Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 278
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(8): 1610-1626, 2023 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-37059067

RESUMEN

Intercellular communication is a key feature of cancer progression and metastasis. Extracellular vesicles (EVs) are generated by all cells, including cancer cells, and recent studies have identified EVs as key mediators of cell-cell communication via packaging and transfer of bioactive constituents to impact the biology and function of cancer cells and cells of the tumor microenvironment. Here, we review recent advances in understanding the functional contribution of EVs to cancer progression and metastasis, as cancer biomarkers, and the development of cancer therapeutics.


Asunto(s)
Vesículas Extracelulares , Neoplasias , Humanos , Neoplasias/patología , Comunicación Celular/fisiología , Biomarcadores de Tumor , Microambiente Tumoral/fisiología
2.
Nat Rev Mol Cell Biol ; 21(6): 341-352, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32300252

RESUMEN

Epithelial-mesenchymal transition (EMT) encompasses dynamic changes in cellular organization from epithelial to mesenchymal phenotypes, which leads to functional changes in cell migration and invasion. EMT occurs in a diverse range of physiological and pathological conditions and is driven by a conserved set of inducing signals, transcriptional regulators and downstream effectors. With over 5,700 publications indexed by Web of Science in 2019 alone, research on EMT is expanding rapidly. This growing interest warrants the need for a consensus among researchers when referring to and undertaking research on EMT. This Consensus Statement, mediated by 'the EMT International Association' (TEMTIA), is the outcome of a 2-year-long discussion among EMT researchers and aims to both clarify the nomenclature and provide definitions and guidelines for EMT research in future publications. We trust that these guidelines will help to reduce misunderstanding and misinterpretation of research data generated in various experimental models and to promote cross-disciplinary collaboration to identify and address key open questions in this research field. While recognizing the importance of maintaining diversity in experimental approaches and conceptual frameworks, we emphasize that lasting contributions of EMT research to increasing our understanding of developmental processes and combatting cancer and other diseases depend on the adoption of a unified terminology to describe EMT.


Asunto(s)
Investigación Biomédica/normas , Transición Epitelial-Mesenquimal , Animales , Movimiento Celular , Plasticidad de la Célula , Consenso , Biología Evolutiva/normas , Humanos , Neoplasias/patología , Terminología como Asunto
3.
Immunity ; 50(3): 547-549, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30893584

RESUMEN

Exosomes are nano-sized extracellular vesicles that contain DNA, RNA, proteins, and lipids. Exosomes likely participate in facilitating intercellular communication and tumor growth. In this issue of Immunity, Zhang et al. (2019) report on the metabolic activity of B cell-derived exosomes in facilitating the suppression of cytotoxic T cells.


Asunto(s)
Exosomas , Neoplasias , Linfocitos B , Linfocitos T CD8-positivos , Comunicación Celular , Humanos
5.
EMBO J ; 41(7): e109470, 2022 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-35212000

RESUMEN

Skin wound repair is essential for organismal survival and failure of which leads to non-healing wounds, a leading health issue worldwide. However, mechanistic understanding of chronic wounds remains a major challenge due to lack of appropriate genetic mouse models. αSMA+ myofibroblasts, a unique class of dermal fibroblasts, are associated with cutaneous wound healing but their precise function remains unknown. We demonstrate that genetic depletion of αSMA+ myofibroblasts leads to pleiotropic wound healing defects, including lack of reepithelialization and granulation, dampened angiogenesis, and heightened hypoxia, hallmarks of chronic non-healing wounds. Other wound-associated FAP+ and FSP1+ fibroblasts do not exhibit such dominant functions. While type I collagen (COL1) expressing cells play a role in the repair process, COL1 produced by αSMA+ myofibroblasts is surprisingly dispensable for wound repair. In contrast, we show that ß1 integrin from αSMA+ myofibroblasts, but not TGFßRII, is essential for wound healing, facilitating contractility, reepithelization, and vascularization. Collectively, our study provides evidence for the functions of myofibroblasts in ß1 integrin-mediated wound repair with potential implications for treating chronic non-healing wounds.


Asunto(s)
Colágeno Tipo I , Miofibroblastos , Cicatrización de Heridas , Animales , Colágeno Tipo I/genética , Fibroblastos , Integrina beta1/genética , Ratones , Piel
6.
Nature ; 577(7791): 549-555, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31942075

RESUMEN

Treatment with immune checkpoint blockade (ICB) has revolutionized cancer therapy. Until now, predictive biomarkers1-10 and strategies to augment clinical response have largely focused on the T cell compartment. However, other immune subsets may also contribute to anti-tumour immunity11-15, although these have been less well-studied in ICB treatment16. A previously conducted neoadjuvant ICB trial in patients with melanoma showed via targeted expression profiling17 that B cell signatures were enriched in the tumours of patients who respond to treatment versus non-responding patients. To build on this, here we performed bulk RNA sequencing and found that B cell markers were the most differentially expressed genes in the tumours of responders versus non-responders. Our findings were corroborated using a computational method (MCP-counter18) to estimate the immune and stromal composition in this and two other ICB-treated cohorts (patients with melanoma and renal cell carcinoma). Histological evaluation highlighted the localization of B cells within tertiary lymphoid structures. We assessed the potential functional contributions of B cells via bulk and single-cell RNA sequencing, which demonstrate clonal expansion and unique functional states of B cells in responders. Mass cytometry showed that switched memory B cells were enriched in the tumours of responders. Together, these data provide insights into the potential role of B cells and tertiary lymphoid structures in the response to ICB treatment, with implications for the development of biomarkers and therapeutic targets.


Asunto(s)
Linfocitos B/inmunología , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/inmunología , Inmunoterapia , Melanoma/tratamiento farmacológico , Melanoma/inmunología , Estructuras Linfoides Terciarias/inmunología , Linfocitos B/citología , Linfocitos B/metabolismo , Biomarcadores de Tumor/análisis , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/cirugía , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/inmunología , Células Clonales/citología , Células Clonales/inmunología , Células Clonales/metabolismo , Células Dendríticas Foliculares/citología , Células Dendríticas Foliculares/inmunología , Regulación Neoplásica de la Expresión Génica , Humanos , Memoria Inmunológica/inmunología , Espectrometría de Masas , Melanoma/patología , Melanoma/cirugía , Metástasis de la Neoplasia/genética , Fenotipo , Pronóstico , RNA-Seq , Receptores Inmunológicos/inmunología , Análisis de la Célula Individual , Linfocitos T/citología , Linfocitos T/inmunología , Transcriptoma
7.
J Pathol ; 262(1): 22-36, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37728068

RESUMEN

Cancer-associated fibroblasts (CAFs) deposit and remodel collagens in the tumor stroma, impacting cancer progression and efficacy of interventions. CAFs are the focus of new therapeutics with the aim of normalizing the tumor microenvironment. To do this, a better understanding of CAF heterogeneity and collagen composition in cancer is needed. In this study, we sought to profile the expression of collagens at multiple levels with the goal of identifying cancer biomarkers. We investigated the collagen expression pattern in various cell types and CAF subtypes in a publicly available single-cell RNA sequencing (RNA-seq) dataset of pancreatic ductal adenocarcinoma. Next, we investigated the collagen expression profile in tumor samples across cancer types from The Cancer Genome Atlas (TCGA) database and evaluated if specific patterns of collagen expression were associated with prognosis. Finally, we profiled circulating collagen peptides using a panel of immunoassays to measure collagen fragments in the serum of cancer patients. We found that pancreatic stellate cells and fibroblasts were the primary producers of collagens in the pancreas. COL1A1, COL3A1, COL5A1, COL6A1 were expressed in all CAF subtypes, whereas COL8A1, COL10A1, COL11A1, COL12A1 were specific to myofibroblast CAFs (myCAF) and COL14A1 specific to inflammatory CAFs (iCAF). In TCGA database, myCAF collagens COL10A1 and COL11A1 were elevated across solid tumor types, and multiple associations between high expression and worse survival were found. Finally, circulating collagen biomarkers were elevated in the serum of patients with cancer relative to healthy controls with COL11A1 (myCAF) having the best diagnostic accuracy of the markers measured. In conclusion, CAFs express a noncanonical collagen profile with specific collagen subtypes associated with iCAFs and myCAFs in PDAC. These collagens are deregulated at the cellular, tumor, and systemic levels across different solid tumors and associate with survival. These findings could lead to new discoveries such as novel biomarkers and therapeutic targets. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Fibroblastos Asociados al Cáncer , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Fibroblastos Asociados al Cáncer/patología , Neoplasias Pancreáticas/patología , Fibroblastos/patología , Carcinoma Ductal Pancreático/patología , Colágeno/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Microambiente Tumoral
8.
EMBO J ; 39(19): e106368, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32909627

RESUMEN

Prominent accumulation of mesenchymal cells within tumors has long been appreciated, and recent studies suggest molecular and functional diversity of stromal components. In this issue, Wu et al (2020) employed single-cell RNA sequencing of primary human breast tumors and identified new subsets of stromal mesenchymal cells with distinct transcriptional profiles, tissue localization, and associative immune microenvironments.


Asunto(s)
Neoplasias de la Mama , Células Madre Mesenquimatosas , Neoplasias de la Mama Triple Negativas , Neoplasias de la Mama/genética , Humanos , Evasión Inmune , Análisis de Secuencia de ARN , Células del Estroma , Microambiente Tumoral
9.
Dev Dyn ; 252(7): 1046-1060, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37002899

RESUMEN

BACKGROUND: Type IV collagen is an abundant component of basement membranes in all multicellular species and is essential for the extracellular scaffold supporting tissue architecture and function. Lower organisms typically have two type IV collagen genes, encoding α1 and α2 chains, in contrast with the six genes in humans, encoding α1-α6 chains. The α chains assemble into trimeric protomers, the building blocks of the type IV collagen network. The detailed evolutionary conservation of type IV collagen network remains to be studied. RESULTS: We report on the molecular evolution of type IV collagen genes. The zebrafish α4 non-collagenous (NC1) domain, in contrast with its human ortholog, contains an additional cysteine residue and lacks the M93 and K211 residues involved in sulfilimine bond formation between adjacent protomers. This may alter α4 chain interactions with other α chains, as supported by temporal and anatomic expression patterns of collagen IV chains during the zebrafish development. Despite the divergence between zebrafish and human α3 NC1 domain (endogenous angiogenesis inhibitor, Tumstatin), the zebrafish α3 NC1 domain exhibits conserved antiangiogenic activity in human endothelial cells. CONCLUSIONS: Our work supports type IV collagen is largely conserved between zebrafish and humans, with a possible difference involving the α4 chain.


Asunto(s)
Colágeno Tipo IV , Pez Cebra , Animales , Humanos , Colágeno Tipo IV/genética , Células Endoteliales , Subunidades de Proteína/análisis , Subunidades de Proteína/metabolismo , Membrana Basal/metabolismo
10.
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
12.
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.

13.
J Biol Chem ; 296: 100523, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33711340

RESUMEN

The Stimulator of Interferon Genes (STING) pathway is implicated in the innate immune response and is important in both oncogenesis and cancer treatment. Specifically, activation of the cytosolic DNA sensor STING in antigen-presenting cells (APCs) induces a type I interferon response and cytokine production that facilitates antitumor immune therapy. However, use of STING agonists (STINGa) as a cancer therapeutic has been limited by unfavorable pharmacological properties and targeting inefficiency due to rapid clearance and limited uptake into the cytosol. Exosomes, a class of extracellular vesicles shed by all cells are under consideration for their use as effective carriers of drugs owing to their innate ability to be taken up by cells and their biocompatibility for optimal drug biodistribution. Therefore, we engineered exosomes to deliver the STING agonist cyclic GMP-AMP (iExoSTINGa), to exploit their favorable pharmacokinetics and pharmacodynamics. Selective targeting of the STING pathway in APCs with iExoSTINGa was associated with superior potency compared with STINGa alone in suppressing B16F10 tumor growth. Moreover, iExoSTINGa showed superior uptake of STINGa into dendritic cells compared with STINGa alone, which led to increased accumulation of activated CD8+ T-cells and an antitumor immune response. Our study highlights the potential of exosomes in general, and iExoSTINGa specifically, in enhancing cancer therapy outcomes.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Exosomas/metabolismo , Inmunidad Innata/inmunología , Melanoma Experimental/inmunología , Proteínas de la Membrana/agonistas , Nucleótidos Cíclicos/farmacología , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Femenino , Células HEK293 , Humanos , Inmunidad Innata/efectos de los fármacos , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Melanoma Experimental/prevención & control , Ratones , Ratones Endogámicos C57BL , Transducción de Señal
14.
Anal Chem ; 94(28): 10045-10053, 2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35792073

RESUMEN

The phosphonate group is a key pharmacophore in many antiviral, antimicrobial, and antineoplastic drugs. Due to its high polarity and short retention time, detecting and quantifying such phosphonate-containing drugs with LC/MS-based methods are challenging and require derivatization with hazardous reagents. Given the emerging importance of phosphonate-containing drugs, developing a practical, accessible, and safe method for their quantitation in pharmacokinetics (PK) studies is desirable. NMR-based methods are often employed in drug discovery but are seldom used for compound quantitation in PK studies. Here, we show that proton-phosphorous (1H-31P) heteronuclear single quantum correlation (HSQC) NMR allows for the quantitation of the phosphonate-containing enolase inhibitor HEX in plasma and tissues at micromolar concentrations. Although mice were shown to rapidly clear HEX from circulation (over 95% in <1 h), the plasma half-life of HEX was more than 1 h in rats and nonhuman primates. This slower clearance rate affords a significantly higher exposure of HEX in rat models compared to that in mouse models while maintaining a favorable safety profile. Similar results were observed for the phosphonate-containing antibiotic, fosfomycin. Our study demonstrates the applicability of the 1H-31P HSQC method to quantify phosphonate-containing drugs in complex biological samples and illustrates an important limitation of mice as preclinical model species for phosphonate-containing drugs.


Asunto(s)
Antineoplásicos , Organofosfonatos , Animales , Antineoplásicos/farmacocinética , Antivirales , Ratones , Organofosfonatos/química , Primates , Protones , Ratas
15.
FASEB J ; 35(5): e21557, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33855751

RESUMEN

Hepatic fibrosis is a wound healing response that results in excessive extracellular matrix (ECM) accumulation in response to chronic hepatic injury. Signal transducer and activator of transcription 3 (STAT3) is an important transcription factor associated with the pathogenesis of liver fibrosis. Though a promising potential therapeutic target, there are no specific drug candidates for STAT3. Exosomes are extracellular vesicles generated by all cell types with a capacity to efficiently enter cells across different biological barriers. Here, we utilize exosomes as delivery conduit to specifically target STAT3 in liver fibrosis. Exosomes derived from clinical grade fibroblast-like mesenchymal stem cells (MSCs) were engineered to carry siRNA or antisense oligonucleotide (ASO) targeting STAT3 (iExosiRNA-STAT3 or iExomASO-STAT3 ). Compared to scrambled siRNA control, siRNA-STAT3, or ASO-STAT3, iExosiRNA-STAT3 or iExomASO-STAT3 showed enhanced STAT3 targeting efficiency. iExosiRNA-STAT3 or iExomASO-STAT3 treatments suppressed STAT3 levels and ECM deposition in established liver fibrosis in mice, and significantly improved liver function. iExomASO-Stat3 restored liver function more efficiently when compared to iExosiRNA-STAT3 . Our results identify a novel anti-fibrotic approach for direct targeting of STAT3 with exosomes with immediate translational potential.


Asunto(s)
Exosomas/genética , Regulación de la Expresión Génica , Cirrosis Hepática/terapia , Oligonucleótidos Antisentido/farmacología , ARN Interferente Pequeño/genética , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Tetracloruro de Carbono/toxicidad , Femenino , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Ratones , Ratones Endogámicos BALB C , Transducción de Señal
16.
PLoS Biol ; 16(12): e2005907, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30592710

RESUMEN

Metastatic dissemination employs both the blood and lymphatic vascular systems. Solid tumors dynamically remodel and generate both vessel types during cancer progression. Lymphatic vessel invasion and cancer cells in the tumor-draining lymph nodes (LNs) are prognostic markers for breast cancer metastasis and patient outcome, and tumor-induced lymphangiogenesis likely influences metastasis. Deregulated tumor tissue fluid homeostasis and immune trafficking associated with tumor lymphangiogenesis may contribute to metastatic spreading; however, the precise functional characterization of lymphatic endothelial cells (LECs) in tumors is challenged by the lack of specific reagents to decipher their rate-limiting role in metastasis. Therefore, we generated novel transgenic mice (PDPN promoter-driven Cre recombinase transgene [PDPN-Cre] and PDPN promoter-driven thymidine kinase transgene [PDPN-tk]) that allow for the identification and genetically controlled depletion of proliferating podoplanin (Pdpn)-expressing LECs. We demonstrate that suppression of lymphangiogenesis is successfully achieved in lymphangioma lesions induced in the PDPN-tk mice. In multiple metastatic breast cancer mouse models, we identified distinct roles for LECs in primary and metastatic tumors. Our findings support the functional contribution of primary tumor lymphangiogenesis in controlling metastasis to axillary LNs and lung parenchyma. Reduced lymphatic vessel density enhanced primary tumor lymphedema and increased the frequency of intratumoral macrophages but was not associated with a significant impact on primary tumor growth despite a marked reduction in metastatic dissemination. Our findings identify the rate-limiting contribution of the breast tumor lymphatic vessels for lung metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Glicoproteínas de Membrana/fisiología , Animales , Neoplasias de la Mama/fisiopatología , Movimiento Celular , Células Endoteliales/patología , Células Endoteliales/fisiología , Femenino , Humanos , Ganglios Linfáticos/patología , Linfangiogénesis/genética , Linfangiogénesis/fisiología , Sistema Linfático/fisiología , Vasos Linfáticos/patología , Macrófagos , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Metástasis de la Neoplasia/fisiopatología , Timidina Quinasa/genética
17.
Nature ; 527(7579): 525-530, 2015 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-26560028

RESUMEN

Diagnosis of pancreatic ductal adenocarcinoma (PDAC) is associated with a dismal prognosis despite current best therapies; therefore new treatment strategies are urgently required. Numerous studies have suggested that epithelial-to-mesenchymal transition (EMT) contributes to early-stage dissemination of cancer cells and is pivotal for invasion and metastasis of PDAC. EMT is associated with phenotypic conversion of epithelial cells into mesenchymal-like cells in cell culture conditions, although such defined mesenchymal conversion (with spindle-shaped morphology) of epithelial cells in vivo is rare, with quasi-mesenchymal phenotypes occasionally observed in the tumour (partial EMT). Most studies exploring the functional role of EMT in tumours have depended on cell-culture-induced loss-of-function and gain-of-function experiments involving EMT-inducing transcription factors such as Twist, Snail and Zeb1 (refs 2, 3, 7-10). Therefore, the functional contribution of EMT to invasion and metastasis remains unclear, and genetically engineered mouse models to address a causal connection are lacking. Here we functionally probe the role of EMT in PDAC by generating mouse models of PDAC with deletion of Snail or Twist, two key transcription factors responsible for EMT. EMT suppression in the primary tumour does not alter the emergence of invasive PDAC, systemic dissemination or metastasis. Suppression of EMT leads to an increase in cancer cell proliferation with enhanced expression of nucleoside transporters in tumours, contributing to enhanced sensitivity to gemcitabine treatment and increased overall survival of mice. Collectively, our study suggests that Snail- or Twist-induced EMT is not rate-limiting for invasion and metastasis, but highlights the importance of combining EMT inhibition with chemotherapy for the treatment of pancreatic cancer.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Transición Epitelial-Mesenquimal , Metástasis de la Neoplasia/patología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Proliferación Celular/efectos de los fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Masculino , Ratones , Invasividad Neoplásica/patología , Proteínas de Transporte de Nucleósidos/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Factores de Transcripción de la Familia Snail , Análisis de Supervivencia , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína 1 Relacionada con Twist/deficiencia , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo , Gemcitabina
18.
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
19.
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
20.
Cancer Sci ; 111(9): 3100-3110, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32639675

RESUMEN

Mesenchymal stem cells (MSC) are multipotent stromal cells with the potential to differentiate into several cell types. MSC-based therapy has emerged as a promising strategy for various diseases. Accumulating evidence suggests that the paracrine effects of MSC are partially exerted by the secretion of soluble factors, in particular exosomes. MSC-derived exosomes are involved in intercellular communication through transfer of proteins, RNA, DNA and bioactive lipids, which might constitute a novel intercellular communication mode. This review illustrates the current knowledge on the composition and biological functions as well as the therapeutic potential of MSC-derived exosomes in cancer, with a focus on clinical translation opportunities.


Asunto(s)
Productos Biológicos/uso terapéutico , Micropartículas Derivadas de Células/metabolismo , Exosomas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Comunicación Celular , Microambiente Celular , Ensayos Clínicos como Asunto , Sistemas de Liberación de Medicamentos , Humanos , Inmunomodulación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA