Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cancer ; 22(1): 4, 2023 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-36624516

RESUMEN

BACKGROUND: Metastatic colonization is one of the critical steps in tumor metastasis. A pre-metastatic niche is required for metastatic colonization and is determined by tumor-stroma interactions, yet the mechanistic underpinnings remain incompletely understood. METHODS: PCR-based miRNome profiling, qPCR, immunofluorescent analyses evaluated the expression of exosomal miR-141 and cell-to-cell communication. LC-MS/MS proteomic profiling and Dual-Luciferase analyses identified YAP1 as the direct target of miR-141. Human cytokine profiling, ChIP, luciferase reporter assays, and subcellular fractionation analyses confirmed YAP1 in modulating GROα production. A series of in vitro tumorigenic assays, an ex vivo model and Yap1 stromal conditional knockout (cKO) mouse model demonstrated the roles of miR-141/YAP1/GROα/CXCR1/2 signaling cascade. RNAi, CRISPR/Cas9 and CRISPRi systems were used for gene silencing. Blood sera, OvCa tumor tissue samples, and tissue array were included for clinical correlations. RESULTS: Hsa-miR-141-3p (miR-141), an exosomal miRNA, is highly secreted by ovarian cancer cells and reprograms stromal fibroblasts into proinflammatory cancer-associated fibroblasts (CAFs), facilitating metastatic colonization. A mechanistic study showed that miR-141 targeted YAP1, a critical effector of the Hippo pathway, reducing the nuclear YAP1/TAZ ratio and enhancing GROα production from stromal fibroblasts. Stromal-specific knockout (cKO) of Yap1 in murine models shaped the GROα-enriched microenvironment, facilitating in vivo tumor colonization, but this effect was reversed after Cxcr1/2 depletion in OvCa cells. The YAP1/GROα correlation was demonstrated in clinical samples, highlighting the clinical relevance of this research and providing a potential therapeutic intervention for impeding premetastatic niche formation and metastatic progression of ovarian cancers. CONCLUSIONS: This study uncovers miR-141 as an OvCa-derived exosomal microRNA mediating the tumor-stroma interactions and the formation of tumor-promoting stromal niche through activating YAP1/GROα/CXCRs signaling cascade, providing new insight into therapy for OvCa patients with peritoneal metastases.


Asunto(s)
MicroARNs , Neoplasias Ováricas , Humanos , Animales , Ratones , Femenino , Cromatografía Liquida , Proteómica , Espectrometría de Masas en Tándem , Neoplasias Ováricas/genética , MicroARNs/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Microambiente Tumoral
2.
Int J Mol Sci ; 23(19)2022 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-36233339

RESUMEN

Ovarian cancer is the deadliest gynecological cancer, leading to over 152,000 deaths each year. A late diagnosis is the primary factor causing a poor prognosis of ovarian cancer and often occurs due to a lack of specific symptoms and effective biomarkers for an early detection. Currently, cancer antigen 125 (CA125) is the most widely used biomarker for ovarian cancer detection, but this approach is limited by a low specificity. In recent years, multimarker panels have been developed by combining molecular biomarkers such as human epididymis secretory protein 4 (HE4), ultrasound results, or menopausal status to improve the diagnostic efficacy. The risk of ovarian malignancy algorithm (ROMA), the risk of malignancy index (RMI), and OVA1 assays have also been clinically used with improved sensitivity and specificity. Ongoing investigations into novel biomarkers such as autoantibodies, ctDNAs, miRNAs, and DNA methylation signatures continue to aim to provide earlier detection methods for ovarian cancer. This paper reviews recent advancements in molecular biomarkers for the early detection of ovarian cancer.


Asunto(s)
MicroARNs , Neoplasias Ováricas , Algoritmos , Autoanticuerpos , Biomarcadores de Tumor , Antígeno Ca-125 , Carcinoma Epitelial de Ovario , Femenino , Humanos , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/patología , Proteínas/metabolismo
3.
Int J Mol Sci ; 23(12)2022 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-35743298

RESUMEN

Ovarian cancer is one of the most lethal gynecological malignancies worldwide, and chemoresistance is a critical obstacle in the clinical management of the disease. Recent studies have suggested that exploiting cancer cell metabolism by applying AMP-activated protein kinase (AMPK)-activating agents and distinctive adjuvant targeted therapies can be a plausible alternative approach in cancer treatment. Therefore, the perspectives about the combination of AMPK activators together with VEGF/PD-1 blockade as a dual-targeted therapy against ovarian cancer were discussed herein. Additionally, ferroptosis, a non-apoptotic regulated cell death triggered by the availability of redox-active iron, have been proposed to be governed by multiple layers of metabolic signalings and can be synergized with immunotherapies. To this end, ferroptosis initiating therapies (FITs) and metabolic rewiring and immunotherapeutic approaches may have substantial clinical potential in combating ovarian cancer development and progression. It is hoped that the viewpoints deliberated in this review would accelerate the translation of remedial concepts into clinical trials and improve the effectiveness of ovarian cancer treatment.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Neoplasias Ováricas , Proteínas Quinasas Activadas por AMP/metabolismo , Carcinoma Epitelial de Ovario , Femenino , Humanos , Lípidos/uso terapéutico , Neoplasias Ováricas/patología , Receptor de Muerte Celular Programada 1 , Factor A de Crecimiento Endotelial Vascular/uso terapéutico
4.
Int J Mol Sci ; 23(23)2022 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-36499591

RESUMEN

Ovarian cancer is one of the most lethal gynecological cancers worldwide. The poor prognosis of this malignancy is substantially attributed to the inadequate symptomatic biomarkers for early diagnosis and effective remedies to cure the disease against chemoresistance and metastasis. Ovarian cancer metastasis is often relatively passive, and the single clusters of ovarian cancer cells detached from the primary ovarian tumor are transcoelomic spread by the peritoneal fluid throughout the peritoneum cavity and omentum. Our earlier studies revealed that lipid-enriched ascitic/omental microenvironment enforced metastatic ovarian cancer cells to undertake metabolic reprogramming and utilize free fatty acids as the main energy source for tumor progression and aggression. Intriguingly, cell susceptibility to ferroptosis has been tightly correlated with the dysregulated fatty acid metabolism (FAM), and enhanced iron uptake as the prominent features of ferroptosis are attributed to the strengthened lipid peroxidation and aberrant iron accumulation, suggesting that ferroptosis induction is a targetable vulnerability to prevent cancer metastasis. Therefore, the standpoints about tackling altered FAM in combination with ferroptosis initiation as a dual-targeted therapy against advanced ovarian cancer were highlighted herein. Furthermore, a discussion on the prospect and challenge of inducing ferroptosis as an innovative therapeutic approach for reversing remedial resistance in cancer interventions was included. It is hoped this proof-of-concept review will indicate appropriate directions for speeding up the translational application of ferroptosis-inducing compounds (FINs) to improve the efficacy of ovarian cancer treatment.


Asunto(s)
Ferroptosis , Neoplasias Ováricas , Neoplasias Peritoneales , Femenino , Humanos , Metabolismo de los Lípidos , Neoplasias Peritoneales/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Epiplón , Microambiente Tumoral
5.
Cancer Cell Int ; 18: 65, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29743815

RESUMEN

BACKGROUND: Ovarian cancer is the most lethal gynaecological malignancy. Chemotherapy is the main stay of treatment for metastatic disease, with modest response rates but significant side effects. Therefore, there is a need for alternative therapies that can control the disease while offering good quality of life. Ovarian cancer cells express both estrogen receptor subtypes (ERα and ERß). There is growing evidence that ERß is anti-oncogenic. Genistein and daidzein are phytoestrogens found in soybeans and they display higher affinity to bind ERß. ERB-041 is a potent selective ERß agonist. In this study, we aimed to investigate the effects of genistein, daidzein and ERB-041 on ovarian cancer. METHODS: Ovarian cancer cell lines were treated with genistein, daidzein and ERB-041 in pharmacological doses. Cell migration, invasion, proliferation, cell cycle arrest, apoptosis and sphere formation were assessed by Transwell migration and invasion assays, XTT assay, focus formation, flow cytometry and sphere formation assay, respectively. Immunoblotting analysis was performed to determine the downstream signaling pathways. RESULTS: We found that genistein, daidzein and ERB-041 significantly inhibited ovarian cancer cell migration, invasion, proliferation, as well as induced cell cycle arrest and apoptosis. Significantly inhibitory effect on the size and number of sphere formed in genistein, daidzein and ERB-041 treated cells was also demonstrated. Moreover, genistein, daidzein and ERB-041 treatment reduced p-FAK, p-PI3K, p-AKT, p-GSK3ß, p21 or cyclin D1 expression in ovarian cancer cells. CONCLUSION: Genistein, daidzein and ERB-041 decreased ovarian cancer cell migration, invasion, proliferation and sphere formation, and induced cell cycle arrest and apoptosis with altered FAK and PI3K/AKT/GSK signaling and p21/cyclin D1 expression, suggesting their roles on ovarian cancer cell metastasis, tumorigenesis and stem-like properties and their potential as alternative therapies for ovarian cancer patients.

6.
BMC Cancer ; 17(1): 606, 2017 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-28859612

RESUMEN

BACKGROUND: Due to the presence of both classical estrogen receptor (ERα) and another ER subtype (ERß) in ovarian cancer, hormonal treatment is an attractive option. However, response to tamoxifen in ovarian cancer is modest. The presence of ERß variants further complicated the issue. We have recently shown that specifically targeting ER subtypes using selective ER modulators showed opposing functions of ER subtypes on cell growth. In the present study, the clinical significance of ERα and ERß variants (ß1, ß2 and ß5) and the functional effects of ERß2 and ERß5 in ovarian cancer was investigated. METHODS: ERα, ERß1, ERß2 and ERß5 expression were evaluated by immunohistochemistry in 106 ovarian cancer tissues. The association between ERs expression and clinicopathological parameters or prognosis was analyzed. Ectopic expression of ERß2 and ERß5 followed by functional assays were performed in ovarian cancer cell lines in order to detect their effects on cell invasion and proliferation. RESULTS: We found significantly higher nuclear (n)ERα and nERß5 and lower cytoplasmic (c)ERα expression in advanced cancers. Significantly lower ERß1 expression was also detected in high grade cancers. Significant loss of nERα and cERß2 expression were observed in clear cell histological subtypes. Higher nERß5 and lower cERß5 expression were associated with serous/clear cell subtypes, poor disease-free and overall survival. Positive cERα and higher cERß1 expression were significantly associated with better disease-free and overall survival. Furthermore, we found nERß5 as an independent prognostic factor for overall survival. Functionally, overexpression of ERß5 enhanced ovarian cancer cell migration, invasion and proliferation via FAK/c-Src activation whereas ERß2 induced cell migration and invasion. CONCLUSIONS: Since tamoxifen binds to both ERα and ERß1 which appear to bear opposing oncogenic roles, the histotypes-specific expression pattern of ERs indicates that personalized treatment for women based on ERs expression using selective estrogen receptor modulators may improve response rate. This study also suggests nERß5 as a potential prognostic marker and therapeutic target in ovarian cancer.


Asunto(s)
Proliferación Celular , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas/metabolismo , Adulto , Anciano , Línea Celular Tumoral , Receptor beta de Estrógeno/metabolismo , Femenino , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Invasividad Neoplásica , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/patología , Pronóstico , Isoformas de Proteínas/genética
7.
Am J Pathol ; 185(7): 2038-48, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26093985

RESUMEN

Human placental trophoblasts can be considered pseudomalignant, with tightly controlled proliferation, apoptosis, and invasiveness. Gestational trophoblastic disease (GTD) represents a family of heterogeneous trophoblastic lesions with aberrant apoptotic and proliferative activities and dysregulation of cell signaling pathways. We characterize the oncogenic effects of factor that binds to the inducer of short transcripts of HIV-1 [FBI-1, alias POZ and Krüppel erythroid myeloid ontogenic factor (POKEMON)/ZBTB7A] in GTD and its role in promoting cell aggressiveness in vitro and tumor growth in vivo. IHC studies showed increased nuclear expression of FBI-1, including hydatidiform moles, choriocarcinoma (CCA), and placental site trophoblastic tumor, in GTD. In JAR and JEG-3 CCA cells, ectopic FBI-1 expression opposed apoptosis through repression of proapoptotic genes (eg, BAK1, FAS, and CASP8). FBI-1 overexpression also promoted Akt activation, as indicated by Akt-pS473 phosphorylation. FBI-1 overexpression promoted mobility and invasiveness of JEG-3 and JAR, but not in the presence of the phosphoinositide 3-kinase inhibitor LY294002. These findings suggest that FBI-1 could promote cell migration and invasion via phosphoinositide 3-kinase/Akt signaling. In vivo, nude mice injected with CCA cells with stable FBI-1 knockdown demonstrated reduced tumor growth compared with that in control groups. These findings suggest that FBI-1 is clinically associated with the progression of, and may be a therapeutic target in, GTD, owing to its diverse oncogenic effects on dysregulated trophoblasts.


Asunto(s)
Coriocarcinoma/patología , Proteínas de Unión al ADN/genética , Enfermedad Trofoblástica Gestacional/patología , Proteína Oncogénica v-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Animales , Anticuerpos , Apoptosis , Pruebas de Carcinogenicidad , Movimiento Celular , Coriocarcinoma/genética , Coriocarcinoma/metabolismo , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Enfermedad Trofoblástica Gestacional/genética , Enfermedad Trofoblástica Gestacional/metabolismo , Humanos , Mola Hidatiforme/genética , Mola Hidatiforme/metabolismo , Mola Hidatiforme/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteína Oncogénica v-akt/genética , Fosfatidilinositol 3-Quinasas/genética , Placenta/metabolismo , Embarazo , Conejos , Factores de Transcripción/metabolismo , Trofoblastos/metabolismo
8.
Carcinogenesis ; 34(9): 2170-7, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23671128

RESUMEN

Gestational choriocarcinoma is a malignant tumor derived from placental trophoblast and the most aggressive member of gestational trophoblastic disease (GTD). Apoptosis-stimulating protein of p53-2 (ASPP2) is a member of ASPP family that transactivates p53 and thereby functions as a tumor suppressor. In this study, the expression profile of ASPP2 in choriocarcinoma was examined in comparison with normal placentas and hydatidiform moles, the latter being a type of GTD that carries malignant potential. Downregulation of ASPP2 messenger RNA and protein was demonstrated in choriocarcinoma by quantitative PCR and immunohistochemistry. ASPP2-transfected choriocarcinoma cells (JEG-3 and JAR) showed an increase in apoptosis and a decrease in cell migration as detected by TdT-mediated dUTP nick end labeling and wound healing assays, respectively, illustrating the complex action of ASPP2 on cell functions other than programmed cell death. Activated Src is known to be important in tumor progression. Transfection of ASPP2 but not ASPP1, another tumor-suppressive ASPP, was found to be related to subsequent decreased Src-pY416 phosphorylation, suggesting an inactivating effect of ASPP2 on Src. Moreover, this ASPP2-mediated inactivation of Src could be abolished by RNA interference with C-terminal Src kinase (Csk), a kinase that can inhibit Src activation. Our findings suggested that the ability of ASPP2 to attenuate Src activation was specific to ASPP2 in a Csk-dependent manner. Taken together, we demonstrated a loss of tumor-suppressive ASPP2 in choriocarcinoma with effects on cell migration and apoptosis. We also unveiled a possible mechanistic link between ASPP2 and Csk/Src signaling pathway, implicating the multiple cellular functions of ASPP2.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Movimiento Celular/genética , Coriocarcinoma/genética , Enfermedad Trofoblástica Gestacional/genética , Familia-src Quinasas/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína Tirosina Quinasa CSK , Línea Celular Tumoral , Coriocarcinoma/patología , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Enfermedad Trofoblástica Gestacional/patología , Humanos , Embarazo , Interferencia de ARN , Transducción de Señal , Familia-src Quinasas/metabolismo
9.
Proc Natl Acad Sci U S A ; 107(43): 18622-7, 2010 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-20926745

RESUMEN

Ovarian cancer is a lethal gynecological malignancy, and to improve survival, it is important to identify novel prognostic and therapeutic targets. In this study, we present a role for p21-activated kinase 4 (Pak4) in ovarian cancer progression. We show a significant association between increased expression of Pak4 and its activated form, phosphorylated (p)-Pak4 Ser(474), with metastasis of ovarian cancers, shorter overall and disease-free survival, advanced stage and high-grade cancers, serous/clear cell histological subtypes, and reduced chemosensitivity. Pak4 overexpression was also observed in ovarian cancer cell lines. Pak4 and p-Pak4 expression were detected both in the nucleus and cytoplasm of ovarian cancer cells, in vitro as well as in vivo. Stable knockdown of Pak4 in ovarian cancer cell lines led to reduced cell migration, invasion, and proliferation, along with reduced c-Src, ERK1/2, and epidermal growth factor receptor (EGFR) activation and decreased matrix metalloproteinase 2 (MMP2) expression. Conversely, Pak4 overexpression promoted ovarian cancer cell migration and invasion in a c-Src, MEK-1, MMP2, and kinase-dependent manner, and induced cell proliferation through the Pak4/c-Src/EGFR pathway that controls cyclin D1 and CDC25A expression. Stable knockdown of Pak4 also impeded tumor growth and dissemination in nude mice. This report reveals the association between Pak4 and important clinicopathologic parameters, suggesting Pak4 to be a significant prognostic marker and potential therapeutic molecular target in ovarian cancer. The implied possible cross-talk between Pak4 and EGFR suggests the potential of dual targeting of EGFR and Pak4 as a unique therapeutic approach for cancer therapy.


Asunto(s)
Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Quinasas p21 Activadas/fisiología , Adulto , Anciano , Animales , Secuencia de Bases , Línea Celular Tumoral , Movimiento Celular , Núcleo Celular/enzimología , Proliferación Celular , Citoplasma/enzimología , Cartilla de ADN/genética , Activación Enzimática , Receptores ErbB/fisiología , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Neoplasias Ováricas/genética , Pronóstico , Proteínas Proto-Oncogénicas pp60(c-src)/fisiología , Transducción de Señal , Quinasas p21 Activadas/antagonistas & inhibidores , Quinasas p21 Activadas/genética
10.
Adv Exp Med Biol ; 763: 186-92, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23397625

RESUMEN

The blood-follicle barrier (BFB) is one of the blood-tissue barriers in mammalian body found in developing follicles in the ovary. The BFB, besides the tight junction (TJ)-permeability barrier of the endothelial cells in the microvessels that surround the developing follicle, is constituted and contributed significantly by the basement membrane of the developing follicle which alters its composition rapidly during follicle development. While the concept of the BFB and its ultrastructure were described more than six decades ago, fewer than 20 reports are found in the literature that were dedicated to investigate the biology, regulation, and function of the BFB either in health or in disease. Furthermore, detailed analysis of the adhesion protein complexes and the regulation of the junction dynamics at the BFB are still missing in the literature. The goal of this short chapter is to provide an update on this important blood-tissue barrier, it is obvious that future investigation is much needed in the field to understand this ultrastructure better in order to treat and better ovarian disorders including ovarian cancer.


Asunto(s)
Folículo Ovárico/patología , Folículo Ovárico/fisiología , Neoplasias Ováricas/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Animales , Membrana Basal/irrigación sanguínea , Membrana Basal/metabolismo , Transporte Biológico , Permeabilidad de la Membrana Celular , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Femenino , Líquido Folicular/metabolismo , Humanos , Inmunoquímica , Folículo Ovárico/irrigación sanguínea , Neoplasias Ováricas/patología , Síndrome del Ovario Poliquístico/irrigación sanguínea , Síndrome del Ovario Poliquístico/patología , Uniones Estrechas/patología
11.
Front Oncol ; 12: 748403, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35155224

RESUMEN

Glycolysis has been reported to be critical for cancer stem cells (CSCs), which are associated with tumor chemoresistance, metastasis and recurrence. Thus, selectively targeting glycolytic enzymes may be a potential therapy for ovarian cancer. 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), the main source of fructose-2,6-bisphosphate, controls the first committed step in glycolysis. We investigate the clinical significance and roles of PFKFB3 in ovarian cancer using in vitro and in vivo experiments. We demonstrate that PFKFB3 is widely overexpressed in ovarian cancer and correlates with advanced stage/grade and poor outcomes. Significant up-regulation of PFKFB3 was found in ascites and metastatic foci, as well as CSC-enriched tumorspheres and ALDH+CD44+ cells. 3PO, a PFKFB3 inhibitor, reduced lactate level and sensitized A2780CP cells to cisplatin treatment, along with the modulation of inhibitors of apoptosis proteins (c-IAP1, c-IAP2 and survivin) and an immune modulator CD70. Blockade of PFKFB3 by siRNA approach in the CSC-enriched subset led to decreases in glycolysis and CSC properties, and activation of the NF-κB cascade. PFK158, another potent inhibitor of PFKFB3, impaired the stemness of ALDH+CD44+ cells in vitro and in vivo, whereas ectopic expression of PFKFB3 had the opposite results. Overall, PFKFB3 was found to mediate metabolic reprogramming, chemoresistance, metastasis and stemness in ovarian cancer, possibly via the modulation of inhibitors of apoptosis proteins and the NF-κB signaling pathway; thus, suggesting that PFKFB3 may be a potential therapeutic target for ovarian cancer.

12.
Carcinogenesis ; 32(5): 765-71, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21325635

RESUMEN

Gestational trophoblastic disease (GTD) includes frankly malignant choriocarcinoma (CCA) and placental site trophoblastic tumor and potentially malignant hydatidiform mole. p21-Activated kinase (PAK) 4 promotes cell motility. This study investigated the role of PAK4 in the pathogenesis of GTD. PAK4 messenger RNA and protein expressions in clinical samples and cell lines of normal placentas and GTD were determined by quantitative real-time polymerase chain reaction and western blot, respectively. The effects of human chorionic gonadotropin (hCG) and phosphoinositide 3 kinase (PI3K) on the expression and activation of PAK4 were investigated by treating CCA JEG3 and JAR cells with anti-hCG antibody and PI3K inhibitor, respectively. The effects of PAK4 on CCA cell proliferation, migration and invasion were assessed by corresponding functional assays. We demonstrated overexpression of PAK4 in GTD and CCA cell lines at both RNA and protein level. hCG is one of the upstream regulators of PAK4 expression, whereas activation of PAK4 is PI3K/PKB dependent in JEG3 and JAR cells. Significant correlation was found between PAK4 expression and proliferation index minichromosome maintenance complex component 7 (P = 0.007). In JEG3 and JAR cells, stably transfected PAK4 increased proliferation, migration and invasion, whereas small interfering RNA knockdown of PAK4 decreased proliferation, migration and invasion along with downregulated CDK6 and membrane-type 1 matrix metalloproteinase (MT1-MMP) and upregulated p16. We further found PAK4-mediated transcription of MT1-MMP in CCA cells by luciferase reporter assay. Our results demonstrated for the first time that overexpressed PAK4 was involved in the pathogenesis of GTD, promoting proliferation and enhancing cell migration and invasion in CCA cells.


Asunto(s)
Movimiento Celular , Proliferación Celular , Coriocarcinoma/patología , Neoplasias Uterinas/patología , Quinasas p21 Activadas/metabolismo , 1-Fosfatidilinositol 4-Quinasa/farmacología , Apoptosis , Western Blotting , Adhesión Celular , Línea Celular Tumoral , Coriocarcinoma/genética , Coriocarcinoma/metabolismo , Gonadotropina Coriónica/farmacología , Femenino , Enfermedad Trofoblástica Gestacional , Humanos , Técnicas para Inmunoenzimas , Metaloproteinasa 14 de la Matriz/genética , Metaloproteinasa 14 de la Matriz/metabolismo , Embarazo , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Trofoblásticas/genética , Neoplasias Trofoblásticas/metabolismo , Neoplasias Trofoblásticas/patología , Neoplasias Uterinas/genética , Neoplasias Uterinas/metabolismo , Quinasas p21 Activadas/antagonistas & inhibidores , Quinasas p21 Activadas/genética
13.
Mod Pathol ; 24(4): 522-32, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21102414

RESUMEN

Gestational trophoblastic disease encompasses a spectrum of trophoblastic lesions including true neoplasms such as choriocarcinomas and the potentially malignant hydatidiform moles, which may develop persistent disease requiring chemotherapy. ASPP1, a member of apoptosis-stimulating proteins of p53 (ASPPs), is a proapoptotic protein that can stimulate apoptosis through its interaction with p53. We evaluated the promoter methylation and expression profiles of ASPP1 in different trophoblastic tissues and its in vitro functional effect on two choriocarcinoma cell lines, namely JEG-3 and JAR. Significant downregulation of ASPP1 mRNA and protein levels was demonstrated in hydatidiform moles and choriocarcinomas, when compared with normal placentas by quantitative-PCR and immunohistochemistry. The ASPP1 mRNA level was significantly correlated with its hypermethylation status, evaluated with methylation-specific PCR, in placenta and gestational trophoblastic disease samples (P=0.024). Moreover, lower ASPP1 immunoreactivity was shown in hydatidiform moles that progressed to persistent gestational trophoblastic neoplasms than in those that regressed (P=0.045). A significant correlation was also found between expression of ASPP1 and proliferative indices (assessed by Ki67 and MCM7), apoptotic activity (M30 CytoDeath antibody), p53 and caspase-8 immunoreactivities. An in vitro study showed that ectopic expression of ASPP1 could trigger apoptosis through intrinsic and extrinsic pathways as indicated by an increase in cleaved caspase-9 and Fas ligand protein expression. The latter suggests a hitherto unreported novel link between ASPP1 and the extrinsic pathway of apoptosis. Our findings suggest that downregulation of ASPP1 by hypermethylation may be involved in the pathogenesis and progress of gestational trophoblastic disease, probably through its effect on apoptosis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis , Biomarcadores de Tumor/metabolismo , Coriocarcinoma/metabolismo , Metilación de ADN , Mola Hidatiforme/metabolismo , Placenta/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Reguladoras de la Apoptosis/genética , Biomarcadores de Tumor/genética , Western Blotting , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular , Distribución de Chi-Cuadrado , Coriocarcinoma/genética , Coriocarcinoma/patología , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Enfermedad Trofoblástica Gestacional , Hong Kong , Humanos , Mola Hidatiforme/genética , Mola Hidatiforme/patología , Inmunohistoquímica , Placenta/patología , Embarazo , Pronóstico , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Neoplasias Trofoblásticas/genética , Neoplasias Trofoblásticas/metabolismo , Neoplasias Trofoblásticas/patología
14.
Am J Pathol ; 176(6): 3015-22, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20413688

RESUMEN

Gestational trophoblastic disease (GTD) includes hydatidiform mole (HM), which can develop persistent gestational trophoblastic neoplasia requiring chemotherapy; choriocarcinoma, which is a frankly malignant tumor; placental site trophoblastic tumor; and epithelioid trophoblastic tumor. p21-Activated kinases (PAKs) promote malignant tumor progression. Therefore, this study investigated PAK1, PAK2, and p-PAK2 Ser(20) in the pathogenesis of GTD. By real-time PCR, PAK1 mRNA was significantly higher in HMs, particularly metastatic HMs (P = 0.046) and HMs that developed persistent disease (P = 0.011), when compared with normal placentas. By immunohistochemistry, significantly increased cytoplasmic PAK1 immunoreactivity in cytotrophoblasts was also detected in HMs (P = 0.042) and choriocarcinomas (P = 0.003). In addition, HMs that developed persistent disease displayed higher PAK1 immunoreactivity than those that regressed (P = 0.016), and elevated PAK1 immunoreactivity was observed in placental site trophoblastic tumors. Indeed, there was significant positive correlation between PAK1 expression and the proliferative indices Ki-67 (P = 0.016) and MCM7 (P = 0.026). Moreover, higher PAK1 mRNA and protein expression was confirmed in the choriocarcinoma cell-lines JEG-3 and JAR; however, PAK2 mRNA and p-PAK2 immunoreactivity showed a similar expression pattern in normal first trimester placentas and GTD. Knockdown of PAK1 in JEG-3 and JAR reduced cell proliferation, migration, and invasion ability, up-regulated p16, and down-regulated vascular endothelial growth factor and MT1-MMP expression. This is the first report revealing the involvement of PAK1 in the pathogenesis and clinical progress of GTD.


Asunto(s)
Proliferación Celular , Enfermedad Trofoblástica Gestacional , Isoenzimas/metabolismo , Quinasas p21 Activadas/metabolismo , Animales , Línea Celular , Movimiento Celular/fisiología , Femenino , Técnicas de Silenciamiento del Gen , Edad Gestacional , Enfermedad Trofoblástica Gestacional/metabolismo , Enfermedad Trofoblástica Gestacional/patología , Humanos , Isoenzimas/genética , Placenta/citología , Placenta/metabolismo , Placenta/patología , Embarazo , Quinasas p21 Activadas/genética
15.
Histopathology ; 59(6): 1163-72, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22175896

RESUMEN

AIMS: Dedicator of cytokinesis I (Dock180) is a novel guanine nucleotide exchange factor for Rho guanosine triphosphates (GTPases) important for cell migration. The aim of this study was to evaluate the role of Dock180 in ovarian carcinogenesis. METHODS AND RESULTS: Using immunohistochemistry, real-time polymerase chain reaction and Western blotting, overexpression of Dock180 RNA and protein was demonstrated in the nucleus and cytoplasm of ovarian cancer cell lines (n = 5) and clinical samples of ovarian borderline tumours (n = 21) and invasive cancers (n = 108) when compared with ovarian epithelial cell lines (n = 3) and benign cystadenomas (n = 10) (P < 0.05). High Dock180 cytoplasmic expression in ovarian cancer (n = 108) was associated significantly with serous histological type, high-grade cancer and advanced stage (P < 0.05), as well as poor overall and disease-free survival (P = 0.004). Using multivariate progression analysis, high Dock180 cytoplasmic expression and advanced cancer stage were found to be independent prognostic factors for short overall survival and disease-free survival (P < 0.05). Exogenous expression of Dock180 by transient transfection enhanced cancer cell migration and invasion, whereas knockdown of Dock180 by an siRNA approach retarded cancer cell migration and invasion in association with down-regulation of matrix metalloproteinase 2. CONCLUSIONS: Our findings suggest that Dock180 contributes to ovarian carcinogenesis and dissemination and is a potential prognostic marker and therapeutic target.


Asunto(s)
Carcinoma/enzimología , Cistoadenoma/enzimología , Neoplasias Ováricas/enzimología , Proteínas de Unión al GTP rac/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Western Blotting , Carcinoma/mortalidad , Carcinoma/patología , Movimiento Celular/genética , Cistoadenoma/mortalidad , Cistoadenoma/patología , Femenino , Humanos , Inmunohistoquímica , Microscopía Confocal , Persona de Mediana Edad , Clasificación del Tumor , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/patología , Fenotipo , Pronóstico , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Regulación hacia Arriba , Adulto Joven , Proteínas de Unión al GTP rac/análisis
16.
Front Oncol ; 11: 738177, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34820325

RESUMEN

Cholesterol is an essential substance in mammalian cells, and cholesterol metabolism plays crucial roles in multiple biological functions. Dysregulated cholesterol metabolism is a metabolic hallmark in several cancers, beyond the Warburg effect. Reprogrammed cholesterol metabolism has been reported to enhance tumorigenesis, metastasis and chemoresistance in multiple cancer types, including ovarian cancer. Ovarian cancer is one of the most aggressive malignancies worldwide. Alterations in metabolic pathways are characteristic features of ovarian cancer; however, the specific role of cholesterol metabolism remains to be established. In this report, we provide an overview of the key proteins involved in cholesterol metabolism in ovarian cancer, including the rate-limiting enzymes in cholesterol biosynthesis, and the proteins involved in cholesterol uptake, storage and trafficking. Also, we review the roles of cholesterol and its derivatives in ovarian cancer and the tumor microenvironment, and discuss promising related therapeutic targets for ovarian cancer.

17.
Cancers (Basel) ; 13(14)2021 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-34298618

RESUMEN

In epithelial ovarian cancer (EOC), carboplatin/cisplatin-induced chemoresistance is a major hurdle to successful treatment. Aerobic glycolysis is a common characteristic of cancer. However, the role of glycolytic metabolism in chemoresistance and its impact on clinical outcomes in EOC are not clear. Here, we show a functional interaction between the key glycolytic enzyme hexokinase II (HKII) and activated P-p53 (Ser15) in the regulation of bioenergetics and chemosensitivity. Using translational approaches with proximity ligation assessment in cancer cells and human EOC tumor sections, we showed that nuclear HKII-P-p53 (Ser15) interaction is increased after chemotherapy, and functions as a determinant of chemoresponsiveness as a prognostic biomarker. We also demonstrated that p53 is required for the intracellular nuclear HKII trafficking in the control of glycolysis in EOC, associated with chemosensitivity. Mechanistically, cisplatin-induced P-p53 (Ser15) recruits HKII and apoptosis-inducing factor (AIF) in chemosensitive EOC cells, enabling their translocation from the mitochondria to the nucleus, eliciting AIF-induced apoptosis. Conversely, in p53-defective chemoresistant EOC cells, HKII and AIF are strongly bound in the mitochondria and, therefore, apoptosis is suppressed. Collectively, our findings implicate nuclear HKII-P-p53(Ser15) interaction in chemosensitivity and could provide an effective clinical strategy as a promising biomarker during platinum-based therapy.

18.
Mol Cancer ; 9: 318, 2010 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-21176152

RESUMEN

BACKGROUND: FBI-1 (factor that binds to the inducer of short transcripts of human immunodeficiency virus-1) is a member of the POK (POZ and Kruppel) family of transcription factors and play important roles in cellular differentiation and oncogenesis. Recent evidence suggests that FBI-1 is expressed at high levels in a subset of human lymphomas and some epithelial solid tumors. However, the function of FBI-1 in human ovarian cancers remains elusive. RESULTS: In this study, we investigated the role of FBI-1 in human ovarian cancers, in particularly, its function in cancer cell invasion via modulating membrane type 1-matrix metalloproteinase (MT1-MMP). Significantly higher FBI-1 protein and mRNA expression levels were demonstrated in ovarian cancers samples and cell lines compared with borderline tumors and benign cystadenomas. Increased FBI-1 mRNA expression was correlated significantly with gene amplification (P = 0.037). Moreover, higher FBI-1 expression was found in metastatic foci (P = 0.036) and malignant ascites (P = 0.021), and was significantly associated with advanced stage (P = 0.012), shorter overall survival (P = 0.032) and disease-free survival (P = 0.016). In vitro, overexpressed FBI-1 significantly enhanced cell migration and invasion both in OVCA 420 and SKOV-3 ovarian carcinoma cells, irrespective of p53 status, accompanied with elevated expression of MT1-MMP, but not MMP-2 or TIMP-2. Moreover, knockdown of MT1-MMP abolished FBI-1-mediated cell migration and invasion. Conversely, stable knockdown of FBI-1 remarkably reduced the motility of these cells with decreased expression of MT1-MMP. Promoter assay and chromatin immunoprecipitation study indicated that FBI-1 could directly interact with the promoter spanning ~600 bp of the 5'-flanking sequence of MT1-MMP and enhanced its expression in a dose-dependent manner. Furthermore, stable knockdown and ectopic expression of FBI-1 decreased and increased cell proliferation respectively in OVCA 420, but not in the p53 null SKOV-3 cells. CONCLUSIONS: Our results suggested an important role of FBI-1 in ovarian cancer cell proliferation, cell mobility, and invasiveness, and that FBI-1 can be a potential target of chemotherapy.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Factores de Transcripción/metabolismo , Adulto , Anciano , Western Blotting , Línea Celular , Línea Celular Tumoral , Movimiento Celular/genética , Movimiento Celular/fisiología , Proliferación Celular , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Femenino , Humanos , Inmunohistoquímica , Técnicas In Vitro , Metaloproteinasa 14 de la Matriz/genética , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Unión Proteica , Proto-Oncogenes Mas , Factores de Transcripción/genética , Adulto Joven
19.
Int J Cancer ; 127(1): 21-31, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19876919

RESUMEN

Ovarian cancer is a gynecological malignancy with high mortality. Therefore, the identification of novel prognostic and therapeutic targets is important. p21-activated kinases (Paks) are involved in cytoskeleton reorganization. This study investigated the clinical significance of total and phosphorylated (p) Pak1 and Pak2 as well as their functional roles in ovarian cancer. Expressions of Pak1, p-Pak1 Thr(212), Pak2 and p-Pak2 Ser(20) in ovarian normal and cancerous cell lines as well as in clinical samples of ovarian tumors were evaluated. The effects of Pak1 and Pak2 on ovarian cancer cell functions were determined. Pak1, p-Pak1 and p-Pak2 were overexpressed in ovarian cancer cell lines, and clinical samples of ovarian cancers were compared with benign ovarian lesions/inclusion cysts. Similar Pak2 expression levels were observed among normal and cancerous cell lines and clinical samples. After multiple testing correction, high Pak1 and nuclear p-Pak1 expression in ovarian cancers was significantly associated with histological type and tumor grade, respectively. Pak1 and p-Pak1 expression was associated with poor overall and disease-free survival. Pak1 was an independent prognostic factor. Knockdown of Pak1 and Pak2 in ovarian cancer cell lines reduced cell migration and invasion but did not affect cell proliferation and apoptosis. Knockdown of Pak1 also reduced p38 activation and downregulated vascular endothelial growth factor. Conversely, ectopic Pak1 overexpression enhanced ovarian cancer cell migration and invasion in a kinase-dependent manner, along with increased p38 activation. Our findings suggest that Pak1, p-Pak1 and p-Pak2 play important roles in ovarian carcinogenesis. Pak1 and p-Pak1 may be potential prognostic markers and therapeutic molecular targets in ovarian cancer.


Asunto(s)
Neoplasias Ováricas/metabolismo , Quinasas p21 Activadas/metabolismo , Apoptosis , Secuencia de Bases , Western Blotting , Línea Celular Transformada , Proliferación Celular , Cartilla de ADN , Femenino , Humanos , Inmunohistoquímica , Invasividad Neoplásica , Neoplasias Ováricas/patología , Fosforilación , Reacción en Cadena de la Polimerasa , Pronóstico , Quinasas p21 Activadas/genética
20.
Oncogenesis ; 9(2): 24, 2020 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-32071289

RESUMEN

Ovarian cancer is the most lethal gynecological malignancies owing to the lack of definitive symptoms until development of widespread metastases. Identification of novel prognostic and therapeutic targets is therefore an urgent need to improve survival. Here, we demonstrated high expression of the mitochondrial gatekeeping enzyme, pyruvate dehydrogenase kinase 1 (PDK1), in both clinical samples and cell lines of ovarian cancer. PDK1 expression was significantly associated with metastasis, reduced chemosensitivity, and poor overall and disease-free survival, and further highlighted as an independent prognostic factor. Silencing of PDK1 retarded lactate production, ovarian cancer cell adhesion, migration, invasion, and angiogenesis, and consequently metastasis, concomitant with decreased α5ß1 integrin expression. Phospho-kinase array profiling and RNA sequencing analyses further revealed reduction of JNK activation and IL-8 expression in PDK1-depleted cells. Conversely, PDK1 overexpression promoted cell adhesion via modulation of α5ß1 integrins, along with cell migration, invasion, and angiogenesis through activation of JNK/IL-8 signaling. PDK1 depletion additionally hindered tumor growth and dissemination in nude mice in vivo. Importantly, PDK1 levels were upregulated upon treatment with conditioned medium from omental tissues, which in turn promoted metastasis. Our findings suggest that PDK1, which is regulated by the tumor microenvironment, controls lactate production and promotes ovarian cancer cell metastasis via modulation of α5ß1 integrin and JNK/IL-8 signaling. To our knowledge, this is the first report to demonstrate an association between PDK1 and survival in patients with ovarian cancer, supporting its efficacy as a valuable prognostic marker and therapeutic molecular target for the disease.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA