mitochondrial SIRT3 is known to be a tumor suppressor via its ability to suppress ROS and hypoxia inducible factor 1α (HIF-1α). However, it is unclear whether increased ROS production by H. pylori is regulated by SIRT3 followed by HIF-1α regulation and whether intracellular CagA acts as a regulator thereof. In this study, we investigated correlations among SIRT3, ROS, and HIF-1α in H. pylori-infected gastric epithelial cells. We observed that SIRT3-deficient AGS cells induce HIF-1α protein stabilization and augmented transcriptional activity under hypoxic conditions. In CagA +H. pylori infected cells, CagA protein localized to mitochondria where it subsequently suppressed SIRT3 proteins. CagA +H. pylori infection also increased HIF-1α activity through the ROS production induced by the downregulated SIRT3 activity, which is similar to the hypoxic condition in gastric epithelial cells. In contrast, overexpression of SIRT3 inhibited the HIF-1α protein stabilization and attenuated the increase in HIF-1α transcriptional activity under hypoxic conditions. Moreover, CagA +H. pylori attenuated HIF-1α stability and decreased transcriptional activity in SIRT3-overexpressing gastric epithelial cells. Taken together, these findings provide valuable insights into the potential role of SIRT3 in CagA +H. pylori-mediated gastric carcinogenesis and a possible target for cancer prevention via inhibition of HIF-1α.

11.
Drug Deliv ; 24(1): 1731-1739, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29124978

RESUMEN

The present study aimed to develop the sustained-release oral dosage form of pelubiprofen (PEL) by using the blended mixture of 3-aminopropyl functionalized-magnesium phyllosilicate (aminoclay) and pH-independent polymers. The sustained-release solid dispersion (SRSD) was prepared by the solvent evaporation method and the optimal composition of SRSD was determined as the weight ratio of drug: Eudragit® RL PO: Eudragit® RS PO of 1:1:2 in the presence of 1% of aminoclay (SRSD(F6)). The dissolution profiles of SRSD(F6) were examined at different pHs and in the simulated intestinal fluids. The drug release from SRSD(F6) was limited at pH 1.2 and gradually increased at pH 6.8, resulting in the best fit to Higuchi equation. The sustained drug release from SRSD(F6) was also maintained in simulated intestinal fluid at fasted-state (FaSSIF) and fed-state (FeSSIF). The structural characteristics of SRSD(F6) were examined by using powder X-ray diffraction (PXRD), differential scanning calorimetry (DSC) and Fourier transform infrared spectroscopy (FT-IR), indicating the change of drug crystallinity to an amorphous form. After oral administration in rats, SRSD(F6) exhibited the prolonged drug exposure in plasma. For both PEL and PEL-transOH (active metabolite), once a day dosing of SRSD(F6) achieved oral exposure (AUC) comparable to those from the multiple dosing (3 times a day) of untreated drug. In addition, the in vivo absorption of SRSD(F6) was well-correlated with the in vitro dissolution data, establishing a good level A in vitro/in vivo correlation. These results suggest that SRSD(F6) should be promising for the sustained-release of PEL, thereby reducing the dosing frequency.


Asunto(s)
Silicatos de Aluminio/química , Preparaciones de Acción Retardada/química , Fenilpropionatos/química , Polímeros/química , Silicatos de Aluminio/metabolismo , Animales , Rastreo Diferencial de Calorimetría/métodos , Química Farmacéutica/métodos , Arcilla , Preparaciones de Acción Retardada/metabolismo , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Liberación de Fármacos , Concentración de Iones de Hidrógeno , Mucosa Intestinal/metabolismo , Masculino , Fenilpropionatos/metabolismo , Polímeros/metabolismo , Ácidos Polimetacrílicos/química , Ácidos Polimetacrílicos/metabolismo , Ratas , Ratas Sprague-Dawley , Solubilidad , Espectroscopía Infrarroja por Transformada de Fourier/métodos , Difracción de Rayos X/métodos
12.
Cancer Chemother Pharmacol ; 78(4): 735-44, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27520631

RESUMEN

PURPOSE: The present study aimed to discover a new potent BCRP inhibitor overcoming multidrug resistance. METHODS: Effects of LW6 on the functional activity and gene expression of two major efflux transporters, BCRP and P-gp, were evaluated by using MDCKII cells overexpressing each transporter (MDCKII-BCRP and MDCKII-MDR1). Its effects on the cytotoxicity and pharmacokinetics of co-administered anticancer drugs were also evaluated in transfected cells and rats, respectively. RESULTS: In MDCKII-BCRP cells overexpressing BCRP, LW6 enhanced significantly (p < 0.05) the cellular accumulation of mitoxantrone, a BCRP substrate, and was more potent than Ko143, a well-known BCRP inhibitor. LW6 also down-regulated BCRP expression at concentrations of 0.1-10 µM. Furthermore, cells became more susceptible to the cytotoxicity of anticancer drugs in the presence of LW6. The CC50 values of mitoxantrone and doxorubicin were reduced by three- and tenfold, respectively, in MDCKII-BCRP cells, while LW6 did not affect the cytotoxicity of anticancer drugs in MDCKII-mock cells lacking BCRP transporter. Furthermore, LW6 improved the oral exposure of methotrexate by twofold in rats. In contrast to BCRP, LW6 had no inhibition effect on the functional activity and gene expression of P-gp. CONCLUSION: LW6 was newly identified as a potent BCRP inhibitor and could be useful to reduce the multidrug resistance of cancer cells via the inhibition of BCRP-mediated drug efflux as well as the down-regulation of BCRP expression.


Asunto(s)
Acetanilidas/farmacología , Adamantano/análogos & derivados , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Descubrimiento de Drogas , Resistencia a Antineoplásicos/efectos de los fármacos , Subfamilia B de Transportador de Casetes de Unión a ATP/antagonistas & inhibidores , Subfamilia B de Transportador de Casetes de Unión a ATP/biosíntesis , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/antagonistas & inhibidores , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/biosíntesis , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Acetanilidas/farmacocinética , Acetanilidas/uso terapéutico , Adamantano/farmacocinética , Adamantano/farmacología , Adamantano/uso terapéutico , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Ratas , Ratas Sprague-Dawley
13.
Oncotarget ; 7(35): 57066-57076, 2016 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-27494849

RESUMEN

Colorectal cancer (CRC) with microsatellite instability (MSI) may exhibit impaired epithelial-mesenchymal transition (EMT), but little is known about the underlying mechanisms of this phenomenon. In this study, we investigated the role of Twist1 and its downstream signaling cascades in EMT induction according to MSI status. To investigate the effects of Twist1 on EMT induction according to MSI status, MSS LS513 and MSI LoVo colon cancer cell lines, which overexpress human Twist1, were generated. Twist1-induced EMT and its downstream signaling pathways were evaluated via in vitro and in vivo experiments. We found that Twist1 induced EMT markers and stem cell-like characteristics via AKT signaling pathways. Twist1 induced activation of AKT and suppression of glycogen synthase kinase (GSK)-3ß, which resulted in the activation of ß-catenin, increasing CD44 expression. In addition, Twist1 activated the AKT-induced NF-κB pathway, increasing CD44 and CD166 expression. Activation of both the AKT/GSK-3ß/ß-catenin and AKT/NF-κB pathways occurred in MSS LS513 cells, while only the AKT/GSK-3ß/ß-catenin pathway was activated in MSI LoVo cells. In conclusion, Twist1 induces stem cell-like characteristics in colon cancer cell lines related to EMT via AKT signaling pathways, and those pathways depend on MSI status.


Asunto(s)
Neoplasias del Colon/metabolismo , Transición Epitelial-Mesenquimal , Inestabilidad de Microsatélites , Proteínas Nucleares/metabolismo , Proteína 1 Relacionada con Twist/metabolismo , Animales , Anticuerpos/química , Antígenos CD/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Recuento de Células , Línea Celular Tumoral , Movimiento Celular , Neoplasias del Colon/genética , Femenino , Proteínas Fetales/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Proteínas Nucleares/genética , Transducción de Señal , Células Madre/metabolismo , Proteína 1 Relacionada con Twist/genética
14.
Oncotarget ; 7(24): 36842-36853, 2016 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-27167205

RESUMEN

Chemokine ligand 7 (CCL7) enhances cancer progression and metastasis via epithelial-mesenchymal transition (EMT). However, little is known about the molecular mechanism of CCL7-induced EMT signaling cascade in colon cancer. Thus, the objective of this study was to investigate CCL7-induced EMT signaling pathway and its role in the progression and metastasis of colon cancer. To demonstrate the effect of CCL7 on EMT induction, HCT116 and HT29 cells overexpressing CCL7 were generated. CCL7-induced EMT and its downstream signaling pathway were evaluated by both in vitro and in vivo experiments. In in vitro studies, CCL7 was found to interplay with CC chemokine receptor 3 (CCR3), resulting in enhanced cellular proliferation, invasion, and migration via ERK and JNK signaling pathway. To validate these findings, we established ectopic and orthotopic mouse models injected with CCL7-overexpressed cells. In ectopic mouse models, we observed that CCL7-overexpressed cells grew significantly faster than control cells. In orthotopic mouse models, we found that liver and lung metastasis developed only in mice injected with CCL7-overexpressed cells. This study is the first one focusing on the EMT cascade via CCL7-CCR3-ERK-JNK signaling axis in colon cancer. Our novel findings will improve our understanding on the mechanism of metastatic process and provide potential therapeutic strategies for preventing metastasis in colon cancer.


Asunto(s)
Quimiocina CCL7/metabolismo , Neoplasias del Colon/patología , Sistema de Señalización de MAP Quinasas/fisiología , Invasividad Neoplásica/patología , Receptores CCR3/metabolismo , Animales , Neoplasias del Colon/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Células HCT116 , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Receptor Cross-Talk/fisiología
15.
Oncotarget ; 6(28): 25368-80, 2015 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-26196590

RESUMEN

SLC22A18, solute carrier family 22, member 18, has been proposed to function as a tumor suppressor based on its chromosomal location at 11p15.5, mutations and aberrant splicing in several types of cancer and down-regulation in glioblastoma. In this study, we sought to demonstrate the significance of SLC22A18 as a tumor suppressor in colorectal cancer (CRC) and provide mechanistic bases for its function. We first showed that the expression of SLC22A18 is significantly down-regulated in tumor tissues using matched normal-tumor samples from CRC patients. This finding was also supported by publically accessible data from The Cancer Genome Atlas (TCGA). Functionally, SLC22A18 inhibits colony formation and induces of G2/M arrest consistent with being a tumor suppressor. Interestingly, suppression of KRAS by RNA interference promotes SLC22A18 expression, and expression of SLC22A18 in turn inhibits KRAS(G12D)-mediated anchorage independent growth of NIH3T3 cells indicating a mutual negative interaction. Finally, we evaluated diagnostic and prognostic values of SLC22A18 using clinical and gene expression data from TCGA which revealed a significantly worse long-term prognosis for patients with low level SLC22A18 expression. In sum, we established SLC22A18 as a tumor suppressor in colon epithelial cells and propose that SLC22A18 is potentially a marker of diagnostic and prognostic values.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Biomarcadores de Tumor/genética , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Bases de Datos Genéticas , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HT29 , Humanos , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células 3T3 NIH , Proteínas de Transporte de Catión Orgánico/genética , Pronóstico , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Interferencia de ARN , Transducción de Señal , Transfección , Carga Tumoral , Proteínas Supresoras de Tumor/genética
16.
Lab Invest ; 94(12): 1370-81, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25286029

RESUMEN

Cancer stem cells have the capacity to form new tumors and are thus considered to be a cause of metastasis and tumor recurrence. However, many of the mechanisms determining cancer stem cell characteristics are still unknown. MicroRNAs (miRNAs) are possible modulators of cancer stem cell generation and may be involved in the retention of cancer stem cell characteristics. The aim of this study was to examine the miRNA expression profiles regulating the cancer stem-like cell characteristics in gastric cancer. We sorted gastric cancer stem-like cells using the stem cell marker CD44 by fluorescence-activated cell sorting. CD44(+) cells formed more and larger spheres compared with CD44(-) cells. Cancer stem cell markers were overexpressed in CD44(+) cells. CD44(+) cells showed increased expression of mesenchymal cell markers, whereas epithelial markers were downregulated. In miRNA microarray, the miR-106b family comprising miR-106b, miR-93, and miR-25 was significantly upregulated in CD44(+) cells than in CD44(-) cells. Smad7, which inhibits transforming growth factor-ß (TGF-ß)/Smad signaling as a target of the miR-106b family, was downregulated in CD44(+) cells. Furthermore, expression of TGF-ß/Smad signal molecules was activated in CD44(+) cells, in accordance with the action of the miR-106b family. Inhibition of miR-106b showed suppression of the TGF-ß/Smad signaling pathway and decreased self-renewal capacity and cell invasiveness. Our study suggests that CD44(+) gastric cancer cells show cancer stem cell properties with epithelial-mesenchymal transition (EMT). Increased miR-106b family expression regulated cancer stem-like cell properties, particularly EMT characteristics, through the TGF-ß/Smad signaling pathway in CD44(+) stem-like cells. Taken together, these results indicate that targeting miR-106b may be an effective form of cancer therapy in gastric cancer through the modulation of cancer stem cell characteristics.


Asunto(s)
Receptores de Hialuranos/análisis , MicroARNs/fisiología , Células Madre Neoplásicas/patología , Transducción de Señal , Proteínas Smad/fisiología , Neoplasias Gástricas/patología , Factor de Crecimiento Transformador beta/fisiología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Humanos , MicroARNs/antagonistas & inhibidores , Invasividad Neoplásica , Transducción de Señal/fisiología
17.
Nat Commun ; 5: 4423, 2014 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-25055241

RESUMEN

Cytotoxin-associated gene A (CagA) is an oncoprotein and a major virulence factor of H. pylori. CagA is delivered into gastric epithelial cells via a type IV secretion system and causes cellular transformation. The loss of epithelial adhesion that accompanies the epithelial-mesenchymal transition (EMT) is a hallmark of gastric cancer. Although CagA is a causal factor in gastric cancer, the link between CagA and the associated EMT has not been elucidated. Here, we show that CagA induces the EMT by stabilizing Snail, a transcriptional repressor of E-cadherin expression. Mechanistically we show that CagA binds GSK-3 in a manner similar to Axin and causes it to shift to an insoluble fraction, resulting in reduced GSK-3 activity. We also find that the level of Snail protein is increased in H. pylori infected epithelium in clinical samples. These results suggest that H. pylori CagA acts as a pathogenic scaffold protein that induces a Snail-mediated EMT via the depletion of GSK-3.


Asunto(s)
Antígenos Bacterianos/fisiología , Proteínas Bacterianas/fisiología , Regulación hacia Abajo/fisiología , Transición Epitelial-Mesenquimal/fisiología , Glucógeno Sintasa Quinasa 3/metabolismo , Helicobacter pylori/fisiología , Factores de Transcripción/fisiología , Biopsia , Carcinogénesis/metabolismo , Carcinogénesis/patología , Células Cultivadas , Células Epiteliales/metabolismo , Células Epiteliales/patología , Mucosa Gástrica/metabolismo , Gastritis/metabolismo , Gastritis/patología , Humanos , Transducción de Señal/fisiología , Factores de Transcripción de la Familia Snail , Estómago/patología , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
18.
Helicobacter ; 19(6): 465-75, 2014 12.
Artículo en Inglés | MEDLINE | ID: mdl-25052887

RESUMEN

BACKGROUND: Chronic infection with Helicobacter pylori (H. pylori) is causally linked with gastric carcinogenesis. Virulent H. pylori strains deliver bacterial CagA into gastric epithelial cells. Induction of high motility and an elongated phenotype is considered to be CagA-dependent process. Casein kinase 2 plays a critical role in carcinogenesis through signaling pathways related to the epithelial mesenchymal transition. This study was aimed to investigate the effect of H. pylori infection on the casein kinase 2-mediated migration and invasion in gastric epithelial cells. MATERIALS AND METHODS: AGS or MKN28 cells as human gastric epithelial cells and H. pylori strains Hp60190 (ATCC 49503, CagA(+)) and Hp8822 (CagA(-)) were used. Cells were infected with H. pylori at multiplicity of infection of 100 : 1 for various times. We measured in vitro kinase assay to examine casein kinase 2 activity and performed immunofluorescent staining to observe E-cadherin complex. We also examined ß-catenin transactivation through promoter assay and MMP7 expression by real-time PCR and ELISA. RESULTS: H. pylori upregulates casein kinase 2 activity and inhibition of casein kinase 2 in H. pylori-infected cells profoundly suppressed cell invasiveness and motility. We confirmed that casein kinase 2 mediates membranous α-catenin depletion through dissociation of the α-/ß-catenin complex in H. pylori-infected cells. We also found that H. pylori induces ß-catenin nuclear translocation and increases MMP7 expressions mediated through casein kinase 2. CONCLUSIONS: We show for the first time that CagA(+) H. pylori upregulates cellular invasiveness and motility through casein kinase 2. The demonstration of a mechanistic interplay between H. pylori and casein kinase 2 provides important insights into the role of CagA(+) H. pylori in the gastric cancer invasion and metastasis.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Quinasa de la Caseína II/metabolismo , Células Epiteliales/enzimología , Infecciones por Helicobacter/enzimología , Helicobacter pylori/metabolismo , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Quinasa de la Caseína II/genética , Movimiento Celular , Células Epiteliales/citología , Células Epiteliales/microbiología , Mucosa Gástrica/citología , Mucosa Gástrica/enzimología , Mucosa Gástrica/microbiología , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/fisiopatología , Helicobacter pylori/genética , Humanos , Transducción de Señal
19.
Oncol Rep ; 31(2): 673-8, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24297371

RESUMEN

Genistein is an isoflavone from soy with multiple action targets in cellular processes. Hedgehog signaling and its activator Gli1 are involved not only in oncogenesis, but also in cancer stemness and overexpression of CD44, a typical cancer stem cell surface marker. It has been shown that levels of Gli1 and CD44 expression are downregulated by genistein. Genistein may modulate distinctive cellular characteristics in cancer stem cells by inhibiting Gli1-related signaling pathways. In the present study, we sorted cells from MKN45, a human gastric cancer cell line, according to CD44 expression. CD44(+) cells showed properties of cancer stem-like cells and formed sphere colonies. In addition, sonic hedgehog (Shh) signaling genes were upregulated in CD44(+) cells when compared with these levels in CD44(-) cells. When CD44(+) cancer stem-like cells were treated with genistein, Gli1 and CD44 mRNA and protein expression was significantly reduced. Moreover, other stem cell markers were downregulated by genistein. Gli1 siRNA was used to confirm the action of genistein in inhibiting Gli1 expression. The high cell migration capacity of CD44(+) cells was blocked by genistein. in conclusion, genistein inhibits Gli1 gene expression, resulting in the attenuation of cancer stem-like properties in gastric cancer cells. In addition, genistein suppresses the cell invasive capacity that is required for tumor growth and metastasis. Our data showed that genistein can be an effective agent for gastric cancer therapy by targeting cancer stem-like characteristics.


Asunto(s)
Genisteína/farmacología , Receptores de Hialuranos/biosíntesis , Células Madre Neoplásicas/citología , Neoplasias Gástricas/tratamiento farmacológico , Factores de Transcripción/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/biosíntesis , Anticarcinógenos/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo , Proteínas Hedgehog/biosíntesis , Humanos , Invasividad Neoplásica/genética , Proteínas de Neoplasias/biosíntesis , Nestina/biosíntesis , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Complejo Represivo Polycomb 1/biosíntesis , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal/efectos de los fármacos , Esferoides Celulares , Neoplasias Gástricas/prevención & control , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Células Tumorales Cultivadas , Proteína con Dedos de Zinc GLI1
20.
Lab Invest ; 92(6): 883-95, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22411066

RESUMEN

SMAD proteins are downstream effectors of the TGF-ß signaling pathway. Smad3-null mice develop colorectal cancer by 6 months of age. In this study, we have examined whether the loss of Smad3 promotes gastric neoplasia in mice. The stomachs of Smad3⁻/⁻ mice were compared with age-matched Smad3 heterozygous and wild-type mice. E-cadherin, Ki-67, phosphoSTAT3, and TFF2/SP expression was analyzed by immunohistochemisty. The short hairpin RNA (ShRNA)-mediated knockdown of Smad3 in AGS and MKN28 cells was also performed. In addition, we examined alterations in DCLK1-expressing cells. Smad3⁻/⁻ mouse stomachs at 6 months of age revealed the presence of exophytic growths along the lesser curvature in the proximal fundus. Six-month-old Smad3⁻/⁻ mouse stomachs showed metaplastic columnar glands initiating from the transition zone junction between the forestomach and the glandular epithelium along the lesser curvature. Ten-month-old Smad3⁻/⁻ mice all exhibited invasive gastric neoplastic changes with increased Ki-67, phosphoSTAT3 expression, and aberrant cytosolic E-cadherin staining in papillary glands within the invading submucosal gland. The shRNA-mediated knockdown of Smad3 in AGS and MKN28 cells promoted the expression of phosphoSTAT3. DCLK1-expressing cells, which also stained for the tuft cell marker acetylated-α-tubulin, were observed in 10-month-old Smad3⁻/⁻ mice within tumors and in fundic invasive lesions. In conclusion, Smad3-null mice develop gastric tumors in the fundus, which arise from the junction between the forestomach and the glandular epithelium and progress to prominent invasive tumors over time. Smad3-null mice represent a novel model of fundic gastric tumor initiated from forestomach/glandular transition zone along the lesser curvature.


Asunto(s)
Unión Esofagogástrica/patología , Fundus Gástrico/patología , Mucosa Gástrica/patología , Proteína smad3/deficiencia , Neoplasias Gástricas/patología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Quinasas Similares a Doblecortina , Unión Esofagogástrica/metabolismo , Femenino , Fundus Gástrico/metabolismo , Mucosa Gástrica/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos , Ratones Noqueados , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Factor de Transcripción STAT3/metabolismo , Neoplasias Gástricas/metabolismo , Factor Trefoil-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...