Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Exp Ther Med ; 25(2): 99, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36761005

RESUMEN

Mitochondrial transplantation is a popular field of research in cell-free therapy. Menstrual stem cells (MenSCs) are potential donor cells for provision of foreign mitochondria. The present study aimed to investigate the potential effects of MenSC-derived mitochondria on ovarian cancer from the perspective of protein expression profiling. MenSCs were harvested from menstrual blood. The mitochondria were isolated from MenSCs and ovarian cancer cell line SKOV3. A label-free mitochondria proteomics and analysis were performed by comparing the protein expression in mitochondria of MenSCs and SKOV3 cells. The differentially expressed proteins with fold-change >2 were analyzed by Gene Ontology, Kyoto Encyclopedia of Genes and Genomes pathway and protein domain enrichment, protein interaction networks and parallel reaction monitoring (PRM) analysis. In total, 592 proteins that were found to have increased expression in the mitochondria of MenSCs were analyzed. Functional enrichment analysis revealed these proteins were enriched in metabolism-associated pathway entries including 'oxidative phosphorylation' (OXPHOS) pathway. PRM analysis confirmed that four of 6 candidate proteins in the OXPHOS pathway showed similar increasing trends. The protein domain enrichment analysis showed that domains such as 'thioredoxin domain' were significantly enriched. Based on these findings, it was hypothesized that mitochondria from MenSCs have the potential to enhance progression of ovarian cancer likely mediated by the enrichment of OXPHOS-associated metabolic pathways.

2.
Kaohsiung J Med Sci ; 37(7): 547-561, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33728810

RESUMEN

Circular RNAs (circRNAs) appear to be significant modulators in various physiological processes. Recently, it is found that circRNA_101996 exerts important roles in various cancers. Our previous studies showed that circRNA_101996 promoted cervical cancer growth and metastasis by regulating miR-8075/TPX2. However, the potential regulatory role of circRNA_101996 in cervical cancer still needs further investigation. Our results in this study suggested that circRNA_101996 was over-expressed in cervical cancer patients. circRNA_101996 up-regulation remarkably assisted cell proliferation, cell cycle progression, and cell migration in cervical cancer, while circRNA_101996 knockdown exerted the inverse effects. The molecular investigations indicated that circRNA_101996 could increase the expression level of miR-1236-3p, tripartite motif-containing 37 (TRIM37), through binding to miR-1236-3p and reducing its expression. Moreover, in vivo results demonstrated that circRNA_101996 shRNA can function as a tumor suppressor through down-regulating TRIM37 in cervical cancer. In conclusion, our data indicated that circRNA_101996/miR-1236-3p/TRIM37 axis accelerated cervical cancer development, providing novel insights into cervical cancer diagnosis and treatment.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , ARN Circular/genética , Proteínas de Motivos Tripartitos/biosíntesis , Ubiquitina-Proteína Ligasas/biosíntesis , Neoplasias del Cuello Uterino/genética , Regiones no Traducidas 3' , Adulto , Anciano , Ciclo Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación hacia Abajo , Epitelio/metabolismo , Femenino , Humanos , Metástasis Linfática , Persona de Mediana Edad , Metástasis de la Neoplasia , ARN/metabolismo , ARN Interferente Pequeño/metabolismo , Neoplasias del Cuello Uterino/metabolismo
3.
Front Biosci (Landmark Ed) ; 26(12): 1411-1421, 2021 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-34994156

RESUMEN

BACKGROUND: Cervical cancer is a prevalent female malignancy with poor survival rates. ARID1A is frequently mutated or deleted in a variety of tumors and YAP signaling is widely activated in human malignancies. Nevertheless, the mechanism of YAP signaling in ARID1A-mutated cervical cancer remains unknown. METHODS: The cell viability was determined by MTT assay. The expression of ARID1A, YAP1 and CTGF were evaluated by western blot. The cell proliferation was detected by colony formation. RESULTS: The bioinformatics analysis suggested that mutation of ARID1A was associated with the activation of YAP1 signaling. In addition, knockdown of YAP1 inhibited ARID1A-mutated cervical cancer cells growth. Verteporfin is an inhibition of YAP1 signaling. Interestingly, knockdown of ARID1A decreased ARID1A-wildtype cervical cancer cells resistance to verteporfin. Meanwhile, overexpression of ARID1A increased ARID1A-mutated cervical cancer cells resistance to verteporfin. Similarly, blocking YAP1 signaling inhibited the tumor formation caused by ARID1A-mutated cervical cancer cells in vivo. CONCLUSION: Inhibition of YAP1 signaling suppresses ARID1A-mutated-induced tumorigenesis of cervical cancer, providing a novel therapeutic strategy for cervical cancer.


Asunto(s)
Proteínas de Unión al ADN , Factores de Transcripción , Neoplasias del Cuello Uterino , Proteínas Señalizadoras YAP/metabolismo , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular , Proteínas de Unión al ADN/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Transducción de Señal , Factores de Transcripción/genética , Neoplasias del Cuello Uterino/genética
4.
Cancer Biother Radiopharm ; 35(9): 650-660, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32186927

RESUMEN

Background: MIAT (myocardial infarction-associated transcript) regulates cell proliferation, apoptosis, and metastasis in several cancers. In this study, the authors aimed to explore the role of MIAT in ovarian cancer. Materials and Methods: The expression of MIAT in ovarian cancer subtypes, normal human ovarian surface epithelial and ovarian cancer cell lines was measured by qualitative real-time polymerase chain reaction (qRT-PCR). OVCAR3 and SKOV3 cells were transfected with MIAT overexpression plasmid or siMIAT. The cell growth ability was then evaluated by CCK-8 and colony formation assays. The cell migration and invasion rate were separately measured by wound-healing and transwell assays. The levels of epithelial-mesenchymal transition (EMT)-associated markers were evaluated by Western blotting. MIAT sponging miR-150-5p was predicted by starBase and confirmed by dual-luciferase reporter assays. The expression of miR-150-5p in OVCAR3 and SKOV3 cells with MIAT overexpression or knockdown, and in ovarian cancer subtypes was also measured by qRT-PCR. Further analyses confirmed the role of MIAT sponging miR-150-5p in ovarian cancer cells. Results: MIAT was highly expressed in mesenchymal subtype ovarian cancer tissues and ovarian cancer cells. In OVCAR3 and SKOV3 cells, overexpression of MIAT promoted, and knockdown of MIAT suppressed the cell growth, migration, invasion, and EMT. miR-150-5p was sponged and regulated by MIAT. miR-150-5p was downregulated in mesenchymal subtype ovarian cancer. Suppression of cell migration, invasion, and EMT caused by miR-150-5p overexpression was rescued by MIAT overexpression. Conclusions: MIAT acts as an oncogene in ovarian cancer cells through sponging miR-150-5p. MIAT or miR-150-5p expression might be a potential prognostic biomarker for ovarian cancer patients. MIAT and miR-150-5p are potential therapeutic targets in treatment of ovarian cancer.


Asunto(s)
Carcinoma Epitelial de Ovario/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Neoplasias Ováricas/genética , ARN Largo no Codificante/metabolismo , Apoptosis/genética , Carcinoma Epitelial de Ovario/patología , Carcinoma Epitelial de Ovario/cirugía , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , MicroARNs/genética , Invasividad Neoplásica/genética , Oncogenes , Neoplasias Ováricas/patología , Neoplasias Ováricas/cirugía , Ovariectomía , Ovario/patología , Ovario/cirugía , ARN Largo no Codificante/genética
5.
Int J Mol Med ; 45(2): 647-657, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31894307

RESUMEN

The molecular mechanisms underlying the activation of primordial follicles are poorly understood. The serine/threonine protein kinase phosphoinositide­dependent kinase 1 (PDK1), a pivotal downstream effector of phosphatidyl inositol­3 kinase (PI3K) signaling, plays a vital role in cellular signaling. In order to identify the function of PDK1 in ovarian follicle development, this study used conditional Pdk1 deletion in mouse oocytes by crossing Pdk1loxP/loxP mice with transgenic mice carrying Gdf­9 promoter­mediated Cre recombinase and found that Pdk1flx/flxGdf9Cre mice were subfertile with increased serum follicle­stimulating hormone (FSH) and luteinizing hormone (LH) levels compared with Pdk1flx/flx mice. The deletion of Pdk1 in oocytes induced massive primordial follicle activation, leading to premature ovarian failure (POF). Further investigation revealed that enhanced Yes­associated protein (YAP) expression and an increased pro­inflammatory response also contributed to massive primordial follicle activation. PDK1 formed the complex with the core kinases of Hippo signaling and regulated the expression levels of YAP. On the whole, the findings of the present study demonstrate that PDK1 serves as an indispensable gatekeeper for maintaining the primordial follicle pool and provide a deeper understanding of POF treatment.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas de Ciclo Celular/genética , Folículo Ovárico/metabolismo , Insuficiencia Ovárica Primaria/genética , Animales , Femenino , Eliminación de Gen , Inflamación/genética , Inflamación/patología , Ratones Endogámicos ICR , Oocitos/metabolismo , Folículo Ovárico/patología , Insuficiencia Ovárica Primaria/patología , Regulación hacia Arriba , Proteínas Señalizadoras YAP
6.
Mol Med ; 25(1): 32, 2019 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-31299894

RESUMEN

BACKGROUND: Emerging evidence shows that Hippo signal pathways can regulate the progression of various cancer. While the roles of Yes-associated protein (YAP), the key transducer of Hippo signals, in the development of endometrial cancer (EC) are rarely investigated. METHODS: The expression of YAP in endometrial cancer cells and tissues was measured. Its roles in proliferation and expression of interleukins (ILs) were investigated by use of its specific siRNA or inhibitor (verteporfin, VP). RESULTS: YAP was upregulated in endometrial cancer cells and tissues. Knockdown of YAP or VP can suppress the proliferation while increase its chemo-sensitivity of EC cells. We found that targeted inhibition of YAP can decrease the expression of interleukin-6 (IL-6) and IL-11 in EC cells. Recombinant IL-6 or IL-11 can attenuate si-YAP suppressed proliferation of EC cells. Chromatin immunoprecipitation (ChIP) assay suggested that YAP can directly bind with the promoter of IL-6 and induce its transcription. As to IL-11, inhibitor of NF-κB (BAY 11-7082) can significantly down regulate the mRNA expression of IL-11. Over expression of p65 abolished si-YAP suppressed transcription of IL-11. It suggested that NF-κB was involved in the YAP regulated expression of IL-11. CONCLUSIONS: YAP can regulate the proliferation and progression of EC cells. It suggested that targeted inhibition of YAP might be a potent potential approach for EC therapy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Interleucina-11/metabolismo , Interleucina-6/metabolismo , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Western Blotting , Línea Celular Tumoral , Proliferación Celular/genética , Proliferación Celular/fisiología , Inmunoprecipitación de Cromatina , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Técnicas In Vitro , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Proteínas Señalizadoras YAP
7.
Biochem Biophys Res Commun ; 516(2): 506-514, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31230753

RESUMEN

Bisphenol A (BPA) is a kind of environmental endocrine disruptors (EEDs) that interfere embryo implantation. Trophoblast invasion plays a crucial role during embryo implantation. In this study, the effects of BPA on invasion ability of human trophoblastic Jeg-3 spheroids and regulation of endothelial and stromal cells on trophoblastic spheroids invasion, and its possible mechanism were investigated. The results showed that BPA at 10 and 100 µM can inhibit the attachment of Jeg-3 spheroid onto Ishikawa cells. BPA at 1-100 µM also activate ERE-Luc reporter expression in the transfected cells, which was through the ERα, but not ERß or GPR30 binding. Endothelial receptivity ability was harmed by BPA treatment since receptivity markers of LIF, EGF, MUC1 and integrin αVß3 were decreased after BPA treatment. The invasion ability of trophoblastic spheroids generated from Jeg-3 cell line was inhibited by BPA and this effect was mediated through canonical ERs pathway and MMP2/MMP9 down-regulation and TIMP1/PAI-1 up-regulation. Besides, BPA treated decidualized stromal cells suppressed Jeg-3 spheroid outgrowth and invasion in co-culture assay. Our study would give a better understanding on the possible mechanism of BPA effect on human embryo implantation process.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Movimiento Celular , Decidua/patología , Células Endoteliales/patología , Fenoles/toxicidad , Esferoides Celulares/patología , Trofoblastos/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Células Endoteliales/efectos de los fármacos , Femenino , Humanos , Receptores de Estrógenos/metabolismo , Esferoides Celulares/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Células del Estroma/patología , Trofoblastos/efectos de los fármacos
8.
J Cell Physiol ; 234(8): 14296-14305, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30633364

RESUMEN

In recent years, circular RNAs have been shown to serve as essential regulators in several human cancers. Nevertheless, the function and mechanism of CircRNA in cervical cancer remain elusive. In the present study, we showed that hsa_circRNA_101996 was highly expressed in cervical cancer tissues compared with matched normal tissues by bioinformatics analysis. We showed that the expression level of hsa_circRNA_101996 in cervical cancer tissues was positively correlated with TNM stage, tumor size, and lymph node metastasis. Moreover, higher levels of hsa_circRNA_101996 were related to poor outcomes of cervical cancer patients. We found that knockdown of hsa_circRNA_101996 significantly inhibited the proliferation, cell cycle, migration, and invasion of cervical cancer cells. Mechanistically, we demonstrated that hsa_circRNA_101996 served as a sponge of miR-8075, which targeted TPX2 in cervical cancer cells. We showed that miR-8075 that was downregulated in cervical cancer tissues repressed cervical cancer cell proliferation, migration, and invasion. Furthermore, we validated that upregulation of TPX2 by hsa_circRNA_101996-mediated inhibition of miR-8075 contributed to cervical cancer proliferation, migration, and invasion. Taken together, our findings revealed a novel mechanism that hsa_circRNA_101996-miR-8075-TPX2 network promoted cervical cancer progression.


Asunto(s)
Proteínas de Ciclo Celular/genética , MicroARNs/genética , Proteínas Asociadas a Microtúbulos/genética , ARN Circular/genética , Neoplasias del Cuello Uterino/genética , Animales , Movimiento Celular/genética , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HeLa , Xenoinjertos , Humanos , Metástasis Linfática , Ratones , Persona de Mediana Edad , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Estadificación de Neoplasias , Supervivencia sin Progresión , Neoplasias del Cuello Uterino/patología
9.
Mol Med Rep ; 12(2): 2211-6, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25824616

RESUMEN

Previous studies have demonstrated that the presence of Hox transcript antisense intergenic RNA (HOTAIR) is correlated with poor survival in several types of cancer, including breast cancer, and promotes tumor metastasis. Currently, little is known regarding the correlation between HOTAIR and chemoresistance in cancer. The current study aimed to investigate the role of HOTAIR in epithelial ovarian cancer, and the correlation between HOTAIR and cisplatin resistance. Reverse transcription-quantitative polymerase chain reaction was conducted to detect HOTAIR expression in the ovarian specimens and ovarian carcinoma cell lines. The results indicated that the expression level of HOTAIR was higher in epithelial ovarian cancer tissues than the level in the benign ovarian tissues. The expression level was also higher in late-stage malignant ovarian tumors compared with the level in early-stage tumors. Levels of HOTAIR were also higher in the SKOV-3CDDP/R cisplatin-resistant ovarian carcinoma cell line than in the SKOV-3 cisplatin-sensitive cell line. The knockdown of HOTAIR using siRNAs with transfection reagent suppressed cell proliferation, reduced the invasion ability of the cells and notably, it restored cisplatin-sensitivity of the cisplatin-resistant cells specifically by enhancing cisplatin-induced cytotoxicity and apoptosis in SKOV-3CDDP/R cells. In conclusion, HOTAIR may be used in the development of novel treatments for ovarian cancer, particularly those that are resistant to conventional therapies.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Ováricas/genética , ARN Largo no Codificante/genética , Apoptosis/efectos de los fármacos , Carcinoma Epitelial de Ovario , Estudios de Casos y Controles , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Estadificación de Neoplasias , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Pronóstico , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal
10.
Asian Pac J Cancer Prev ; 15(7): 3051-6, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24815446

RESUMEN

Previous studies have demonstrated that JMJD2A is a potential oncogene and is overexpressed in human tumors. However, its role in the endometrial carcinoma remains largely unknown. In this study, we discovered that JMJD2A was overexpressed in endometrial carcinoma, using immunohistochemistry, quantitative real- time polymerase chain reaction, and western blotting. Downregulation of JMJD2A led to reduced endometrial carcinoma RL95-2 and ISK cell proliferation, invasion and metastasis as asessed with cell counting kit-8, cell migration and invasive assays. Collectively, our results support that JMJD2A is a promoter of endometrial carcinoma cell proliferation and survival, and is a potential novel drug target.


Asunto(s)
Neoplasias Endometriales/patología , Histona Demetilasas con Dominio de Jumonji/biosíntesis , Histona Demetilasas con Dominio de Jumonji/genética , Invasividad Neoplásica/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Histonas/metabolismo , Humanos , Inmunohistoquímica , Metástasis de la Neoplasia/genética , Interferencia de ARN , ARN Interferente Pequeño
11.
J Cancer Res Clin Oncol ; 136(6): 873-81, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19921255

RESUMEN

INTRODUCTION: Endostatin is the most potent inhibitor of tumor angiogenesis. However, endostatin protein has a short half-time and virus-mediated endostatin gene therapy has serious toxicity, which limits the application of endostatin in clinical therapy. Mesenchymal stem cells (MSCs) are considered to be able to accumulate at the site of cancers with high specificity and may be used as a new delivery of endostatin. MATERIALS AND METHODS: The MSCs from the human bone marrow were transfected with recombinant adenovirus encoding endostatin and EGFP (MSC-EN cells). The tropism capacity of MSCs was quantitatively assayed in vitro using the Millicell system. To investigate the impact of secreted endostatin on cancer cells, SKOV3 cells were co-cultured with MSC-EN cells in Millicell for 48 h, then apoptosis and cell cycle were analyzed on a flow cytometer. RESULTS: In contrast with 293 cells and saline, SKOV3 cells significantly stimulated migration of MSCs, the number reached 919.67 +/- 19.96 (P < 0.05). The endostatin produced by MSC-EN cells made 13.08 +/- 0.21% SKOV3 cells undergo early stage apoptosis (control 3.23 +/- 0.73%, P < 0.05) and 82.05 +/- 2.65% SKOV3 cells accumulate in the G0/G1 phase (control 66.51 +/- 2.91%, P < 0.05). CONCLUSION: We found that MSCs possessed great migratory capacity in vitro and the human ovarian adenocarcinoma cell line SKOV3 could significantly induce the migration of MSCs. Our results provided evidence that MSCs could be utilized as a powerful delivery system of endostatin. The endostatin produced by MSC-EN cells could inhibit the proliferation of SKOV3 cells.


Asunto(s)
Adenocarcinoma/terapia , Inhibidores de la Angiogénesis/farmacología , Apoptosis/efectos de los fármacos , Endostatinas/farmacología , Terapia Genética/métodos , Células Madre Mesenquimatosas , Neoplasias Ováricas/terapia , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/genética , Adenoviridae , Western Blotting , Línea Celular Tumoral , Proliferación Celular , Estudios de Factibilidad , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Neoplasias Ováricas/irrigación sanguínea , Neoplasias Ováricas/genética , Transfección
12.
Int J Gynecol Cancer ; 19(6): 998-1003, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19820359

RESUMEN

Matrix metalloproteinase 2 (MMP-2) and membrane type 1 matrix metalloproteinase (MT1-MMP) have been identified as important participants in tumor invasion, metastasis, and angiogenesis. Membrane type 1 matrix metalloproteinase has also been recognized as a major activator of MMP-2. The purpose of this study was to investigate epidermal growth factor (EGF) mediating signal pathways in the regulation of MMP-2 and MT1-MMP in SiHa cells, a cervical cancer cell line. We showed here that EGF induced the expression of MT1-MMP and inhibited the expression of MMP-2 at both the mRNA and protein levels. Membrane type 1 matrix metalloproteinase induction was blocked by mitogen-activated protein kinase or extracellular signal-regulated kinase inhibitors PD98059 and U0126 but not by phosphatidylinositol-3 kinase (PI3-K) inhibitors LY294002 and wortmannin. Interestingly, the mitogen-activated protein kinase or extracellular signal-regulated kinase inhibitors PD98059 and U0126 actually increased MMP-2 mRNA and protein synthesis, whereas the PI3-K inhibitors LY294002 and wortmannin further suppressed the expression of MMP-2. Our results suggest that EGF receptor up-regulated the expression of MT1-MMP and down-regulated the synthesis of MMP-2 through the mitogen-activated protein kinase/extracellular signal-regulated kinase pathway while concomitantly transmitting a mild positive regulatory signal to the expression of MMP-2 via the PI3-K/AKT pathway in SiHa cells. Furthermore, we found that EGF elevated the activity of MMP-2 in culture media.


Asunto(s)
Carcinoma/genética , Receptores ErbB/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Metaloproteinasa 14 de la Matriz/biosíntesis , Metaloproteinasa 2 de la Matriz/biosíntesis , Proteína Oncogénica v-akt/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Neoplasias del Cuello Uterino/genética , Carcinoma/patología , Línea Celular Tumoral , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/genética , Metaloproteinasa 14 de la Matriz/genética , Metaloproteinasa 14 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Proteína Oncogénica v-akt/metabolismo , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Neoplasias del Cuello Uterino/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...