Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
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
11.
J Biol Chem ; 289(7): 3869-75, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24398677

RESUMEN

Exosomes are small vesicles (50-150 nm) of endocytic origin that are released by many different cell types. Exosomes in the tumor microenvironment may play a key role in facilitating cell-cell communication. Exosomes are reported to predominantly contain RNA and proteins. In this study, we investigated whether exosomes from pancreatic cancer cells and serum from patients with pancreatic ductal adenocarcinoma contain genomic DNA. Our results provide evidence that exosomes contain >10-kb fragments of double-stranded genomic DNA. Mutations in KRAS and p53 can be detected using genomic DNA from exosomes derived from pancreatic cancer cell lines and serum from patients with pancreatic cancer. In addition, using whole genome sequencing, we demonstrate that serum exosomes from patients with pancreatic cancer contain genomic DNA spanning all chromosomes. These results indicate that serum-derived exosomes can be used to determine genomic DNA mutations for cancer prediction, treatment, and therapy resistance.


Asunto(s)
Cromosomas Humanos , ADN de Neoplasias , Exosomas , Mutación , Neoplasias Pancreáticas , Proteínas Proto-Oncogénicas , Proteína p53 Supresora de Tumor , Proteínas ras , Línea Celular Tumoral , Cromosomas Humanos/genética , Cromosomas Humanos/metabolismo , Análisis Mutacional de ADN , ADN de Neoplasias/sangre , ADN de Neoplasias/genética , Exosomas/genética , Exosomas/metabolismo , Femenino , Humanos , Masculino , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas/sangre , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras) , Proteína p53 Supresora de Tumor/sangre , Proteína p53 Supresora de Tumor/genética , Proteínas ras/sangre , Proteínas ras/genética
12.
Curr Genomics ; 16(5): 295-303, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27047249

RESUMEN

Exosomes are lipid bilayer extracellular vesicles (EVs) of 50-150nm in size, which contain nucleic acids (mRNA, ncRNAs and DNA), proteins and lipids. They are secreted by all cells and circulate in all body fluids. Exosomes are key mediators of several processes in cancer that mediate tumor progression and metastasis. These nano-vesicles, when secreted from cancer cells, are enriched in non-coding RNAs (e.g. microRNAs) complexed with the RNA-Induced Silencing Complex (RISC), that mediate an efficient and rapid silencing of mRNAs at the recipient cell, reprogramming their transcriptome. MicroRNAs in circulation encapsulated in exosomes are protected from degradation by a lipid bilayer and might serve as potential non-invasive diagnostic and screening tools to detect early stage cancer, to facilitate treatment options and possible help in curative surgical therapy decisions. Additionally, engineered exosomes can be used as therapy vehicles for targeted delivery of RNAi molecules, escaping the immune system detection.

13.
Int J Mol Sci ; 16(6): 12324-44, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26039235

RESUMEN

Heterologous expression of a putative manganese superoxide dismutase gene (SOD2) of the basidiomycete Moniliophthora perniciosa complemented the phenotypes of a Saccharomyces cerevisiae sod2Δ mutant. Sequence analysis of the cloned M. perniciosa cDNA revealed an open reading frame (ORF) coding for a 176 amino acid polypeptide with the typical metal-binding motifs of a SOD2 gene, named MpSOD2. Phylogenetic comparison with known manganese superoxide dismutases (MnSODs) located the protein of M. perniciosa (MpSod2p) in a clade with the basidiomycete fungi Coprinopsis cinerea and Laccaria bicolor. Haploid wild-type yeast transformants containing a single copy of MpSOD2 showed increased resistance phenotypes against oxidative stress-inducing hydrogen peroxide and paraquat, but had unaltered phenotype against ultraviolet-C (UVC) radiation. The same transformants exhibited high sensitivity against treatment with the pro-mutagen diethylnitrosamine (DEN) that requires oxidation to become an active mutagen/carcinogen. Absence of MpSOD2 in the yeast sod2Δ mutant led to DEN hyper-resistance while introduction of a single copy of this gene restored the yeast wild-type phenotype. The haploid yeast wild-type transformant containing two SOD2 gene copies, one from M. perniciosa and one from its own, exhibited DEN super-sensitivity. This transformant also showed enhanced growth at 37 °C on the non-fermentable carbon source lactate, indicating functional expression of MpSod2p. The pro-mutagen dihydroethidium (DHE)-based fluorescence assay monitored basal level of yeast cell oxidative stress. Compared to the wild type, the yeast sod2Δ mutant had a much higher level of intrinsic oxidative stress, which was reduced to wild type (WT) level by introduction of one copy of the MpSOD2 gene. Taken together our data indicates functional expression of MpSod2 protein in the yeast S. cerevisiae.


Asunto(s)
Basidiomycota/genética , Mutación , Saccharomyces cerevisiae/crecimiento & desarrollo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Basidiomycota/clasificación , Dietilnitrosamina/farmacología , Farmacorresistencia Bacteriana , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Prueba de Complementación Genética , Peróxido de Hidrógeno/farmacología , Estrés Oxidativo/efectos de los fármacos , Paraquat/farmacología , Filogenia , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética
14.
Proc Natl Acad Sci U S A ; 108(11): 4394-9, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21368194

RESUMEN

MicroRNAs (miRNAs) are small RNA molecules that regulate gene expression at the posttranscriptional level and are critical for many cellular pathways. The disruption of miRNAs and their processing machineries also contributes to the development of human tumors. A common scenario for miRNA expression in carcinogenesis is emerging that shows that impaired miRNA production and/or down-regulation of these transcripts occurs in many neoplasms. Several of these lost miRNAs have tumor-suppressor features, so strategies to restore their expression globally in malignancies would be a welcome addition to the current therapeutic arsenal against cancer. Herein, we show that the small molecule enoxacin, a fluoroquinolone used as an antibacterial compound, enhances the production of miRNAs with tumor suppressor functions by binding to the miRNA biosynthesis protein TAR RNA-binding protein 2 (TRBP). The use of enoxacin in human cell cultures and xenografted, orthotopic, and metastatic mouse models reveals a TRBP-dependent and cancer-specific growth-inhibitory effect of the drug. These results highlight the key role of disrupted miRNA expression patterns in tumorigenesis, and suggest a unique strategy for restoring the distorted microRNAome of cancer cells to a more physiological setting.


Asunto(s)
Antineoplásicos/farmacología , Enoxacino/farmacología , MicroARNs/metabolismo , Neoplasias/patología , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Proteínas de Unión al ARN/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , MicroARNs/biosíntesis , Mutación/genética , Neoplasias/metabolismo , Unión Proteica/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Am Soc Nephrol ; 24(3): 385-92, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23274427

RESUMEN

Hypoxia is associated with tissue injury and fibrosis but its functional role in fibroblast activation and tissue repair/regeneration is unknown. Using kidney injury as a model system, we demonstrate that injured epithelial cells produce an increased number of exosomes with defined genetic information to activate fibroblasts. Exosomes released by injured epithelial cells promote proliferation, α-smooth muscle actin expression, F-actin expression, and type I collagen production in fibroblasts. Fibroblast activation is dependent on exosomes delivering TGF-ß1 mRNA among other yet to be identified moieties. This study suggests that TGF-ß1 mRNA transported by exosomes constitutes a rapid response to initiate tissue repair/regenerative responses and activation of fibroblasts when resident parenchyma is injured. The results also inform potential utility of exosome-targeted therapies to control tissue fibrosis.


Asunto(s)
Riñón/lesiones , Riñón/fisiopatología , Regeneración/fisiología , Factor de Crecimiento Transformador beta1/fisiología , Animales , Hipoxia de la Célula/fisiología , Células Cultivadas , Células Epiteliales/fisiología , Exosomas/fisiología , Fibroblastos/fisiología , Fibrosis , Humanos , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Células 3T3 NIH , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regeneración/genética , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/genética
16.
Trends Cancer ; 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39168775

RESUMEN

Exosomes are regarded as having transformative potential for clinical applications. Exosome-based liquid biopsies offer a noninvasive method for early cancer detection and real-time disease monitoring. Clinical trials are underway to validate the efficacy of exosomal biomarkers for enhancing diagnostic accuracy and predicting treatment responses. Additionally, engineered exosomes are being developed as targeted drug delivery systems that can navigate the bloodstream to deliver therapeutic agents to tumor sites, thus enhancing treatment efficacy while minimizing systemic toxicity. Exosomes also exhibit immunomodulatory properties, which are being harnessed to boost antitumor immune responses. In this review, we detail the latest advances in clinical trials and research studies, underscoring the potential of exosomes to revolutionize cancer care.

17.
Nat Commun ; 15(1): 1496, 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38383468

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC), a lethal disease, requires a grasp of its biology for effective therapies. Exosomes, implicated in cancer, are poorly understood in living systems. Here we use the genetically engineered mouse model (ExoBow) to map the spatiotemporal distribution of exosomes from healthy and PDAC pancreas in vivo to determine their biological significance. We show that, within the PDAC microenvironment, cancer cells establish preferential communication routes through exosomes with cancer associated fibroblasts and endothelial cells. The latter being a conserved event in the healthy pancreas. Inhibiting exosomes secretion in both scenarios enhances angiogenesis, underscoring their contribution to vascularization and to cancer. Inter-organ communication is significantly increased in PDAC with specific organs as most frequent targets of exosomes communication occurring in health with the thymus, bone-marrow, brain, and intestines, and in PDAC with the kidneys, lungs and thymus. In sum, we find that exosomes mediate an organized intra- and inter- pancreas communication network with modulatory effects in vivo.


Asunto(s)
Carcinoma Ductal Pancreático , Exosomas , Neoplasias Pancreáticas , Ratones , Animales , Exosomas/patología , Células Endoteliales/patología , Línea Celular Tumoral , Movimiento Celular , Neoplasias Pancreáticas/patología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Páncreas/patología , Microambiente Tumoral
18.
Mater Today Bio ; 28: 101189, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39221219

RESUMEN

The inflammatory response to wear particles derived from hip prothesis is considered a hallmark of periprosthetic osteolysis, which can ultimately lead to the need for revision surgery. Exosomes (Exos) have been associated with various bone pathologies, and there is increasing recognition in the literature that they actively transport molecules throughout the body. The role of wear particles in osteoblast-derived Exos is unknown, and the potential contribution of Exos to osteoimmune communication and periprosthetic osteolysis niche is still in its infancy. Given this, we investigate how titanium dioxide nanoparticles (TiO2 NPs), similar in size and composition to prosthetic wear particles, affect Exos biogenesis. Two osteoblastic cell models commonly used to study the response of osteoblasts to wear particles were selected as a proof of concept. The contribution of Exos to periprosthetic osteolysis was assessed by functional assays in which primary human macrophages were stimulated with bone-derived Exos. We demonstrated that TiO2 NPs enter multivesicular bodies, the nascent of Exos, altering osteoblast-derived Exos secretion and molecular cargo. No significant differences were observed in Exos morphology and size. However, functional assays reveal that Exos cargo enriched in uPA stimulates macrophages to a mixed M1 and M2 phenotype, inducing the release of pro- and anti-inflammatory signals characteristic of periprosthetic osteolysis. In addition, we demonstrated the expression of uPA in exosomes derived from the urine of patients with osteolysis. These results suggest that uPA can be a potential biomarker of osteolysis. In the future, uPa may serve as a possible non-invasive biomarker to identify patients at risk for peri-implant osteolysis.

20.
Biomater Sci ; 11(7): 2427-2444, 2023 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-36756939

RESUMEN

Titanium (Ti) and its alloys are the most widely used metallic biomaterials in total joint replacement; however, increasing evidence supports the degradation of its surface due to corrosion and wear processes releasing debris (ions, and micro and nanoparticles) and contribute to particle-induced osteolysis and implant loosening. Cell-to-cell communication involving several cell types is one of the major biological processes occurring during bone healing and regeneration at the implant-bone interface. In addition to the internal response of cells to the uptake and intracellular localization of wear debris, a red flag is the ability of titanium dioxide nanoparticles (mimicking wear debris) to alter cellular communication with the tissue background, disturbing the balance between osseous tissue integrity and bone regenerative processes. This study aims to understand whether titanium dioxide nanoparticles (TiO2 NPs) alter osteoblast-derived exosome (Exo) biogenesis and whether exosomal protein cargos affect the communication of osteoblasts with human mesenchymal stem/stromal cells (HMSCs). Osteoblasts are derived from mesenchymal stem cells coexisting in the bone microenvironment during development and remodelling. We observed that TiO2 NPs stimulate immature osteoblast- and mature osteoblast-derived Exo secretion that present a distinct proteomic cargo. Functional tests confirmed that Exos derived from both osteoblasts decrease the osteogenic differentiation of HMSCs. These findings are clinically relevant since wear debris alter extracellular communication in the bone periprosthetic niche, contributing to particle-induced osteolysis and consequent prosthetic joint failure.


Asunto(s)
Exosomas , Células Madre Mesenquimatosas , Nanopartículas , Osteólisis , Humanos , Osteogénesis , Titanio/efectos adversos , Osteólisis/inducido químicamente , Exosomas/metabolismo , Proteómica , Osteoblastos , Diferenciación Celular , Factores Inmunológicos , Comunicación Celular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA