Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
2.
J Cell Mol Med ; 26(1): 123-132, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34894055

RESUMEN

In normal pregnancy, hepatic metabolism adaptation occurs with an increase in lipid biosynthesis. Placental shedding of syncytiotrophoblast-derived extracellular vesicles (STBEVs) into the maternal circulation constitutes a major signalling mechanism between foetus and mother. We investigated whether STBEVs from normal pregnant women might target liver cells in vitro and induce changes in lipid synthesis. This study was performed at the Nuffield Department of Women's & Reproductive Health, Oxford, UK. STBEVs were obtained by dual-lobe placental perfusion from 11 normal pregnancies at term. Medium/large and small STBEVs were collected by ultracentrifugation at 10,000g and 150,000g, respectively. STBEVs were analysed by Western blot analysis and flow cytometry for co-expression of apolipoprotein-E (apoE) and placental alkaline phosphatase (PLAP). The uptake of STBEVs by liver cells and the effect on lipid metabolism was evaluated using a hepatocarcinoma cell line (HepG2 cells). Data were analysed by one-way ANOVA and Student's t test. We demonstrated that: (a) STBEVs carry apoE; (b) HepG2 cells take up STBEVs through an apoE-LDL receptor interaction; (c) STBEV incorporation into HepG2 cells resulted in (i) increased cholesterol release (ELISA); (ii) increased expression of the genes SQLE and FDPS (microarray) involved in cholesterol biosynthesis; (iii) downregulation of the CLOCK gene (microarray and PCR), involved in the circadian negative control of lipid synthesis in liver cells. In conclusion, the placenta may orchestrate the metabolic adaptation of the maternal liver through release of apoE-positive STBEVs, by increasing lipid synthesis in a circadian-independent fashion, meeting the nutritional needs of the growing foetus.


Asunto(s)
Vesículas Extracelulares , Trofoblastos , Apolipoproteínas/metabolismo , Apolipoproteínas E/metabolismo , Vesículas Extracelulares/metabolismo , Femenino , Humanos , Lípidos , Hígado , Placenta/metabolismo , Embarazo , Trofoblastos/metabolismo
3.
Nat Commun ; 9(1): 476, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29396402

RESUMEN

Though used widely in cancer therapy, paclitaxel only elicits a response in a fraction of patients. A strong determinant of paclitaxel tumor response is the state of microtubule dynamic instability. However, whether the manipulation of this physiological process can be controlled to enhance paclitaxel response has not been tested. Here, we show a previously unrecognized role of the microtubule-associated protein CRMP2 in inducing microtubule bundling through its carboxy terminus. This activity is significantly decreased when the FER tyrosine kinase phosphorylates CRMP2 at Y479 and Y499. The crystal structures of wild-type CRMP2 and CRMP2-Y479E reveal how mimicking phosphorylation prevents tetramerization of CRMP2. Depletion of FER or reducing its catalytic activity using sub-therapeutic doses of inhibitors increases paclitaxel-induced microtubule stability and cytotoxicity in ovarian cancer cells and in vivo. This work provides a rationale for inhibiting FER-mediated CRMP2 phosphorylation to enhance paclitaxel on-target activity for cancer therapy.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Paclitaxel/farmacología , Proteínas Tirosina Quinasas/genética , Tratamiento con ARN de Interferencia , Moduladores de Tubulina/farmacología , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Desnudos , Microscopía Confocal , Microscopía Fluorescente , Microtúbulos/efectos de los fármacos , Microtúbulos/ultraestructura , Simulación de Dinámica Molecular , Terapia Molecular Dirigida , Trasplante de Neoplasias , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/ultraestructura , Fosforilación/efectos de los fármacos , Fosforilación/genética , Multimerización de Proteína/efectos de los fármacos , Multimerización de Proteína/genética , Proteínas Tirosina Quinasas/metabolismo , ARN Interferente Pequeño
4.
Oncotarget ; 8(11): 17960-17980, 2017 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-28152500

RESUMEN

Taxanes represent some of the most commonly used chemotherapeutic agents for ovarian cancer treatment. However, they are only effective in approximately 40% of patients. Novel therapeutic strategies are required to potentiate their effect and improve patient outcome. A hallmark of many cancers is the constitutive activation of the PI3K/AKT pathway, which drives cell survival and metabolism. We discovered a striking decrease in AKT activity coupled with a significant reduction in glucose 6-phosphate and ATP levels during mitotic arrest in the majority of ovarian cancer cell lines tested, indicating a potential metabolic vulnerability. A high-content siRNA screen to detect novel metabolic targets in mitotically arrested ovarian cancer cells identified the glycolytic enzyme PFKFB4. PFKFB4 depletion increased caspase 3/7 activity, and levels of reactive oxygen species only in mitotically arrested cells, and significantly enhanced mitotic cell death after paclitaxel treatment. Depletion of PFKFB3 demonstrated a similar phenotype. The observation that some ovarian cancer cells lose AKT activity during mitotic arrest and become vulnerable to metabolic targeting is a new concept in cancer therapy. Thus, combining mitotic-targeted therapies with glycolytic inhibitors may act to potentiate the effects of antimitotics in ovarian cancer through mitosis-specific cell death.


Asunto(s)
Puntos de Control del Ciclo Celular/fisiología , Muerte Celular/fisiología , Neoplasias Ováricas/patología , Fosfofructoquinasa-2/metabolismo , Antineoplásicos/farmacología , Western Blotting , Línea Celular Tumoral , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Humanos , Mutagénesis Sitio-Dirigida , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Paclitaxel/farmacología , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas c-akt/metabolismo
6.
Cancer Cell ; 30(2): 273-289, 2016 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-27478041

RESUMEN

The adipocyte-rich microenvironment forms a niche for ovarian cancer metastasis, but the mechanisms driving this process are incompletely understood. Here we show that salt-inducible kinase 2 (SIK2) is overexpressed in adipocyte-rich metastatic deposits compared with ovarian primary lesions. Overexpression of SIK2 in ovarian cancer cells promotes abdominal metastasis while SIK2 depletion prevents metastasis in vivo. Importantly, adipocytes induce calcium-dependent activation and autophosphorylation of SIK2. Activated SIK2 plays a dual role in augmenting AMPK-induced phosphorylation of acetyl-CoA carboxylase and in activating the PI3K/AKT pathway through p85α-S154 phosphorylation. These findings identify SIK2 at the apex of the adipocyte-induced signaling cascades in cancer cells and make a compelling case for targeting SIK2 for therapy in ovarian cancer.


Asunto(s)
Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Adipocitos/enzimología , Adipocitos/metabolismo , Adipocitos/patología , Animales , Femenino , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Metástasis de la Neoplasia , Proteína Oncogénica v-akt/metabolismo , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal
7.
EBioMedicine ; 10: 137-49, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27492892

RESUMEN

Current screening methods for ovarian cancer can only detect advanced disease. Earlier detection has proved difficult because the molecular precursors involved in the natural history of the disease are unknown. To identify early driver mutations in ovarian cancer cells, we used dense whole genome sequencing of micrometastases and microscopic residual disease collected at three time points over three years from a single patient during treatment for high-grade serous ovarian cancer (HGSOC). The functional and clinical significance of the identified mutations was examined using a combination of population-based whole genome sequencing, targeted deep sequencing, multi-center analysis of protein expression, loss of function experiments in an in-vivo reporter assay and mammalian models, and gain of function experiments in primary cultured fallopian tube epithelial (FTE) cells. We identified frequent mutations involving a 40kb distal repressor region for the key stem cell differentiation gene SOX2. In the apparently normal FTE, the region was also mutated. This was associated with a profound increase in SOX2 expression (p<2(-16)), which was not found in patients without cancer (n=108). Importantly, we show that SOX2 overexpression in FTE is nearly ubiquitous in patients with HGSOCs (n=100), and common in BRCA1-BRCA2 mutation carriers (n=71) who underwent prophylactic salpingo-oophorectomy. We propose that the finding of SOX2 overexpression in FTE could be exploited to develop biomarkers for detecting disease at a premalignant stage, which would reduce mortality from this devastating disease.


Asunto(s)
Trompas Uterinas/metabolismo , Trompas Uterinas/patología , Expresión Génica , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/genética , Lesiones Precancerosas , Factores de Transcripción SOXB1/genética , Adulto , Anciano , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor , Diferenciación Celular/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Femenino , Genes BRCA1 , Genes BRCA2 , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Biopsia Guiada por Imagen , Laparoscopía , Persona de Mediana Edad , Modelos Biológicos , Mutación , Estadificación de Neoplasias , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neoplasias Ováricas/tratamiento farmacológico , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción SOXB1/metabolismo
8.
Mol Cell Biol ; 32(9): 1633-44, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22371483

RESUMEN

The TFAP2C transcription factor has been shown to downregulate transcription of the universal cell cycle inhibitor p21(cip) (CDKN1A). In examining the mechanism of TFAP2C-mediated repression, we have identified a ternary complex at the proximal promoter containing TFAP2C, the oncoprotein Myc, and the trimethylated lysine 4 of histone H3 (H3K4me3) demethylase, KDM5B. We demonstrated that while TFAP2C and Myc can downregulate the CDKN1A promoter independently, KDM5B acts as a corepressor dependent on the other two proteins. All three factors collaborate for optimal CDKN1A repression, which requires the AP-2 binding site at -111/-103 and KDM5B demethylase activity. Silencing of TFAP2C-KDM5B-Myc led to increased H3K4me3 at the endogenous promoter and full induction of CDKN1A expression. Coimmunoprecipitation assays showed that TFAP2C and Myc associate with distinct domains of KDM5B and the TFAP2C C-terminal 270 amino acids (aa) are required for Myc and KDM5B interaction. Overexpression of all three proteins resulted in forced S-phase entry and attenuation of checkpoint activation, even in the presence of chemotherapy drugs. Since each protein has been linked to poor prognosis in breast cancer, our findings suggest that the TFAP2C-Myc-KDM5B complex promotes cell cycle progression via direct CDKN1A repression, thereby contributing to tumorigenesis and therapy failure.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Genes myc , Histona Demetilasas con Dominio de Jumonji/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Factor de Transcripción AP-2/metabolismo , Complejo 2 de Proteína Adaptadora/genética , Complejo 2 de Proteína Adaptadora/metabolismo , Sitios de Unión , Ciclo Celular , Línea Celular Tumoral , Regulación hacia Abajo , Sitios Genéticos , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Represoras/genética , Factor de Transcripción AP-2/genética
9.
Mol Cancer Res ; 9(2): 225-33, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21325284

RESUMEN

Although innumerable investigations regarding the biology of lung cancer have been carried out, many aspects thereof remain to be addressed, including the role played by the retinoblastoma-related protein Rb2/p130 during the evolution of this disease. Here we report novel findings on the mechanisms that control Rb2/p130 gene expression in lung fibroblasts and characterize the effects of Rb2/p130 deregulation on the proliferative features of lung cancer cells. We revealed for the first time that in lung fibroblasts the expression of Rb2/p130 gene is directly controlled by the chromatin insulator CCCTC-binding factor, CTCF, which by binding to the Rb2/p130 gene promoter induces, and/or maintains, a specific local chromatin organization that in turn governs the transcriptional activity of Rb2/p130 gene. However, in lung cancer cells the activity of CTCF in controlling Rb2/p130 gene expression is impaired by BORIS, a CTCF-paralogue, which by binding to the Rb2/p130 gene could trigger changes in the chromatin asset established by CTCF, thereby affecting CTCF regulatory activity on Rb2/p130 transcription. These studies not only provide essential basic insights into the molecular mechanisms that control Rb2/p130 gene expression in lung cancer, but also offer a potential paradigm for the actions of other activators and/or corepressors, such as CTCF and BORIS, that could be crucial in explaining how alterations in the mechanism regulating Rb2/p130 gene expression may accelerate the progression of lung tumors, or favor the onset of recurrence after cancer treatment.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Proteínas Represoras/metabolismo , Proteína p130 Similar a la del Retinoblastoma/genética , Transcripción Genética , Sitios de Unión , Factor de Unión a CCCTC , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Pequeñas/genética , Carcinoma de Células Pequeñas/patología , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Posicionamiento de Cromosoma/genética , Fibroblastos/metabolismo , Humanos , Neoplasias Pulmonares/patología , Sistemas de Lectura Abierta/genética , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteína p130 Similar a la del Retinoblastoma/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA