Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Anticancer Res ; 44(3): 1131-1142, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38423649

RESUMEN

BACKGROUND/AIM: Cancer stem cells (CSCs) contribute significantly to the poor prognosis of patients with epithelial ovarian cancer (EOC) due to their roles in drug resistance and tumor metastasis. Autotaxin (ATX) plays a pivotal role in the maintenance of the CSC-like properties of EOC tumors. BBT-877 is a novel ATX inhibitor used in clinical treatment of idiopathic pulmonary fibrosis. However, the effects of BBT-877 on drug resistance and metastasis in ovarian CSCs remain unknown. In this study, we aimed to investigate the effects of BBT-877 on drug resistance and intraperitoneal metastasis of EOC. MATERIALS AND METHODS: Spheroid-forming CSCs, which were isolated from two EOC cell lines, A2780 and SKOV3, were investigated by cell viability, western blot, PCR, Spheroid-forming assay, and in vivo experiments. RESULTS: Spheroid-forming CSCs exhibited increased CSC-like properties and paclitaxel (PTX) resistance. BBT-877 treatment inhibited the viability of spheroid-forming CSCs more potently than that of adherent ovarian cancer cell lines. Combinatorial treatment with BBT-877 and PTX significantly attenuated the viability of spheroid-forming CSCs. In a SKOV3 cells-derived intraperitoneal metastasis model, BBT-877 treatment reduced the number of metastatic tumor nodes, while combinatorial treatment with BBT-877 and PTX more potently attenuated the formation of metastatic nodes and accumulation of ascitic fluid. CONCLUSION: These results suggest that BBT-877 can be combined with conventional anticancer drugs for the treatment of patients with recurrent or drug-resistant EOC.


Asunto(s)
Neoplasias Ováricas , Oxazoles , Piperazinas , Humanos , Femenino , Carcinoma Epitelial de Ovario/patología , Neoplasias Ováricas/patología , Línea Celular Tumoral , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Resistencia a Antineoplásicos , Células Madre Neoplásicas/metabolismo
2.
Int J Mol Sci ; 24(12)2023 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-37373457

RESUMEN

Epithelial ovarian cancer (EOC) is the most lethal gynecological malignancy owing to relapse caused by resistance to chemotherapy. We previously reported that cluster of differentiation 109 (CD109) expression is positively correlated with poor prognosis and chemoresistance in patients with EOC. To further explore the role of CD109 in EOC, we explored the signaling mechanism of CD109-induced drug resistance. We found that CD109 expression was upregulated in doxorubicin-resistant EOC cells (A2780-R) compared with that in their parental cells. In EOC cells (A2780 and A2780-R), the expression level of CD109 was positively correlated with the expression level of ATP-binding cassette (ABC) transporters, such as ABCB1 and ABCG2, and paclitaxel (PTX) resistance. Using a xenograft mouse model, it was confirmed that PTX administration in xenografts of CD109-silenced A2780-R cells significantly attenuated in vivo tumor growth. The treatment of CD109-overexpressed A2780 cells with cryptotanshinone (CPT), a signal transducer and activator of transcription 3 (STAT3) inhibitor, inhibited the CD109 overexpression-induced activation of STAT3 and neurogenic locus notch homolog protein 1 (NOTCH1), suggesting a STAT3-NOTCH1 signaling axis. The combined treatment of CD109-overexpressed A2780 cells with CPT and N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester (DAPT), a NOTCH inhibitor, markedly abrogated PTX resistance. These results suggest that CD109 plays a key role in the acquisition of drug resistance by activating the STAT3-NOTCH1 signaling axis in patients with EOC.


Asunto(s)
Neoplasias Ováricas , Humanos , Femenino , Animales , Ratones , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Factor de Transcripción STAT3/metabolismo , Línea Celular Tumoral , Recurrencia Local de Neoplasia , Paclitaxel/farmacología , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Resistencia a Antineoplásicos , Transportadoras de Casetes de Unión a ATP , Proteínas de Neoplasias/metabolismo , Antígenos CD/uso terapéutico , Proteínas Ligadas a GPI/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo
3.
Int J Mol Sci ; 24(7)2023 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-37047234

RESUMEN

NANOG, a stemness-associated transcription factor, is highly expressed in many cancers and plays a critical role in regulating tumorigenicity. Transformation/transcription domain-associated protein (TRRAP) has been reported to stimulate the tumorigenic potential of cancer cells and induce the gene transcription of NANOG. This study aimed to investigate the role of the TRRAP-NANOG signaling pathway in the tumorigenicity of cancer stem cells. We found that TRRAP overexpression specifically increases NANOG protein stability by interfering with NANOG ubiquitination mediated by FBXW8, an E3 ubiquitin ligase. Mapping of NANOG-binding sites using deletion mutants of TRRAP revealed that a domain of TRRAP (amino acids 1898-2400) is responsible for binding to NANOG and that the overexpression of this TRRAP domain abrogated the FBXW8-mediated ubiquitination of NANOG. TRRAP knockdown decreased the expression of CD44, a cancer stem cell marker, and increased the expression of P53, a tumor suppressor gene, in HCT-15 colon cancer cells. TRRAP depletion attenuated spheroid-forming ability and cisplatin resistance in HCT-15 cells, which could be rescued by NANOG overexpression. Furthermore, TRRAP knockdown significantly reduced tumor growth in a murine xenograft transplantation model, which could be reversed by NANOG overexpression. Together, these results suggest that TRRAP plays a pivotal role in the regulation of the tumorigenic potential of colon cancer cells by modulating NANOG protein stability.


Asunto(s)
Neoplasias del Colon , Animales , Humanos , Ratones , Carcinogénesis/genética , Línea Celular Tumoral , Neoplasias del Colon/patología , Regulación Neoplásica de la Expresión Génica , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Células Madre Neoplásicas/metabolismo , Estabilidad Proteica
4.
Mol Cells ; 44(7): 481-492, 2021 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-34326276

RESUMEN

Tribbles homolog 2 (TRIB2) is implicated in tumorigenesis and drug resistance in various types of cancers. However, the role of TRIB2 in the regulation of tumorigenesis and drug resistance of cancer stem cells (CSCs) is still elusive. In the present study, we showed increased expression of TRIB2 in spheroid-forming and aldehyde dehydrogenase-positive CSC populations of A2780 epithelial ovarian cancer cells. Short hairpin RNA-mediated silencing of TRIB2 expression attenuates the spheroid-forming, migratory, tumorigenic, and drug-resistant properties of A2780 cells, whereas overexpression of TRIB2 increases the CSC-like characteristics. TRIB2 overexpression induced GSK3ß inactivation by augmenting AKT-dependent phosphorylation of GSK3ß at Ser9, followed by increasing ß-catenin level via reducing the GSK3ß-mediated phosphorylation of ß-catenin. Treatment of TRIB2-ovexpressed A2780 cells with the phosphoinositide-3-kinase inhibitor LY294002 abrogated TRIB2-stimulated proliferation, migration, drug resistance of A2780 cells. These results suggest a critical role for TRIB2 in the regulation of CSC-like properties by increasing the stability of ß-catenin protein via the AKT-GSK3ß-dependent pathways.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , beta Catenina/metabolismo , Humanos , Transducción de Señal
5.
BMB Rep ; 53(12): 622-627, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32843129

RESUMEN

Cancer stem cells (CSCs) or tumor-initiating cells are thought to play critical roles in tumorigenesis, metastasis, drug resistance, and tumor recurrence. For the diagnosis and targeted therapy of CSCs, the molecular identity of biomarkers or therapeutic targets for CSCs needs to be clarified. In this study, we identified CD166 as a novel marker expressed in the sphereforming CSC population of A2780 epithelial ovarian cancer cells and primary ovarian cancer cells. The CD166+ cells isolated from A2780 cells and primary ovarian cancer cells highly expressed CSC markers, including ALDH1a1, OCT4, and SOX2, and ABC transporters, which are implicated in the drug resistance of CSCs. The CD166+ cells exhibited enhanced CSC-like properties, such as increased sphere-forming ability, cell migration and adhesion abilities, resistance to conventional anticancer drugs, and high tumorigenic potential in a xenograft mouse model. Knockdown of CD166 expression in the sphereforming ovarian CSCs abrogated their CSC-like properties. Moreover, silencing of CD166 expression in the sphere-forming CSCs suppressed the phosphorylation of focal adhesion kinase, paxillin, and SRC. These results suggest that CD166 plays a key role in the regulation of CSC-like properties and focal adhesion kinase signaling in ovarian cancer. [BMB Reports 2020; 53(12): 622-627].


Asunto(s)
Antígenos CD/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas Fetales/metabolismo , Células Madre Neoplásicas/metabolismo , Neoplasias Ováricas/metabolismo , Transportadoras de Casetes de Unión a ATP , Familia de Aldehído Deshidrogenasa 1 , Animales , Antígenos CD/genética , Biomarcadores de Tumor/metabolismo , Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular Neuronal/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Proteínas Fetales/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Células Madre Neoplásicas/fisiología , Factor 3 de Transcripción de Unión a Octámeros , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Ovario/metabolismo , Retinal-Deshidrogenasa , Factores de Transcripción SOXB1 , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Menopausal Med ; 25(1): 55-62, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31080790

RESUMEN

OBJECTIVES: Body mass index (BMI) is commonly used in epidemiological study or clinical center. However, it is not exactly correlated with body fat composition and does not reflect sex, age, or race. The aim of this article is to evaluate the validity of BMI standards relative to total body fat (TBF) and to estimate new BMI criteria that correspond to TBF for obesity, especially for Asian postmenopausal women. METHODS: A total 3,936 patients were included in this cross-sectional study, including 1,565 premenopausal and 2,371 postmenopausal women. At the time of visit, demographic data were collected. We demonstrated the validity of BMI cut-point of 25 kg/m2 by using area under the curve (AUC), and presented the empirical optimal BMI cut-point by using Youden's index and overall accuracy in both premenopausal and postmenopausal women. RESULTS: BMI-defined obesity (≥ 25 kg/m2) represents high AUC values (> 0.9) for each TBF. In premenopausal women, TBF ≥ 38% and corresponding BMI value was 29.45 kg/m2 indicated the highest both Youden's index and overall accuracy. In comparison, postmenopausal women who were TBF ≥ 38% showed the highest Youden's index and overall accuracy, and corresponding BMI value was 26.45 kg/m2. CONCLUSIONS: We proposed new BMI criteria for obesity by using TBF reference. With application of bioelectrical impedance analysis, the diagnosis of obesity using BMI criteria may differ between premenopausal and postmenopausal women.

7.
Ginekol Pol ; 89(11): 611-617, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30508213

RESUMEN

OBJECTIVES: The purpose of this study was to identify clinical risk factors for the recurrence of ovarian endometrioma after ovarian cystectomy in Korean women with long-term postoperative medical therapy. MATERIAL AND METHODS: A total of 134 patients who were surgically treated for endometriotic cysts at Pusan National University Hospital were included in this retrospective study. All patients received long-term postoperative medical treatment for at least 12 months after the first-line conservative surgery. Several epidemiologic variables were analyzed as possible risk factors for recurrence. Endometrioma recurrence was considered when a cystic mass was observed on transvaginal or transrectal sonography. Statistical analysis was performed using independent t-tests for parametric continuous variables. RESULTS: The mean follow-up period for the 134 patients was 56.5 ± 14.3 months (range, 36-120 months) and the mean duration of the medical therapy was 17.9 ± 17.3 months (range, 12-120 months). The overall recurrence rate was 35/134 (26.12%). Our univariate analysis showed statistically significant differences between the recurrent and non-recurrent groups in terms of weight (P = 0.013), body mass index (P = 0.007), age at the time of surgery (P = 0.013), the diameter of the largest cyst (P = 0.001), the presence of dysmenorrhea (P < 0.0001), and postoperative pregnancy (P = 0.016). Multivariate analysis showed that body mass index (OR 1.153, 95% CI 1.003-1.326, P = 0.046), age at the time of surgery (OR 0.924, 95% CI 0.860-0.992, P = 0.029), and presence of dysmenorrhea (OR 12.226, 95% CI 3.543-42.188, P < 0.0001) were significantly correlated with the recurrence of endometrioma. CONCLUSIONS: We found that patients with dysmenorrhea after surgery, and a younger age of the patient at the time of surgery were the highest risk factors associated with the recurrence of endometrioma, despite long-term postoperative medication.


Asunto(s)
Anticonceptivos Hormonales Orales/uso terapéutico , Dismenorrea/epidemiología , Endometriosis/terapia , Procedimientos Quirúrgicos Ginecológicos , Enfermedades del Ovario/terapia , Progestinas/uso terapéutico , Adulto , Factores de Edad , Índice de Masa Corporal , Femenino , Humanos , Análisis Multivariante , Oportunidad Relativa , Cuidados Posoperatorios , Periodo Posoperatorio , Embarazo/estadística & datos numéricos , Recurrencia , República de Corea , Estudios Retrospectivos , Factores de Riesgo , Factores de Tiempo
8.
BMB Rep ; 51(10): 514-519, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29936929

RESUMEN

Ovarian cancer is the most fatal gynecological malignancy in women and identification of new therapeutic targets is essential for the continued development of therapy for ovarian cancer. TRRAP (transformation/transcription domain-associated protein) is an adaptor protein and a component of histone acetyltransferase complex. The present study was undertaken to investigate the roles played by TRRAP in the proliferation and tumorigenicity of ovarian cancer stem cells. TRRAP expression was found to be up-regulated in the sphere cultures of A2780 ovarian cancer cells. Knockdown of TRRAP significantly decreased cell proliferation and the number of A2780 spheroids. In addition, TRRAP knockdown induced cell cycle arrest and increased apoptotic percentages of A2780 sphere cells. Notably, the mRNA levels of stemness-associated markers, that is, OCT4, SOX2, and NANOG, were suppressed in TRRAP-silenced A2780 sphere cells. In addition, TRRAP overexpression increased the mRNA level of NANOG and the transcriptional activity of NANOG promoter in these cells. Furthermore, TRRAP knockdown significantly reduced tumor growth in a murine xenograft transplantation model. Taken together, the findings of the present study suggest that TRRAP plays an important role in the regulation of the proliferation and stemness of ovarian cancer stem cells. [BMB Reports 2018; 51(10): 515-520].


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas Nucleares/metabolismo , Neoplasias Ováricas/patología , Animales , Apoptosis , Biomarcadores de Tumor/metabolismo , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos
9.
Cancer Metastasis Rev ; 37(2-3): 509-518, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29926310

RESUMEN

Stem cells are a rare subpopulation defined by the potential to self-renew and differentiate into specific cell types. A population of stem-like cells has been reported to possess the ability of self-renewal, invasion, metastasis, and engraftment of distant tissues. This unique cell subpopulation has been designated as cancer stem cells (CSC). CSC were first identified in leukemia, and the contributions of CSC to cancer progression have been reported in many different types of cancers. The cancer stem cell hypothesis attempts to explain tumor cell heterogeneity based on the existence of stem cell-like cells within solid tumors. The elimination of CSC is challenging for most human cancer types due to their heightened genetic instability and increased drug resistance. To combat these inherent abilities of CSC, multi-pronged strategies aimed at multiple aspects of CSC biology are increasingly being recognized as essential for a cure. One of the most challenging aspects of cancer biology is overcoming the chemotherapeutic resistance in CSC. Here, we provide an overview of autotaxin (ATX), lysophosphatidic acid (LPA), and their signaling pathways in CSC. Increasing evidence supports the role of ATX and LPA in cancer progression, metastasis, and therapeutic resistance. Several studies have demonstrated the ATX-LPA axis signaling in different cancers. This lipid mediator regulatory system is a novel potential therapeutic target in CSC. In this review, we summarize the evidence linking ATX-LPA signaling to CSC and its impact on cancer progression and metastasis. We also provide evidence for the efficacy of cancer therapy involving the pharmacological inhibition of this signaling pathway.


Asunto(s)
Neoplasias/enzimología , Neoplasias/patología , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/patología , Hidrolasas Diéster Fosfóricas/metabolismo , Animales , Humanos , Lisofosfolípidos/metabolismo
10.
Oncotarget ; 7(34): 55624-55638, 2016 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-27489349

RESUMEN

Hypoxia and NOTCH signaling have been reported to be associated with the self-renewal and drug resistance of cancer stem cells (CSCs). However, the molecular mechanisms by which hypoxia and NOTCH signaling stimulate the self-renewal and drug resistance of ovarian CSCs are poorly understood. In the present study, we identified SOX2 as a key transcription factor for CSC-like characteristics in the downstream of hypoxia-induced NOTCH signaling in epithelial ovarian cancer cells. Hypoxic treatment or overexpression of intracellular domain of NOTCH1 (NICD1) in ovarian cancer cells increased sphere formation, drug resistance, and expression of CSC-associated genes such as SOX2, ALDH, and ABC transporters. Hypoxic treatment increased the expression of NICD1, and hypoxic treatment or NICD1 overexpression increased SOX2 promoter activity, which was inhibited by deletion of HIF-1 or CSL binding sites. Furthermore, DAPT treatment decreased the effect of hypoxic treatment, and SOX2 knockdown decreased the effect of hypoxic treatment and NICD overexpression on sphere formation and drug resistance in established ovarian cancer cell lines and primary ovarian cancer cells. These results suggest that hypoxia-NOTCH1-SOX2 signaling axis is important for activation of ovarian CSCs, which may provide a novel opportunity for developing therapeutics to eradicate CSCs in ovarian cancer patients.


Asunto(s)
Hipoxia de la Célula , Células Madre Neoplásicas/fisiología , Neoplasias Ováricas/patología , Receptor Notch1/fisiología , Factores de Transcripción SOXB1/fisiología , Transducción de Señal/fisiología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Femenino , Humanos , Regiones Promotoras Genéticas , Factores de Transcripción SOXB1/genética
11.
Exp Mol Med ; 48: e255, 2016 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-27561949

RESUMEN

Cancer stem cells are a subpopulation of cancer cells characterized by self-renewal ability, tumorigenesis and drug resistance. The aim of this study was to investigate the role of HMGA1, a chromatin remodeling factor abundantly expressed in many different cancers, in the regulation of cancer stem cells in ovarian cancer. Spheroid-forming cancer stem cells were isolated from A2780, SKOV3 and PA1 ovarian cancer cells by three-dimensional spheroid culture. Elevated expression of HMGA1 was observed in spheroid cells along with increased expression of stemness-related genes, such as SOX2, KLF4, ALDH, ABCB1 and ABCG2. Furthermore, spheroid A2780 cells, compared with adherent cells, showed higher resistance to chemotherapeutic agents such as paclitaxel and doxorubicin. HMGA1 knockdown in spheroid cells reduced the proliferative advantage and spheroid-forming efficiency of the cells and the expression of stemness-related genes. HMGA1 overexpression in adherent A2780 cells increased cancer stem cell properties, including proliferation, spheroid-forming efficiency and the expression of stemness-related genes. In addition, HMGA1 regulated ABCG2 promoter activity through HMGA1-binding sites. Knockdown of HMGA1 in spheroid cells reduced resistance to chemotherapeutic agents, whereas the overexpression of HMGA1 in adherent ovarian cancer cells increased resistance to chemotherapeutic agents in vitro. Furthermore, HMGA1-overexpressing A2780 cells showed a significant survival advantage after chemotherapeutic agent treatment in a xenograft tumorigenicity assay. Together, our results provide novel insights regarding the critical role of HMGA1 in the regulation of the cancer stem cell characteristics of ovarian cancer cells, thus suggesting that HMGA1 may be an important target in the development of therapeutics for ovarian cancer patients.


Asunto(s)
Proliferación Celular , Resistencia a Antineoplásicos , Proteína HMGA1a/metabolismo , Células Madre Neoplásicas/patología , Neoplasias Ováricas/patología , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteína HMGA1a/análisis , Proteína HMGA1a/genética , Humanos , Factor 4 Similar a Kruppel , Ratones Endogámicos BALB C , Ratones Desnudos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Ovario/efectos de los fármacos , Ovario/metabolismo , Ovario/patología , Esferoides Celulares , Células Tumorales Cultivadas
12.
Stem Cells ; 34(3): 551-64, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26800320

RESUMEN

Ovarian cancer shows high mortality due to development of resistance to chemotherapy and relapse. Cancer stem cells (CSCs) have been suggested to be a major contributor in developing drug resistance and relapse in ovarian cancer. In this study, we isolated CSCs through sphere culture of A2780, SKOV3, OVCAR3 epithelial ovarian cancer cells and primary ovarian cancer cells from patients. We identified heat-stable factors secreted from ovarian CSCs stimulated migration and proliferation of CSCs. Mass spectrometry and ELISA analysis revealed that lysophosphatidic acid (LPA) was significantly elevated in CSC culture media compared with non-CSC culture media. Treatment of CSCs with LPA resulted in augmented CSC characteristics such as sphere-forming ability, resistance to anticancer drugs, tumorigenic potential in xenograft transplantation, and high expression of CSC-associated genes, including OCT4, SOX2, and aldehyde dehydrogenase 1. Treatment of CSCs with LPA receptor 1-specific inhibitors or silencing of LPA receptor 1 expression abrogated the LPA-stimulated CSC properties. Autotaxin, an LPA-producing enzyme, is highly secreted from ovarian CSCs, and pharmacological inhibition or knockdown of autotaxin markedly attenuated the LPA-producing, tumorigenic, and drug resistance potentials of CSCs. Clinicopathological analysis showed a significant survival disadvantage of patients with positive staining of autotaxin. In addition, we further identified that AKT1 activity was upregulated in ovarian CSCs through an LPA-dependent mechanism and silencing of AKT1 expression led to suppression of CSC characteristics. These results suggest that autotaxin-LPA-LPA receptor 1-AKT1 signaling axis is critical for maintaining CSC characteristics through an autocrine loop and provide a novel therapeutic target for ovarian CSCs.


Asunto(s)
Lisofosfolípidos/administración & dosificación , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Hidrolasas Diéster Fosfóricas/genética , Receptores del Ácido Lisofosfatídico/genética , Ataxina-1/genética , Comunicación Autocrina/efectos de los fármacos , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Glandulares y Epiteliales/patología , Células Madre Neoplásicas/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal/efectos de los fármacos
13.
Oncotarget ; 7(3): 3506-19, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26654944

RESUMEN

Ovarian cancer has the highest mortality rate of all gynecological cancers with a high recurrence rate. It is important to understand the nature of recurring cancer cells to terminally eliminate ovarian cancer. The winged helix transcription factor Forkhead box P1 (FOXP1) has been reported to function as either oncogene or tumor-suppressor in various cancers. In the current study, we show that FOXP1 promotes cancer stem cell-like characteristics in ovarian cancer cells. Knockdown of FOXP1 expression in A2780 or SKOV3 ovarian cancer cells decreased spheroid formation, expression of stemness-related genes and epithelial to mesenchymal transition-related genes, cell migration, and resistance to Paclitaxel or Cisplatin treatment, whereas overexpression of FOXP1 in A2780 or SKOV3 ovarian cancer cells increased spheroid formation, expression of stemness-related genes and epithelial to mesenchymal transition-related genes, cell migration, and resistance to Paclitaxel or Cisplatin treatment. In addition, overexpression of FOXP1 increased promoter activity of ABCG2, OCT4, NANOG, and SOX2, among which the increases in ABCG2, OCT4, and SOX2 promoter activity were dependent on the presence of FOXP1-binding site. In xenotransplantation of A2780 ovarian cancer cells into nude mice, knockdown of FOXP1 expression significantly decreased tumor size. These results strongly suggest FOXP1 functions as an oncogene by promoting cancer stem cell-like characteristics in ovarian cancer cells. Targeting FOXP1 may provide a novel therapeutic opportunity for developing a relapse-free treatment for ovarian cancer patients.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Madre Neoplásicas/patología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Proteínas Represoras/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis , Western Blotting , Adhesión Celular , Movimiento Celular , Proliferación Celular , Resistencia a Antineoplásicos , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Factores de Transcripción Forkhead/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Neoplasias Ováricas/enzimología , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Células Tumorales Cultivadas , Cicatrización de Heridas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
PLoS One ; 10(7): e0131785, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26148001

RESUMEN

Endothelial progenitor cells (EPCs) can be isolated from human bone marrow or peripheral blood and reportedly contribute to neovascularization. Aptamers are 40-120-mer nucleotides that bind to a specific target molecule, as antibodies do. To utilize apatmers for isolation of EPCs, in the present study, we successfully generated aptamers that recognize human CD31, an endothelial cell marker. CD31 aptamers bound to human umbilical cord blood-derived EPCs and showed specific interaction with human CD31, but not with mouse CD31. However, CD31 aptamers showed non-specific interaction with CD31-negative 293FT cells and addition of polyanionic competitor dextran sulfate eliminated non-specific interaction without affecting cell viability. From the mixture of EPCs and 293FT cells, CD31 aptamers successfully isolated EPCs with 97.6% purity and 94.2% yield, comparable to those from antibody isolation. In addition, isolated EPCs were decoupled from CD31 aptamers with a brief treatment of high concentration dextran sulfate. EPCs isolated with CD31 aptamers and subsequently decoupled from CD31 aptamers were functional and enhanced the restoration of blood flow when transplanted into a murine hindlimb ischemia model. In this study, we demonstrated isolation of foreign material-free EPCs, which can be utilized as a universal protocol in preparation of cells for therapeutic transplantation.


Asunto(s)
Aptámeros de Nucleótidos/metabolismo , Células Progenitoras Endoteliales/fisiología , Isquemia/terapia , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Animales , Anticuerpos/inmunología , Diferenciación Celular/fisiología , Células Cultivadas , Células Progenitoras Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Células HEK293 , Miembro Posterior/irrigación sanguínea , Miembro Posterior/metabolismo , Miembro Posterior/fisiología , Humanos , Isquemia/metabolismo , Ratones , Neovascularización Fisiológica/fisiología , Trasplante de Células Madre/métodos
15.
Oncol Res Treat ; 37(1-2): 30-4, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24613906

RESUMEN

PURPOSE: Positron emission tomography/computer tomography (PET/CT) utilizing [(18)F]fluorodeoxyglucose ((18)F-FDG) has been recommended for the diagnosis, staging, therapy monitoring, prediction of prognosis, and detection of recurrence in endometrial cancer. We aimed to define the correlations of biological markers with (18)F-FDG uptake in endometrial cancer. METHODS: 29 patients (55 ± 8.93 years) with endometrial cancer were included in this study. All patients underwent hysterectomy and bilateral salpingo-oophorectomy with or without para-aortic lymphadenectomy at the department of Obstetrics and Gynecology of Pusan National University Hospital. Immunohistochemical studies were performed for glucose transporter 1 (GLUT-1), hexokinase II (HK-II), hypoxia-inducible factor 1α (HIF-1α), vascular endothelial growth factor (VEGF) and carbonic anhydrase IX (CA-IX). RESULTS: There were positive correlations between FDG uptake and GLUT-1 (r = 0.375, p = 0.0452), and HK-II (r = 0.537, p = 0.0027). However, HIF-1α (r = 0.153, p = 0.4283), VEGF (r = -0,101, p = 0.6032) and CA-IX (r = 0.240, p = 0.2105) were not significantly associated with FDG uptake. No significant correlations were found among the expression levels of biological markers. CONCLUSION: FDG uptake in endometrial cancer was significantly associated with GLUT-1 and HK-II, while HIF-1α, VEGF and CA-IX were not associated with FDG uptake. No significant correlations were found among the expression levels of biological markers.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Endometriales/diagnóstico por imagen , Neoplasias Endometriales/metabolismo , Fluorodesoxiglucosa F18/farmacocinética , Proteínas de Neoplasias/metabolismo , Tomografía de Emisión de Positrones/métodos , Adulto , Anciano , Femenino , Humanos , Persona de Mediana Edad , Radiofármacos/farmacocinética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Estadística como Asunto
16.
Obstet Gynecol Sci ; 56(2): 76-83, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24327985

RESUMEN

OBJECTIVE: To investigate the clinical significance of atypical glandular cells (AGC) by analyzing the prevalence and histologic outcomes of patients with AGC according to Pap smear. METHODS: The medical records of 83 patients who were diagnosed AGC on Pap tests at the Pusan National University Hospital outpatient department and health care center from January 1998 to March 2006 were reviewed. RESULTS: The prevalence of AGC was 55 of 54,160 (0.10%) and 28 of 54,160 (0.05%) for AGC-not otherwise specified (NOS) and neoplastic associated AGC, respectively. The histopathologic results of the AGC-NOS group (n=55) were as follows: low-grade squamous intraepithelial lesion, 7 (12.7%); high-grade squamous intraepithelial lesion, 4 (7.2%); adenocarcinoma of cervix, 3 (5.4%); endometrial carcinoma, 2 (3.6%); and other malignancies including 2 ovarian cancer cases and 1 breast cancer case, 3 (5.4%). The histopathologic results for the AGC-associated neoplastic group (n=28) were as follows: low-grade squamous intraepithelial lesion, 1 (3.5%); high-grade squamous intraepithelial lesion, 3 (10.7%); adenocarcinoma of cervix, 5 (17.8%); endometrial carcinoma, 4 (4.8%); and additional malignancies including 3 stomach cancer cases, 2 ovarian cancer cases, and 2 breast cancer cases; 7 (25%). CONCLUSION: AGCs may represent a variety of benign and malignant lesions. AGC-associated neoplastic findings may be related to gynecological or extrauterine malignancies. Thus, when AGCs, especially neoplastic AGCs, are encountered, it is best to evaluate the cervix not only for typical maladies, but also for gynecological and non-gynecological malignancies.

17.
Cell Physiol Biochem ; 32(2): 253-63, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23942151

RESUMEN

BACKGROUND: Transcriptional co-activator with PDZ-binding motif (TAZ), a downstream effector of the Hippo pathway, has been reported to regulate organ size, tissue homeostasis, and tumorigenesis by acting as a transcriptional co-activator. Lysophosphatidic acid (LPA) is a bioactive lipid implicated in tumorigenesis and metastasis of ovarian cancer through activation of G protein-coupled receptors. However, the involvement of TAZ in LPA-induced tumorigenesis of ovarian cancer has not been elucidated. METHODS: In order to demonstrate the role of TAZ in LPA-stimulated tumorigenesis, the effects of LPA on TAZ expression and cell migration were determined by Western blotting and chemotaxis analyses in R182 human epithelial ovarian cancer cells. RESULTS AND CONCLUSION: Treatment of R182 cells with the LPA receptor inhibitor Ki16425 blocked LPA-induced cell migration. In addition, transfection of R182 cells with small interfering RNA specific for LPA receptor 1 resulted in abrogation of LPA-stimulated cell migration. LPA induced phosphorylation of ERK and p38 MAP kinase in R182 cells and pretreatment of cells with the MEK-ERK pathway inhibitor U0126, but not the p38 MAPK inhibitor SB202190, resulted in abrogation of LPA-induced cell migration. Pretreatment of R182 cells with U0126 attenuated LPA-induced mRNA levels of TAZ and its transcriptional target genes, such as CTGF and CYR61, without affecting phosphorylation level of YAP. These results suggest that MEK-ERK pathway plays a key role in LPA-induced cell migration and mRNA expression of TAZ in R182 cells, without affecting stability of TAZ protein. In addition, small interfering RNA-mediated silencing of TAZ expression attenuated LPA-stimulated migration of R182 cells. These results suggest that TAZ plays a key role in LPA-stimulated migration of epithelial ovarian cancer cells.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisofosfolípidos/toxicidad , Butadienos/farmacología , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Isoxazoles/farmacología , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Nitrilos/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Propionatos/farmacología , Estabilidad Proteica/efectos de los fármacos , Transactivadores , Factores de Transcripción , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ
18.
Int J Biochem Cell Biol ; 45(8): 1869-77, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23735324

RESUMEN

Oncostatin M, a member of the interleukin-6 family of cytokines, has been implicated in tumorigenesis of human prostate cancer. In the current study, we demonstrate that oncostatin M promotes human adipose tissue-derived mesenchymal stem cell-stimulated tumor growth in an in vivo xenograft transplantation model of the human prostate cancer cell line PC-3M-luc-C6, a PC3M cell line expressing the luciferase gene. Conditioned medium derived from oncostatin M-treated mesenchymal stem cells stimulated adhesion of PC-3M-luc-C6 cells. We identified TGFBI and periostin, extracellular matrix proteins implicated in tumorigenesis and metastasis, as oncostatin M-induced secreted proteins in mesenchymal stem cells. Treatment with oncostatin M stimulated secretion of periostin and TGFBI from mesenchymal stem cells in a time-dependent manner. Immunodepletion of TGFBI and periostin from conditioned medium derived from oncostatin M-treated mesenchymal stem cells resulted in abrogation of adhesion of PC-3M-luc-C6 cells stimulated by oncostatin M-conditioned medium. In addition, small interfering RNA-mediated silencing of TGFBI and periostin resulted in abrogation of cell adhesion stimulated by oncostatin M-conditioned medium. These results suggest that mesenchymal stem cell-derived TGFBI and periostin play a key role in tumorigenesis by stimulating adhesion of prostate cancer cells.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Células Madre Mesenquimatosas/citología , Oncostatina M/farmacología , Comunicación Paracrina/efectos de los fármacos , Neoplasias de la Próstata/patología , Factor de Crecimiento Transformador beta/metabolismo , Tejido Adiposo/citología , Animales , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Silenciador del Gen/efectos de los fármacos , Humanos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Desnudos , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/farmacología , Trasplante de Células Madre , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Child Psychiatry Hum Dev ; 44(3): 460-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23073610

RESUMEN

Due to increased multiculturalism in the US and abroad, there is a need for increased understanding of the different ways in which parenting stress is related to child problems across cultures. In the present study, we investigated (a) differences in reported parenting stress and childhood problem behaviors across a Korean (n = 71) and US (n = 71) sample, as well as (b) differences in the ways in which parenting stress and childhood problems were related across Korean and US children based on mothers' reports. Results revealed that Korean mothers reported significantly higher parenting stress yet significantly lower childhood problem behaviors compared to US mothers. In addition, mother-based reports of child problems were significantly associated with parenting stress in the US sample, but not in the Korean sample. Clinical implications and culturally-relevant issues relevant to these findings are addressed, including a potential under-reporting bias of child problems among Asian parents.


Asunto(s)
Trastornos de la Conducta Infantil/etnología , Comparación Transcultural , Madres/psicología , Responsabilidad Parental/etnología , Estrés Psicológico/etnología , Adolescente , Adulto , Femenino , Humanos , Masculino , República de Corea/etnología , Estados Unidos/etnología
20.
Int J Biochem Cell Biol ; 44(11): 2069-76, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22903068

RESUMEN

Lysophosphatidic acid (LPA) is involved in mesenchymal stem cell-stimulated tumor growth in vivo. However, the molecular mechanism by which mesenchymal stem cells promote tumorigenesis remains elusive. In the present study, we demonstrate that conditioned medium from A549 human lung adenocarcinoma cells (A549 CM) induced the expression of ADAM12, a disintegrin and metalloproteases family member, in human adipose tissue-derived mesenchymal stem cells (hASCs). A549 CM-stimulated ADAM12 expression was abrogated by pretreatment of hASCs with the LPA receptor 1 inhibitor Ki16425 or by small interfering RNA-mediated silencing of LPA receptor 1, suggesting a key role for the LPA-LPA receptor 1 signaling axis in A549 CM-stimulated ADAM12 expression. Silencing of ADAM12 expression using small interfering RNA or short hairpin RNA abrogated LPA-induced expression of both α-smooth muscle actin, a marker of carcinoma-associated fibroblasts, and ADAM12 in hASCs. Using a xenograft transplantation model of A549 cells, we demonstrated that silencing of ADAM12 inhibited the hASC-stimulated in vivo growth of A549 xenograft tumors and the differentiation of transplanted hASCs to α-smooth muscle actin-positive carcinoma-associated fibroblasts. LPA-conditioned medium from hASCs induced the adhesion of A549 cells and silencing of ADAM12 inhibited LPA-induced expression of extracellular matrix proteins, periostin and ßig-h3, in hASCs and LPA-conditioned medium-stimulated adhesion of A549 cells. These results suggest a pivotal role for LPA-stimulated ADAM12 expression in tumor growth and the differentiation of hASCs to carcinoma-associated fibroblasts expressing α-smooth muscle actin, periostin, and ßig-h3.


Asunto(s)
Proteínas ADAM/metabolismo , Tejido Adiposo/citología , Lisofosfolípidos/farmacología , Proteínas de la Membrana/metabolismo , Células Madre Mesenquimatosas/enzimología , Neoplasias/patología , Proteína ADAM12 , Actinas/metabolismo , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Animales , Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Proteínas de la Matriz Extracelular/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Silenciador del Gen/efectos de los fármacos , Humanos , Neoplasias Pulmonares/patología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Desnudos , Neoplasias/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA