Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 4549, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38811525

RESUMEN

Breast cancer metastasis to the brain is a clinical challenge rising in prevalence. However, the underlying mechanisms, especially how cancer cells adapt a distant brain niche to facilitate colonization, remain poorly understood. A unique metabolic feature of the brain is the coupling between neurons and astrocytes through glutamate, glutamine, and lactate. Here we show that extracellular vesicles from breast cancer cells with a high potential to develop brain metastases carry high levels of miR-199b-5p, which shows higher levels in the blood of breast cancer patients with brain metastases comparing to those with metastatic cancer in other organs. miR-199b-5p targets solute carrier transporters (SLC1A2/EAAT2 in astrocytes and SLC38A2/SNAT2 and SLC16A7/MCT2 in neurons) to hijack the neuron-astrocyte metabolic coupling, leading to extracellular retention of these metabolites and promoting cancer cell growth. Our findings reveal a mechanism through which cancer cells of a non-brain origin reprogram neural metabolism to fuel brain metastases.


Asunto(s)
Astrocitos , Neoplasias Encefálicas , Neoplasias de la Mama , MicroARNs , Neuronas , Humanos , MicroARNs/metabolismo , MicroARNs/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/genética , Neoplasias Encefálicas/secundario , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Femenino , Animales , Línea Celular Tumoral , Astrocitos/metabolismo , Astrocitos/patología , Neuronas/metabolismo , Neuronas/patología , Ratones , Transportador 2 de Aminoácidos Excitadores/metabolismo , Transportador 2 de Aminoácidos Excitadores/genética , Vesículas Extracelulares/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Regulación Neoplásica de la Expresión Génica , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Ácido Láctico/metabolismo , Proliferación Celular
2.
Nat Cell Biol ; 24(6): 954-967, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35637408

RESUMEN

Epidemiological studies demonstrate an association between breast cancer (BC) and systemic dysregulation of glucose metabolism. However, how BC influences glucose homeostasis remains unknown. We show that BC-derived extracellular vesicles (EVs) suppress pancreatic insulin secretion to impair glucose homeostasis. EV-encapsulated miR-122 targets PKM in ß-cells to suppress glycolysis and ATP-dependent insulin exocytosis. Mice receiving high-miR-122 EVs or bearing BC tumours exhibit suppressed insulin secretion, enhanced endogenous glucose production, impaired glucose tolerance and fasting hyperglycaemia. These effects contribute to tumour growth and are abolished by inhibiting EV secretion or miR-122, restoring PKM in ß-cells or supplementing insulin. Compared with non-cancer controls, patients with BC have higher levels of circulating EV-encapsulated miR-122 and fasting glucose concentrations but lower fasting insulin; miR-122 levels are positively associated with glucose and negatively associated with insulin. Therefore, EV-mediated impairment of whole-body glycaemic control may contribute to tumour progression and incidence of type 2 diabetes in some patients with BC.


Asunto(s)
Neoplasias de la Mama , Diabetes Mellitus Tipo 2 , Vesículas Extracelulares , MicroARNs , Animales , Neoplasias de la Mama/patología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Vesículas Extracelulares/metabolismo , Femenino , Glucosa/metabolismo , Homeostasis , Humanos , Insulina/metabolismo , Secreción de Insulina , Ratones , MicroARNs/genética , MicroARNs/metabolismo
3.
EMBO Rep ; 22(2): e51239, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33345445

RESUMEN

Metabolic reprogramming of non-cancer cells residing in a tumor microenvironment, as a result of the adaptations to cancer-derived metabolic and non-metabolic factors, is an emerging aspect of cancer-host interaction. We show that in normal and cancer-associated fibroblasts, breast cancer-secreted extracellular vesicles suppress mTOR signaling upon amino acid stimulation to globally reduce mRNA translation. This is through delivery of cancer-derived miR-105 and miR-204, which target RAGC, a component of Rag GTPases that regulate mTORC1 signaling. Following amino acid starvation and subsequent re-feeding, 13 C-arginine labeling of de novo synthesized proteins shows selective translation of proteins that cluster to specific cellular functional pathways. The repertoire of these newly synthesized proteins is altered in fibroblasts treated with cancer-derived extracellular vesicles, in addition to the overall suppressed protein synthesis. In human breast tumors, RAGC protein levels are inversely correlated with miR-105 in the stroma. Our results suggest that through educating fibroblasts to reduce and re-prioritize mRNA translation, cancer cells rewire the metabolic fluxes of amino acid pool and dynamically regulate stroma-produced proteins during periodic nutrient fluctuations.


Asunto(s)
MicroARNs , Proteínas de Unión al GTP Monoméricas , Neoplasias , Aminoácidos , Fibroblastos/metabolismo , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , MicroARNs/genética , Proteínas de Unión al GTP Monoméricas/metabolismo
4.
Breast Cancer Res ; 20(1): 127, 2018 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-30348200

RESUMEN

BACKGROUND: Bone is one of the most frequent metastatic sites of advanced breast cancer. Current therapeutic agents aim to inhibit osteoclast-mediated bone resorption but only have palliative effects. During normal bone remodeling, the balance between bone resorption and osteoblast-mediated bone formation is essential for bone homeostasis. One major function of osteoblast during bone formation is to secrete type I procollagen, which will then be processed before being crosslinked and deposited into the bone matrix. METHODS: Small RNA sequencing and quantitative real-time PCR were used to detect miRNA levels in patient blood samples and in the cell lysates as well as extracellular vesicles of parental and bone-tropic MDA-MB-231 breast cancer cells. The effects of cancer cell-derived extracellular vesicles isolated by ultracentrifugation and carrying varying levels of miR-218 were examined in osteoblasts by quantitative real-time PCR, Western blot analysis, and P1NP bone formation marker analysis. Cancer cells overexpressing miR-218 were examined by transcriptome profiling through RNA sequencing to identify intrinsic genes and pathways influenced by miR-218. RESULTS: We show that circulating miR-218 is associated with breast cancer bone metastasis. Cancer-secreted miR-218 directly downregulates type I collagen in osteoblasts, whereas intracellular miR-218 in breast cancer cells regulates the expression of inhibin ß subunits. Increased cancer secretion of inhibin ßA results in elevated Timp3 expression in osteoblasts and the subsequent repression of procollagen processing during osteoblast differentiation. CONCLUSIONS: Here we identify a twofold function of cancer-derived miR-218, whose levels in the blood are associated with breast cancer metastasis to the bone, in the regulation of type I collagen deposition by osteoblasts. The adaptation of the bone niche mediated by miR-218 might further tilt the balance towards osteolysis, thereby facilitating other mechanisms to promote bone metastasis.


Asunto(s)
Neoplasias Óseas/genética , Neoplasias de la Mama/patología , MicroARN Circulante/metabolismo , Colágeno Tipo I/metabolismo , MicroARNs/metabolismo , Osteoblastos/metabolismo , Adulto , Animales , Células de la Médula Ósea , Neoplasias Óseas/sangre , Neoplasias Óseas/secundario , Neoplasias de la Mama/sangre , Neoplasias de la Mama/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Cadena alfa 1 del Colágeno Tipo I , Regulación hacia Abajo , Femenino , Humanos , Subunidades beta de Inhibinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Osteoclastos/fisiología , Osteogénesis/genética , Cultivo Primario de Células
5.
Nat Cell Biol ; 20(5): 597-609, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29662176

RESUMEN

Cancer and other cells residing in the same niche engage various modes of interactions to synchronize and buffer the negative effects of environmental changes. Extracellular microRNAs (miRNAs) have recently been implicated in the intercellular crosstalk. Here we show a mechanistic model involving breast-cancer-secreted, extracellular-vesicle-encapsulated miR-105, which is induced by the oncoprotein MYC in cancer cells and, in turn, activates MYC signalling in cancer-associated fibroblasts (CAFs) to induce a metabolic program. This results in the capacity of CAFs to display different metabolic features in response to changes in the metabolic environment. When nutrients are sufficient, miR-105-reprogrammed CAFs enhance glucose and glutamine metabolism to fuel adjacent cancer cells. When nutrient levels are low and metabolic by-products accumulate, these CAFs detoxify metabolic wastes, including lactic acid and ammonium, by converting them into energy-rich metabolites. Thus, the miR-105-mediated metabolic reprogramming of stromal cells contributes to sustained tumour growth by conditioning the shared metabolic environment.


Asunto(s)
Neoplasias de la Mama/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Proliferación Celular , Reprogramación Celular , Metabolismo Energético , Exosomas/metabolismo , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Células del Estroma/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer/patología , Línea Celular Tumoral , Exosomas/genética , Exosomas/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , MicroARNs/genética , Células 3T3 NIH , Comunicación Paracrina , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal , Células del Estroma/patología , Factores de Tiempo , Carga Tumoral , Células Tumorales Cultivadas , Microambiente Tumoral
6.
Cell Res ; 26(2): 217-28, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26794868

RESUMEN

MicroRNAs (miRNAs) are critical regulators of gene expression, and exert extensive impacts on development, physiology, and disease of eukaryotes. A high degree of parallelism is found in the molecular basis of miRNA biogenesis and action in plants and animals. Recent studies interestingly suggest a potential cross-kingdom action of plant-derived miRNAs, through dietary intake, in regulating mammalian gene expression. Although the source and scope of plant miRNAs detected in mammalian specimens remain controversial, these initial studies inspired us to determine whether plant miRNAs can be detected in Western human sera and whether these plant miRNAs are able to influence gene expression and cellular processes related to human diseases such as cancer. Here we found that Western donor sera contained the plant miRNA miR159, whose abundance in the serum was inversely correlated with breast cancer incidence and progression in patients. In human sera, miR159 was predominantly detected in the extracellular vesicles, and was resistant to sodium periodate oxidation suggesting the plant-originated 2'-O-methylation on the 3' terminal ribose. In breast cancer cells but not non-cancerous mammary epithelial cells, a synthetic mimic of miR159 was capable of inhibiting proliferation by targeting TCF7 that encodes a Wnt signaling transcription factor, leading to a decrease in MYC protein levels. Oral administration of miR159 mimic significantly suppressed the growth of xenograft breast tumors in mice. These results demonstrate for the first time that a plant miRNA can inhibit cancer growth in mammals.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , MicroARNs/genética , MicroARNs/farmacología , ARN de Planta/genética , ARN de Planta/farmacología , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Epiteliales/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Ratones , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
8.
Nat Cell Biol ; 17(2): 183-94, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25621950

RESUMEN

Reprogrammed glucose metabolism as a result of increased glycolysis and glucose uptake is a hallmark of cancer. Here we show that cancer cells can suppress glucose uptake by non-tumour cells in the premetastatic niche, by secreting vesicles that carry high levels of the miR-122 microRNA. High miR-122 levels in the circulation have been associated with metastasis in breast cancer patients, and we show that cancer-cell-secreted miR-122 facilitates metastasis by increasing nutrient availability in the premetastatic niche. Mechanistically, cancer-cell-derived miR-122 suppresses glucose uptake by niche cells in vitro and in vivo by downregulating the glycolytic enzyme pyruvate kinase. In vivo inhibition of miR-122 restores glucose uptake in distant organs, including brain and lungs, and decreases the incidence of metastasis. These results demonstrate that, by modifying glucose utilization by recipient premetastatic niche cells, cancer-derived extracellular miR-122 is able to reprogram systemic energy metabolism to facilitate disease progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Glucosa/metabolismo , MicroARNs/metabolismo , Astrocitos/metabolismo , Secuencia de Bases , Neoplasias de la Mama/ultraestructura , Bromodesoxiuridina/metabolismo , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Exosomas/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Luciferasas/metabolismo , Pulmón/patología , MicroARNs/genética , Datos de Secuencia Molecular , Metástasis de la Neoplasia , Piruvato Quinasa/metabolismo
9.
Mol Cancer Res ; 12(11): 1597-609, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25103497

RESUMEN

UNLABELLED: Transforming growth factor beta (TGFß) proteins are multitasking cytokines, in which high levels at tumor sites generally correlate with poor prognosis in human patients with cancer. Previously, it was reported that TGFß downregulates the expression of ataxia telangiectasia-mutated (ATM) and mutS homolog 2 (MSH2) in breast cancer cells through an miRNA-mediated mechanism. In this study, expression of a panel of DNA-repair genes was examined, identifying breast cancer 1, early onset (BRCA1) as a target downregulated by TGFß through the miR181 family. Correlations between the expression levels of TGFß1 and the miR181/BRCA1 axis were observed in primary breast tumor specimens. By downregulating BRCA1, ATM, and MSH2, TGFß orchestrates DNA damage response in certain breast cancer cells to induce a "BRCAness" phenotype, including impaired DNA-repair efficiency and synthetic lethality to the inhibition of poly (ADP-ribose) polymerase (PARP). Xenograft tumors with active TGFß signaling exhibited resistance to the DNA-damaging agent doxorubicin but increased sensitivity to the PARP inhibitor ABT-888. Combination of doxorubicin with ABT-888 significantly improved the treatment efficacy in TGFß-active tumors. Thus, TGFß can induce "BRCAness" in certain breast cancers carrying wild-type BRCA genes and enhance the responsiveness to PARP inhibition, and the molecular mechanism behind this is characterized. IMPLICATIONS: These findings enable better selection of patients with sporadic breast cancer for PARP interventions, which have exhibited beneficial effects in patients carrying BRCA mutations.


Asunto(s)
Proteína BRCA1/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Reparación del ADN/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Factor de Crecimiento Transformador beta/farmacología , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular Tumoral , Reparación del ADN/efectos de los fármacos , Progresión de la Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Doxorrubicina/farmacología , Femenino , Inestabilidad Genómica/efectos de los fármacos , Humanos , Ratones , MicroARNs , Proteína 2 Homóloga a MutS/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología
10.
Sci Rep ; 4: 5750, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-25034888

RESUMEN

Growing evidence links tumor progression with chronic inflammatory processes and dysregulated activity of various immune cells. In this study, we demonstrate that various types of macrophages internalize microvesicles, called exosomes, secreted by breast cancer and non-cancerous cell lines. Although both types of exosomes targeted macrophages, only cancer-derived exosomes stimulated NF-κB activation in macrophages resulting in secretion of pro-inflammatory cytokines such as IL-6, TNFα, GCSF, and CCL2. In vivo mouse experiments confirmed that intravenously injected exosomes are efficiently internalized by macrophages in the lung and brain, which correlated with upregulation of inflammatory cytokines. In mice bearing xenografted human breast cancers, tumor-derived exosomes were internalized by macrophages in axillary lymph nodes thereby triggering expression of IL-6. Genetic ablation of Toll-like receptor 2 (TLR2) or MyD88, a critical signaling adaptor in the NF-κB pathway, completely abolished the effect of tumor-derived exosomes. In contrast, inhibition of TLR4 or endosomal TLRs (TLR3/7/8/9) failed to abrogate NF-κB activation by exosomes. We further found that palmitoylated proteins present on the surface of tumor-secreted exosomes contributed to NF-κB activation. Thus, our results highlight a novel mechanism used by breast cancer cells to induce pro-inflammatory activity of distant macrophages through circulating exosomal vesicles secreted during cancer progression.


Asunto(s)
Exosomas/fisiología , Macrófagos/inmunología , FN-kappa B/metabolismo , Receptor Toll-Like 2/metabolismo , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Comunicación Celular , Citocinas/metabolismo , Femenino , Humanos , Inmunomodulación , Células MCF-7 , Macrófagos/metabolismo , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Trasplante de Neoplasias , Transducción de Señal
11.
Cancer Cell ; 25(4): 501-15, 2014 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-24735924

RESUMEN

Cancer-secreted microRNAs (miRNAs) are emerging mediators of cancer-host crosstalk. Here we show that miR-105, which is characteristically expressed and secreted by metastatic breast cancer cells, is a potent regulator of migration through targeting the tight junction protein ZO-1. In endothelial monolayers, exosome-mediated transfer of cancer-secreted miR-105 efficiently destroys tight junctions and the integrity of these natural barriers against metastasis. Overexpression of miR-105 in nonmetastatic cancer cells induces metastasis and vascular permeability in distant organs, whereas inhibition of miR-105 in highly metastatic tumors alleviates these effects. miR-105 can be detected in the circulation at the premetastatic stage, and its levels in the blood and tumor are associated with ZO-1 expression and metastatic progression in early-stage breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Endotelio Vascular/patología , MicroARNs/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular , Endotelio Vascular/metabolismo , Femenino , Humanos , MicroARNs/genética , Metástasis de la Neoplasia
12.
Methods Mol Biol ; 1049: 239-53, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23913221

RESUMEN

Most of the research on tumor cell metabolism has focused on glucose utilization. However, when glucose is limited, solid tumors are forced to catabolize alternative substrates such as fatty acids and amino acids as an energy source. Measuring these alternations in tumor cell metabolism enables us to track neoplastic changes in the tissue to lead towards a more reliable diagnostic outcome. Although a very small number of elements are used in biochemistry, the metabolome is structurally diverse for the production of simple compounds such as phosphate and amino acids as well as more structurally complex compounds such as nucleotides, oligosaccharides, and complex lipids. Characterization of the metabolome, therefore, requires analytical methods that can handle a wide range of molecular structures and physicochemical properties, including solubility, polarity, and molecular weight. A further factor for consideration in the selection of technology for metabolomics is the wide range of concentrations of biochemical typically present in biological systems. MS has established itself as the high-throughput, information-rich, industrially stable approach to assess both the composition of diverse sample types as well as changes to that composition following perturbation.


Asunto(s)
Carcinoma/metabolismo , Metabolómica , Biología Molecular/métodos , Neoplasias Ováricas/metabolismo , Aminoácidos/metabolismo , Carcinoma/patología , Femenino , Cromatografía de Gases y Espectrometría de Masas/métodos , Humanos , Neoplasias Ováricas/patología
13.
BMC Cancer ; 12: 532, 2012 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-23164239

RESUMEN

BACKGROUND: Pituitary tumor-transforming gene (PTTG) is an oncogene that is overexpressed in variety of tumors and exhibits characteristics of a transforming gene. Previous transgenic mouse models to access the tumorigenic potential in the pituitary and ovary have resulted in dysplasia without formation of visible tumors, possibly due to the insufficient expression of PTTG. PTTG expression level is critical for ovarian tumorigenesis in a xenograft model. Therefore, the tumorigenic function of PTTG in vivo remains unclear. We generated a transgenic mouse that overexpresses PTTG driven by the CMV promoter to determine whether PTTG functions as a transforming oncogene that is capable of initiating tumorigenesis. METHODS: Transgenic animals were generated by microinjection of PTTG transgene into the male pronucleus of FVB 0.5 day old embryos. Expression levels of PTTG in tissues of transgenic animals were analyzed using an immunohistochemical analysis. H&E staining and immunohistostaining were performed to examine the type of tumor in transgenic and PTTG transgenic/p53+/- animals. RESULTS: PTTG transgenic offspring (TgPTTG) were monitored for tumor development at various ages. H&E analysis was performed to identify the presence of cancer and hyperplastic conditions verified with the proliferation marker PCNA and the microvessel marker CD31. Immunohistochemistry was performed to determine transgene expression, revealing localization to the epithelium of the fallopian tube, with more generalized expression in the liver, lung, kidney, and spleen. At eight months of age, 2 out of 15 TgPTTG developed ovarian cancer, 2 out of 15 developed benign tumors, 2 out of 15 developed cervical dysplasia, and 3 out of 15 developed adenomyosis of the uterus. At ten months of age, 2 out of 10 TgPTTG developed adenocarcinoma of the ovary, 1 out of 10 developed a papillary serous adenocarcinoma, and 2 out of 10 presented with atypia of ovarian epithelial cells. Tumorigenesis is a multi-step process, often requiring multiple oncogenes and/or inactivation of tumor suppressor genes. Therefore, to understand the contribution of p53 to PTTG induced tumorigenesis, we crossbred TgPTTG to p53+/- mice and maintained those 8 to 10 months. TgPTTG/p53+/- animals developed sarcomas faster than p53+/- alone as well as different tumor types in addition to cervical carcinomas in situ in 10 out of 17 females. CONCLUSIONS: We conclude that while PTTG is a functional transforming oncogene, it requires an additional partner to effectively promote tumorigenesis through the loss of p53 include or between function or modulation.


Asunto(s)
Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Proteínas de Neoplasias/genética , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Oncogenes , Proteína p53 Supresora de Tumor/genética , Animales , Cruzamiento/métodos , Epitelio/metabolismo , Epitelio/patología , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Antígeno Nuclear de Célula en Proliferación/genética , Regiones Promotoras Genéticas , Securina
14.
PLoS One ; 7(7): e42265, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22860102

RESUMEN

Application of doxorubicin (Dox) for the treatment of cancer is restricted due to its severe side effects. We used combination strategy by combining doxorubicin (Dox) with withaferin A (WFA) to minimize the ill effects of Dox. Treatment of various epithelial ovarian cancer cell lines (A2780, A2780/CP70 and CaOV3) with combination of WFA and Dox (WFA/DOX) showed a time- and dose-dependent synergistic effect on inhibition of cell proliferation and induction of cell death, thus reducing the dosage requirement of Dox. Combination treatment resulted in a significant enhancement of ROS production resulting in immense DNA damage, induction of autophagy analyzed by transmission electron microscope and increase in expression of autophagy marker LC3B, and culminated in cell death analyzed by cleaved caspase 3. We validated combination therapy on tumor growth using an in vitro 3Dimension (3D) tumor model and the more classic in vivo xenograft model of ovarian cancer. Both tumor models showed a 70 to 80% reduction in tumor growth compared to control or animals treated with WFA or Dox alone. Immunohistochemical analysis of the tumor tissues from animals treated with WFA/Dox combination showed a significant reduction in cell proliferation and formation of microvessels accompanied by increased in LC3B level, cleaved caspase 3, and DNA damage. Taken together, our data suggest that combining WFA with Dox decreases the dosage requirement of Dox, therefore, minimizing/eliminating the severe side effects associated with high doses of DOX, suggesting the application of this combination strategy for the treatment of ovarian and other cancers with no or minimum side effects.


Asunto(s)
Antineoplásicos/uso terapéutico , Autofagia/efectos de los fármacos , Doxorrubicina/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Witanólidos/farmacología , Animales , Línea Celular Tumoral , Daño del ADN , Doxorrubicina/farmacología , Sinergismo Farmacológico , Femenino , Citometría de Flujo , Humanos , Ratones , Neoplasias Ováricas/patología
15.
Biochem Biophys Res Commun ; 423(4): 819-25, 2012 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-22713472

RESUMEN

Cisplatin derivatives are used as the mainline treatment of ovarian cancer, despite their severe side effects and development of resistance. We developed a novel combination therapy by combining cisplatin with withaferin A. Treatment of ovarian cancer cell lines with combination therapy acted synergistically to induce cell death, thus required a lower dose of cisplatin to achieve the same therapeutic effect. WFA and cisplatin combination induced cell death through the generation of reactive oxygen species (ROS) for WFA, while DNA damage for cisplatin, suggesting that cisplatin binds directly to DNA to form adducts while WFA indirectly damages DNA through ROS generation. Our results for the first time suggest that combining low dose of cisplatin with suboptimal dose of WFA can serve as a potential combination therapy for the treatment of ovarian cancer with the potential to minimize/eliminate the side effects associated with high doses of cisplatin.


Asunto(s)
Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias Ováricas/tratamiento farmacológico , Línea Celular Tumoral , Cisplatino/administración & dosificación , Sinergismo Farmacológico , Femenino , Humanos , Especies Reactivas de Oxígeno/metabolismo , Witanólidos/administración & dosificación
16.
PLoS One ; 6(5): e19963, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21625518

RESUMEN

In this study, we characterized the metabolome of the human ovary and identified metabolic alternations that coincide with primary epithelial ovarian cancer (EOC) and metastatic tumors resulting from primary ovarian cancer (MOC) using three analytical platforms: gas chromatography mass spectrometry (GC/MS) and liquid chromatography tandem mass spectrometry (LC/MS/MS) using buffer systems and instrument settings to catalog positive or negative ions. The human ovarian metabolome was found to contain 364 biochemicals and upon transformation of the ovary caused changes in energy utilization, altering metabolites associated with glycolysis and ß-oxidation of fatty acids--such as carnitine (1.79 fold in EOC, p<0.001; 1.88 fold in MOC, p<0.001), acetylcarnitine (1.75 fold in EOC, p<0.001; 2.39 fold in MOC, p<0.001), and butyrylcarnitine (3.62 fold, p<0.0094 in EOC; 7.88 fold, p<0.001 in MOC). There were also significant changes in phenylalanine catabolism marked by increases in phenylpyruvate (4.21 fold; p = 0.0098) and phenyllactate (195.45 fold; p<0.0023) in EOC. Ovarian cancer also displayed an enhanced oxidative stress response as indicated by increases in 2-aminobutyrate in EOC (1.46 fold, p = 0.0316) and in MOC (2.25 fold, p<0.001) and several isoforms of tocopherols. We have also identified novel metabolites in the ovary, specifically N-acetylasparate and N-acetyl-aspartyl-glutamate, whose role in ovarian physiology has yet to be determined. These data enhance our understanding of the diverse biochemistry of the human ovary and demonstrate metabolic alterations upon transformation. Furthermore, metabolites with significant changes between groups provide insight into biochemical consequences of transformation and are candidate biomarkers of ovarian oncogenesis. Validation studies are warranted to determine whether these compounds have clinical utility in the diagnosis or clinical management of ovarian cancer patients.


Asunto(s)
Neoplasias Ováricas/metabolismo , Ovario/metabolismo , Metabolismo de los Hidratos de Carbono , Ácidos Grasos/metabolismo , Femenino , Humanos , Metástasis de la Neoplasia , Neoplasias Ováricas/patología , Estrés Oxidativo
17.
Histol Histopathol ; 25(1): 113-20, 2010 01.
Artículo en Inglés | MEDLINE | ID: mdl-19924647

RESUMEN

Ovarian cancer is the most lethal cancer of the female reproductive tract, accounting for ~15,000 deaths per year according to the National Cancer Institute and American Cancer Society. This review article covers risk factors for the development of ovarian cancer, current detection strategies, prognostic markers, treatment strategies, etiology of tumorigenesis, and ovarian somatic stem cells. While the etiology of ovarian cancer is still unknown, several theories have been proposed as the mechanism of carcinogenesis. One theory states that the surface epithelium undergoing invagination and forming inclusion cysts that are exposed to growth factors and cytokines. The "gonadotropin theory" has also been proposed. Other reigning models for tumorigenesis include the stochastical model where a distinct population of cells acquires somatic mutations leading to metastasis, and the hierarchical model where the tumor is initiated by cancer stem cells (CSCs). CSCs isolated from primary tumors have the ability to regenerate the tumor and reconstitute the original tumor phenotype with as few as 100 cells. CSCs from ovarian carcinomas display the cell surface markers CD44+CD117+CD133+. CSCs are also thought to account for chemotherapy resistance through the expression of highly selective transporters ABCG2 and MDR1 and activation of TLR4/MyD88. The side population has been characterized by their ability to efflux lipophilic substrates, including the dye Hoechst 33342 and many chemotherapy agents. This ability has been attributed to the expression of the transporters ABCG2 and MDR1.


Asunto(s)
Carcinoma/patología , Células Madre Neoplásicas/patología , Neoplasias Ováricas/patología , Animales , Biomarcadores de Tumor , Carcinoma/diagnóstico , Carcinoma/epidemiología , Carcinoma/genética , Carcinoma/terapia , Femenino , Humanos , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/epidemiología , Neoplasias Ováricas/genética , Neoplasias Ováricas/terapia , Pronóstico , Factores de Riesgo
18.
J Ovarian Res ; 2(1): 12, 2009 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-19781107

RESUMEN

Development of mouse models representing human spontaneous ovarian cancer has been hampered by the lack of understanding of the etiology of this very complex disease. Mouse models representing the different types of ovarian cancer are needed to understand how epithelial ovarian cancer differs from granulosa cell tumors. Many different methods have been used to generate a viable genetic model with limited success. This review focuses on the methods of various investigators and the limitations of each model in establishing a reproducible and inheritable line to study this disease.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...