Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
J Transl Med ; 21(1): 578, 2023 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-37641131

RESUMEN

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is expected to soon surpass colorectal cancer as a leading cause of cancer mortality in both males and females in the US, only lagging behind lung cancer. The lethality of PDAC is driven by late diagnosis and inefficient therapies. The complex biology of PDAC involves various cellular components, including exosomes that carry molecular information between cells. Thus, recipient cells can be reprogrammed, impacting tumorigenesis. Rab27a is a GTPase responsible for the last step of exosomes biogenesis. Hence, dissecting the mechanisms that regulate the expression of Rab27a and that control exosomes biogenesis can provide fundamental insights into the molecular underpinnings regulating PDAC progression. METHODS: To assess the mechanism that regulates Rab27a expression in PDAC, we used PDAC cell lines. The biological significance of these findings was validated in PDAC genetically engineered mouse models (GEMMs) and human samples. RESULTS: In this work we demonstrate in human PDAC samples and GEMMs that Rab27a expression decreases throughout the development of the disease, and that Rab27a knockout promotes disease progression. What is more, we demonstrate that Rab27a expression is epigenetically regulated in PDAC. Treatment with demethylating agents increases Rab27a expression specifically in human PDAC cell lines. We found that SMC3, a component of the cohesin complex, regulates Rab27a expression in PDAC. SMC3 methylation is present in human PDAC specimens and treatment with demethylating agents increases SMC3 expression in human PDAC cell lines. Most importantly, high levels of SMC3 methylation are associated with a worse prognosis in PDAC. Mechanistically, we identified an enhancer region within the Rab27a gene that recruits SMC3, and modulates Rab27a expression. CONCLUSION: Overall, we dissected a mechanism that regulates Rab27a expression during PDAC progression and impacts disease prognosis.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Femenino , Humanos , Masculino , Animales , Ratones , Neoplasias Pancreáticas/genética , Páncreas , Carcinoma Ductal Pancreático/genética , Epigénesis Genética , Proteínas Cromosómicas no Histona , Proteoglicanos Tipo Condroitín Sulfato , Proteínas de Ciclo Celular , Proteínas rab27 de Unión a GTP/genética , Neoplasias Pancreáticas
2.
Nature ; 546(7659): 498-503, 2017 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-28607485

RESUMEN

The mutant form of the GTPase KRAS is a key driver of pancreatic cancer but remains a challenging therapeutic target. Exosomes are extracellular vesicles generated by all cells, and are naturally present in the blood. Here we show that enhanced retention of exosomes, compared to liposomes, in the circulation of mice is likely due to CD47-mediated protection of exosomes from phagocytosis by monocytes and macrophages. Exosomes derived from normal fibroblast-like mesenchymal cells were engineered to carry short interfering RNA or short hairpin RNA specific to oncogenic KrasG12D, a common mutation in pancreatic cancer. Compared to liposomes, the engineered exosomes (known as iExosomes) target oncogenic KRAS with an enhanced efficacy that is dependent on CD47, and is facilitated by macropinocytosis. Treatment with iExosomes suppressed cancer in multiple mouse models of pancreatic cancer and significantly increased overall survival. Our results demonstrate an approach for direct and specific targeting of oncogenic KRAS in tumours using iExosomes.


Asunto(s)
Exosomas/metabolismo , Silenciador del Gen , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Proteínas Proto-Oncogénicas p21(ras)/genética , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Animales , Antígeno CD47/metabolismo , Modelos Animales de Enfermedad , Exosomas/inmunología , Femenino , Terapia Genética , Liposomas/inmunología , Ratones , Monocitos/citología , Monocitos/inmunología , Metástasis de la Neoplasia/prevención & control , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/patología , Pinocitosis , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Tasa de Supervivencia
4.
Gut ; 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35012996

RESUMEN

OBJECTIVE: Intratumor heterogeneity drives cancer progression and therapy resistance. However, it has yet to be determined whether and how subpopulations of cancer cells interact and how this interaction affects the tumour. DESIGN: We have studied the spontaneous flow of extracellular vesicles (EVs) between subpopulations of cancer cells: cancer stem cells (CSC) and non-stem cancer cells (NSCC). To determine the biological significance of the most frequent communication route, we used pancreatic ductal adenocarcinoma (PDAC) orthotopic models, patient-derived xenografts (PDXs) and genetically engineered mouse models (GEMMs). RESULTS: We demonstrate that PDAC tumours establish an organised communication network between subpopulations of cancer cells using EVs called the EVNet). The EVNet is plastic and reshapes in response to its environment. Communication within the EVNet occurs preferentially from CSC to NSCC. Inhibition of this communication route by impairing Rab27a function in orthotopic xenographs, GEMMs and PDXs is sufficient to hamper tumour growth and phenocopies the inhibition of communication in the whole tumour. Mechanistically, we provide evidence that CSC EVs use agrin protein to promote Yes1 associated transcriptional regulator (YAP) activation via LDL receptor related protein 4 (LRP-4). Ex vivo treatment of PDXs with antiagrin significantly impairs proliferation and decreases the levels of activated YAP.Patients with high levels of agrin and low inactive YAP show worse disease-free survival. In addition, patients with a higher number of circulating agrin+ EVs show a significant increased risk of disease progression. CONCLUSION: PDAC tumours establish a cooperation network mediated by EVs that is led by CSC and agrin, which allows tumours to adapt and thrive. Targeting agrin could make targeted therapy possible for patients with PDAC and has a significant impact on CSC that feeds the tumour and is at the centre of therapy resistance.

5.
Nature ; 523(7559): 177-82, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-26106858

RESUMEN

Exosomes are lipid-bilayer-enclosed extracellular vesicles that contain proteins and nucleic acids. They are secreted by all cells and circulate in the blood. Specific detection and isolation of cancer-cell-derived exosomes in the circulation is currently lacking. Using mass spectrometry analyses, we identify a cell surface proteoglycan, glypican-1 (GPC1), specifically enriched on cancer-cell-derived exosomes. GPC1(+) circulating exosomes (crExos) were monitored and isolated using flow cytometry from the serum of patients and mice with cancer. GPC1(+) crExos were detected in the serum of patients with pancreatic cancer with absolute specificity and sensitivity, distinguishing healthy subjects and patients with a benign pancreatic disease from patients with early- and late-stage pancreatic cancer. Levels of GPC1(+) crExos correlate with tumour burden and the survival of pre- and post-surgical patients. GPC1(+) crExos from patients and from mice with spontaneous pancreatic tumours carry specific KRAS mutations, and reliably detect pancreatic intraepithelial lesions in mice despite negative signals by magnetic resonance imaging. GPC1(+) crExos may serve as a potential non-invasive diagnostic and screening tool to detect early stages of pancreatic cancer to facilitate possible curative surgical therapy.


Asunto(s)
Exosomas/metabolismo , Glipicanos , Neoplasias Pancreáticas/diagnóstico , Animales , Biomarcadores/sangre , Línea Celular Tumoral , Exosomas/genética , Femenino , Glipicanos/sangre , Glipicanos/metabolismo , Células HCT116 , Humanos , Células MCF-7 , Masculino , Ratones , Células 3T3 NIH , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas p21(ras) , Proteínas ras/metabolismo
6.
Mol Cell ; 49(3): 524-35, 2013 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-23273978

RESUMEN

Binding within or nearby target genes involved in cell proliferation and survival enables the p53 tumor suppressor gene to regulate their transcription and cell-cycle progression. Using genome-wide chromatin-binding profiles, we describe binding of p53 also to regions located distantly from any known p53 target gene. Interestingly, many of these regions possess conserved p53-binding sites and all known hallmarks of enhancer regions. We demonstrate that these p53-bound enhancer regions (p53BERs) indeed contain enhancer activity and interact intrachromosomally with multiple neighboring genes to convey long-distance p53-dependent transcription regulation. Furthermore, p53BERs produce, in a p53-dependent manner, enhancer RNAs (eRNAs) that are required for efficient transcriptional enhancement of interacting target genes and induction of a p53-dependent cell-cycle arrest. Thus, our results ascribe transcription enhancement activity to p53 with the capacity to regulate multiple genes from a single genomic binding site. Moreover, eRNA production from p53BERs is required for efficient p53 transcription enhancement.


Asunto(s)
Elementos de Facilitación Genéticos , ARN/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo , Puntos de Control del Ciclo Celular/genética , Cromatina/metabolismo , Cromosomas Humanos/metabolismo , Regulación Neoplásica de la Expresión Génica , Genes , Humanos , Células MCF-7 , Modelos Genéticos , Unión Proteica/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN no Traducido/metabolismo
7.
Semin Cell Dev Biol ; 78: 13-21, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-28803894

RESUMEN

Exosomes are small extracellular vesicles with a significant role in most processes associated with cancer. On one hand, exosomes role in the different hallmarks of cancer has been widely described, highlighting the urge to understand the potential to target communication mediated by exosomes as a novel therapeutic approach in cancer. On the other hand, exosomes stability in circulation and tumor-targeting capacity shows their applicability in the delivery of anti-cancer molecules. This review will discuss the dual applicability of exosomes in cancer focusing on their usage for therapy improvement, or their targeting to block their supportive role in tumor progression and response to therapy. We highlight the current developments and the strategies used to enhance the potential of exosomes to become clinical partners in the treatment of cancer.


Asunto(s)
Comunicación Celular/fisiología , Exosomas/patología , Neoplasias/patología , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/sangre , Comunicación Celular/efectos de los fármacos , Exosomas/efectos de los fármacos , Humanos , Neoplasias/tratamiento farmacológico
8.
Pancreatology ; 20(3): 571-575, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32024605

RESUMEN

The clinical management of patients with pancreatic cystic lesions is of utmost importance to identify those at high risk for pathological progression. Current recommendations are guided by clinical presentation and radiologic criteria, but the results fall short for a disease that the only curative option is surgical resection. There is an urgent need for the introduction of biomarkers that can help in risk assessment of such lesions. We report a case of a pancreatic cystic lesion without imagiological findings suggestive of advanced disease, and high levels of a circulating biomarker, glypican-1 (GPC-1), which parallel those of patients with pancreatic cancer. One year after, the patient revealed malignant progression at follow-up. Our report is unprecedented in the literature. It describes a clinical case in which a biomarker was positive for a patient that only showed progression one year after its detection. This clinical information goes beyond the current knowledge in the field because it shows that the introduction of liquid biopsy and biomarkers is a highly promising clinical tool for the non-invasive assessment of pancreatic cancer precursor lesions, ultimately increasing the rate of patients eligible for surgical resection.


Asunto(s)
Exosomas/metabolismo , Glipicanos/química , Quiste Pancreático/diagnóstico , Biopsia con Aguja Fina , Diagnóstico Diferencial , Progresión de la Enfermedad , Femenino , Humanos , Persona de Mediana Edad , Pancreatectomía , Quiste Pancreático/diagnóstico por imagen , Quiste Pancreático/patología , Neoplasias Pancreáticas/patología , Medición de Riesgo
9.
Int J Mol Sci ; 20(3)2019 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-30699928

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease, associated with a late diagnosis and a five-year survival rate of 8%. Currently available treatments fall short in improving the survival and quality of life of PDAC patients. The only possible curative option is still the surgical resection of the tumor. Exosomes are extracellular vesicles secreted by cells that transport proteins, lipids, and nucleic acids to other cells, triggering phenotypic changes in the recipient cells. Tumor cells often secrete increased amounts of exosomes. Tumor exosomes are now accepted as important players in the remodeling of PDAC tumor stroma, particularly in the establishment of an immunosuppressive microenvironment. This has sparked the interest in their usefulness as mediators of immunomodulatory effects for the treatment of PDAC. In fact, exosomes are now under study to understand their potential as nanocarriers to stimulate an immune response against cancer. This review highlights the latest findings regarding the function of exosomes in tumor-driven immunomodulation, and the challenges and advantages associated with the use of these vesicles to potentiate immunotherapy in PDAC.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/terapia , Exosomas/metabolismo , Inmunoterapia/métodos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/terapia , Animales , Carcinoma Ductal Pancreático/fisiopatología , Humanos , Neoplasias Pancreáticas/fisiopatología , Calidad de Vida
10.
Semin Cancer Biol ; 27: 46-51, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24607282

RESUMEN

MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression post-transcriptionally. MicroRNAs target about 80% of the protein-coding mRNAs and therefore can be considered master regulators of multiple cellular pathways, contributing to the fine-tuning the cell's most important processes, like the ones involved in cellular growth and proliferation. Deregulation of miRNAs plays a fundamental role in the onset, progression and dissemination of many cancers; therefore impairment of miRNA biosynthesis is an important event in the tumorigenic cascade. MicroRNA synthesis is a multistep regulated process that requires transport of RNA molecules from the nucleus to the cytoplasm. The immature miRNA species that are produced in the nucleus are exported through the nuclear pore complexes via mobile export receptors. Small RNAs such as precursors of miRNAs (pre-miRNAs) are transported out of the nucleus by a specific nuclear transport receptor, exportin-5 (XPO5). Pre-miRNA nuclear export is a fundamental step in miRNAs biosynthesis and its deregulation through inactivating mutations in the XPO5 gene can lead to pre-miRNA nuclear accumulation and disturbance of mature miRNA expression. In addition, it is becoming increasingly evident that mature miRNAs also function as gene regulators in the nuclear compartment. In this review, we will discuss the export of miRNA precursors and its impairment in human cancer as well as the recently described nuclear functions of miRNAs.


Asunto(s)
MicroARNs/genética , MicroARNs/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Transporte Activo de Núcleo Celular , Regulación Neoplásica de la Expresión Génica , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA