Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Plants (Basel) ; 13(6)2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38592828

RESUMEN

Common bean (Phaseolus vulgaris L.) is the primary grain legume cultivated worldwide for direct human consumption due to the high nutritional value of its seeds and pods. The high protein content of common beans highlights it as the most promising source of plant-based protein for the food industry. Additionally, landraces of common bean have great variability in nutritional traits, which is necessary to increase the nutritional quality of elite varieties. Therefore, the main objective of this study was to nutritionally characterize 23 Chilean landraces and 5 commercial varieties of common bean to identify genotypes with high nutritional value that are promising for the food industry and for genetic improvement programs. The landrace Phv23 ('Palo') was the most outstanding with high concentrations of minerals such as P (7.53 g/kg), K (19.8 g/kg), Mg (2.43 g/kg), Zn (52.67 mg/kg), and Cu (13.67 mg/kg); essential amino acids (364.8 mg/g protein); and total proteins (30.35 g/100 g seed). Additionally, the landraces Phv9 ('Cimarrón'), Phv17 ('Juanita'), Phv3 ('Araucano'), Phv8 ('Cabrita/Señorita'), and Phv4 ('Arroz') had a high protein content. The landrace Phv24 ('Peumo') stood out for its phenolic compounds (TPC = 218.1 mg GA/100 g seed) and antioxidant activity (ORAC = 22,167.9 µmol eq trolox/100 g extract), but it has moderate to low mineral and protein concentrations. In general, the concentration of nutritional compounds in some Chilean landraces was significantly different from the commercial varieties, highlighting their high nutritional value and their potential use for the food industry and for genetic improvement purposes.

2.
Transl Res ; 175: 92-102.e2, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27083386

RESUMEN

Emerging evidence suggest that many high-grade serous "ovarian" cancers (HGSOC) start in the fallopian tube. Cancer cells are then recruited to the ovary and then spread diffusely through the abdomen. The mechanism of ovarian cancer spread was thought to be largely due to direct shedding of tumor cells into the peritoneal cavity with vascular spread being of limited importance. Recent work challenges this dogma, suggesting hematogenous spread of ovarian cancer may play a larger role in ovarian cancer cell metastasis than previously thought. One reason the role of vascular spread of ovarian cancer has not been fully elucidated is the lack of easily accessible models of vascular ovarian cancer metastasis. Here, we present 3 metastatic models of ovarian cancer which confirm the ability of ovarian cancer to hematogenously spread. Strikingly, we observe a high rate of metastasis to the ovary with the development of ascites in these models. Interestingly, oophorectomy resulted in a complete loss of peritoneal metastases and ascites. Taken together, our data indicate that hematogenously disseminated HGSOC cells have a unique tropism for the ovary and that hematogenous spread in ovarian cancer may be more common than appreciated. Furthermore, our studies support a critical role for the ovary in promoting HGSOC cell metastasis to the abdomen. The models developed here represent important new tools to evaluate both the mechanism of cancer cell recruitment to the ovary and understand and target key steps in ovarian cancer metastasis.


Asunto(s)
Abdomen/patología , Vasos Sanguíneos/patología , Modelos Biológicos , Neoplasias Ováricas/secundario , Ovario/patología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Inyecciones Intravenosas , Ratones , Neoplasias Ováricas/patología , Ovariectomía
3.
Mol Cancer Ther ; 14(7): 1717-27, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25969154

RESUMEN

Ovarian cancer is known to be composed of distinct populations of cancer cells, some of which demonstrate increased capacity for cancer initiation and/or metastasis. The study of human cancer cell populations is difficult due to long requirements for tumor growth, interpatient variability, and the need for tumor growth in immune-deficient mice. We therefore characterized the cancer initiation capacity of distinct cancer cell populations in a transgenic murine model of ovarian cancer. In this model, conditional deletion of Apc, Pten, and Trp53 in the ovarian surface epithelium (OSE) results in the generation of high-grade metastatic ovarian carcinomas. Cell lines derived from these murine tumors express numerous putative stem cell markers, including CD24, CD44, CD90, CD117, CD133, and ALDH. We show that CD24(+) and CD133(+) cells have increased tumor sphere-forming capacity. CD133(+) cells demonstrated a trend for increased tumor initiation while CD24(+) cells versus CD24(-) cells had significantly greater tumor initiation and tumor growth capacity. No preferential tumor-initiating or growth capacity was observed for CD44(+), CD90(+), CD117(+), or ALDH(+) versus their negative counterparts. We have found that CD24(+) cells, compared with CD24(-) cells, have increased phosphorylation of STAT3 and increased expression of STAT3 target Nanog and c-myc. JAK2 inhibition of STAT3 phosphorylation preferentially induced cytotoxicity in CD24(+) cells. In vivo JAK2 inhibitor therapy dramatically reduced tumor metastases, and prolonged overall survival. These findings indicate that CD24(+) cells play a role in tumor migration and metastasis and support JAK2 as a therapeutic target in ovarian cancer.


Asunto(s)
Antígeno CD24/genética , Janus Quinasa 2/genética , Células Madre Neoplásicas/metabolismo , Neoplasias Ováricas/genética , Proteína de la Poliposis Adenomatosa del Colon/deficiencia , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Western Blotting , Antígeno CD24/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Cisplatino/administración & dosificación , Cisplatino/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Janus Quinasa 2/metabolismo , Ratones , Ratones Noqueados , Metástasis de la Neoplasia , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/genética , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología , Análisis de Supervivencia , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética
4.
Cancer Res ; 73(12): 3555-65, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23576551

RESUMEN

Human tumor vessels express tumor vascular markers (TVM), proteins that are not expressed in normal blood vessels. Antibodies targeting TVMs could act as potent therapeutics. Unfortunately, preclinical in vivo studies testing anti-human TVM therapies have been difficult to do due to a lack of in vivo models with confirmed expression of human TVMs. We therefore evaluated TVM expression in a human embryonic stem cell-derived teratoma (hESCT) tumor model previously shown to have human vessels. We now report that in the presence of tumor cells, hESCT tumor vessels express human TVMs. The addition of mouse embryonic fibroblasts and human tumor endothelial cells significantly increases the number of human tumor vessels. TVM induction is mostly tumor-type-specific with ovarian cancer cells inducing primarily ovarian TVMs, whereas breast cancer cells induce breast cancer specific TVMs. We show the use of this model to test an anti-human specific TVM immunotherapeutics; anti-human Thy1 TVM immunotherapy results in central tumor necrosis and a three-fold reduction in human tumor vascular density. Finally, we tested the ability of the hESCT model, with human tumor vascular niche, to enhance the engraftment rate of primary human ovarian cancer stem-like cells (CSC). ALDH(+) CSC from patients (n = 6) engrafted in hESCT within 4 to 12 weeks whereas none engrafted in the flank. ALDH(-) ovarian cancer cells showed no engraftment in the hESCT or flank (n = 3). Thus, this model represents a useful tool to test anti-human TVM therapy and evaluate in vivo human CSC tumor biology.


Asunto(s)
Células Madre Neoplásicas/metabolismo , Neovascularización Patológica/metabolismo , Neoplasias Ováricas/metabolismo , Teratoma/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/inmunología , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Células Madre Embrionarias/metabolismo , Femenino , Humanos , Inmunohistoquímica , Inmunotoxinas/administración & dosificación , Inmunotoxinas/inmunología , Células MCF-7 , Ratones , Ratones Endogámicos NOD , Ratones SCID , Terapia Molecular Dirigida , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Teratoma/tratamiento farmacológico , Teratoma/patología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cancer Lett ; 322(1): 1-7, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22334034

RESUMEN

Cancer stem cells are rare chemotherapy resistant cells within a tumor which can serve to populate the bulk of a tumor with more differentiated daughter cells and potentially contribute to recurrent disease. Ovarian cancer is a disease for which at the time of initial treatment we can obtain complete clinical remission in the majority of patients. Unfortunately, most will relapse and succumb to their disease. This clinical course is in line with the cancer stem cell model. In the past 5 years a significant amount of work has been done to identify cells with characteristics of ovarian cancer stem cells. This review will focus specifically on the markers used to define human ovarian cancer stem cells, the prognostic implications of the expression of these cancer stem cell markers in patient's primary tumors, and the potential of these cancer stem cell markers to serve as therapeutic targets.


Asunto(s)
Células Madre Neoplásicas/química , Neoplasias Ováricas/patología , Antígeno AC133 , Aldehído Deshidrogenasa/análisis , Antígenos CD/análisis , Biomarcadores de Tumor/análisis , Antígeno CD24/análisis , Femenino , Glicoproteínas/análisis , Humanos , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias Ováricas/química , Neoplasias Ováricas/tratamiento farmacológico , Péptidos/análisis , Pronóstico , Proteínas Proto-Oncogénicas c-kit/análisis
6.
Cancer Res ; 70(21): 8674-83, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20959470

RESUMEN

Recent studies indicate that ovarian cancer may be highly responsive to antivascular therapeutics. We have developed an antivascular tumor therapeutic using the F3 peptide to target cisplatin-loaded nanoparticles (F3-Cis-Np) to tumor vessels. We show that although F3-Cis-Np bind with high specificity to both human ovarian tumor cells and tumor endothelial cells in vitro, they only show cytotoxic activity against the tumor endothelial cells. In vivo these nanoparticles bind primarily to tumor endothelial cells. Therapeutic studies in both flank and orthotopic i.p. murine ovarian tumor models, as well as human tumor xenograft models, show rapid tumor regression with treatment. Treatment was associated with significant vascular necrosis consistent with an antivascular effect. Furthermore, treatment was active in both platinum-sensitive and platinum-resistant cell lines. Importantly, we show that F3-Cis-Np bind to human tumor endothelial cells in vitro and to human tumor vessels in vivo. Therapy targeting human vasculature in vivo with F3-Cis-Np led to near complete loss of all human tumor vessels in a murine model of human tumor vasculature. Our studies indicate that F3-targeted vascular therapeutics may be an effective treatment modality in human ovarian cancer.


Asunto(s)
Cisplatino/uso terapéutico , Endotelio Vascular/efectos de los fármacos , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Nanopartículas , Neoplasias Ováricas/prevención & control , Fragmentos de Péptidos/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Femenino , Humanos , Técnicas para Inmunoenzimas , Inyecciones Intraperitoneales , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Neovascularización Patológica/prevención & control , Neoplasias Ováricas/irrigación sanguínea , Neoplasias Ováricas/patología , Tasa de Supervivencia , Teratoma/irrigación sanguínea , Teratoma/patología , Teratoma/prevención & control , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA