Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Biophys J ; 120(13): 2644-2656, 2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34087211

RESUMEN

The leukocyte-specific ß2-integrin LFA-1 and its ligand ICAM-1, expressed on endothelial cells (ECs), are involved in the arrest, adhesion, and transendothelial migration of leukocytes. Although the role of mechanical forces on LFA-1 activation is well established, the impact of forces on its major ligand ICAM-1 has received less attention. Using a parallel-plate flow chamber combined with confocal and super-resolution microscopy, we show that prolonged shear flow induces global translocation of ICAM-1 on ECs upstream of flow direction. Interestingly, shear forces caused actin rearrangements and promoted actin-dependent ICAM-1 nanoclustering before LFA-1 engagement. T cells adhered to mechanically prestimulated ECs or nanoclustered ICAM-1 substrates developed a promigratory phenotype, migrated faster, and exhibited shorter-lived interactions with ECs than when adhered to non mechanically stimulated ECs or to monomeric ICAM-1 substrates. Together, our results indicate that shear forces increase ICAM-1/LFA-1 bonds because of ICAM-1 nanoclustering, strengthening adhesion and allowing cells to exert higher traction forces required for faster migration. Our data also underscore the importance of mechanical forces regulating the nanoscale organization of membrane receptors and their contribution to cell adhesion regulation.


Asunto(s)
Células Endoteliales , Molécula 1 de Adhesión Intercelular , Adhesión Celular , Movimiento Celular , Antígeno-1 Asociado a Función de Linfocito
2.
STAR Protoc ; 1(2): 100103, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-33000005

RESUMEN

Here, we describe a protocol to detect and visualize protein synthesis by click-chemistry-based immunofluorescence in patient-derived organoids (PDOs) in vitro. The protocol uses O-propargyl puromycin (OPP), an analog of puromycin that enters the acceptor site of ribosomes and is incorporated into nascent polypeptides. OPP can be detected by a click chemistry reaction and can be combined with conventional antibody staining. We describe procedures for imaging intact organoids in 3D format or imaging sections of organoids from paraffin blocks. For complete details on the use and execution of this protocol, please refer to Morral, Stanisavljevic et al. (2020).


Asunto(s)
Química Clic/métodos , Técnica del Anticuerpo Fluorescente/métodos , Proteínas/análisis , Puromicina/análogos & derivados , Técnicas de Cultivo de Célula/métodos , Neoplasias Colorrectales/metabolismo , Humanos , Organoides/metabolismo , Biosíntesis de Proteínas/fisiología , Puromicina/química , Ribosomas/metabolismo , Coloración y Etiquetado/métodos
3.
Cell Stem Cell ; 26(6): 845-861.e12, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32396863

RESUMEN

Colorectal cancers (CRCs) are composed of an amalgam of cells with distinct genotypes and phenotypes. Here, we reveal a previously unappreciated heterogeneity in the biosynthetic capacities of CRC cells. We discover that the majority of ribosomal DNA transcription and protein synthesis in CRCs occurs in a limited subset of tumor cells that localize in defined niches. The rest of the tumor cells undergo an irreversible loss of their biosynthetic capacities as a consequence of differentiation. Cancer cells within the biosynthetic domains are characterized by elevated levels of the RNA polymerase I subunit A (POLR1A). Genetic ablation of POLR1A-high cell population imposes an irreversible growth arrest on CRCs. We show that elevated biosynthesis defines stemness in both LGR5+ and LGR5- tumor cells. Therefore, a common architecture in CRCs is a simple cell hierarchy based on the differential capacity to transcribe ribosomal DNA and synthesize proteins.


Asunto(s)
Neoplasias Colorrectales , Células Madre Neoplásicas , Línea Celular Tumoral , Neoplasias Colorrectales/genética , ADN Ribosómico , Humanos , Receptores Acoplados a Proteínas G
4.
Int J Cancer ; 145(11): 3064-3077, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31032902

RESUMEN

Myofibroblasts are a population of highly contractile fibroblasts that express and require the activity of the transcription factor Snail1. Cancer-associated fibroblasts (CAFs) correlate with low survival of cancer patients when present in the stroma of primary tumors. Remarkably, the presence of myofibroblastic CAFs (which express Snail1) creates mechanical properties in the tumor microenvironment that support metastasis. However, therapeutic blockage of fibroblast activity in patients with cancer is a double-edged sword, as normal fibroblast activities often restrict tumor cell invasion. We used fibroblasts depleted of Snail1 or protein arginine methyltransferases 1 and 4 (PRMT1/-4) to identify specific epigenetic modifications induced by TGFß/Snail1. Furthermore, we analyzed the in vivo efficiency of methyltransferase inhibitors using mouse models of wound healing and metastasis, as well as fibroblasts isolated from patients with idiopathic pulmonary fibrosis (IPF). Mechanistically, TGFß-induced Snail1 promotes the epigenetic mark of asymmetrically dimethylated arginine. Critically, we found that inhibitors of methyltransferases prevent myofibroblast activity (but not regular fibroblast activity) in the extracellular matrix, both in cell culture and in vivo. In a mouse breast cancer model, the inhibitor sinefungin reduces both the myofibroblast activity in the tumor stroma and the metastatic burden in the lung. Two distinct inhibitors effectively blocked the exacerbated myofibroblast activity of patient-derived IPF fibroblasts. Our data reveal epigenetic regulation of myofibroblast transdifferentiation in both wound healing and in disease (fibrosis and breast cancer). Thus, methyltransferase inhibitors are good candidates as therapeutic reagents for these diseases.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Inhibidores Enzimáticos/administración & dosificación , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Metiltransferasas/antagonistas & inhibidores , Miofibroblastos/efectos de los fármacos , Factores de Transcripción de la Familia Snail/genética , Adenosina/administración & dosificación , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Neoplasias de la Mama/enzimología , Fibroblastos Asociados al Cáncer/citología , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Transdiferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Epigénesis Genética , Femenino , Eliminación de Gen , Humanos , Fibrosis Pulmonar Idiopática/enzimología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/enzimología , Metiltransferasas/genética , Ratones , Miofibroblastos/citología , Miofibroblastos/enzimología , Factores de Transcripción de la Familia Snail/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cancer Res ; 75(2): 284-95, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25488750

RESUMEN

Crosstalk between tumor and stromal cells in the tumor microenvironment alter its properties in ways that facilitate the invasive behavior of tumor cells. Here, we demonstrate that cancer-associated fibroblasts (CAF) increase the stiffness of the extracellular matrix (ECM) and promote anisotropic fiber orientation, two mechanical signals generated through a Snail1/RhoA/αSMA-dependent mechanism that sustains oriented tumor cell migration and invasiveness. Snail1-depleted CAF failed to acquire myofibroblastic traits in response to TGFß, including RhoA activation, αSMA-positive stress fibers, increased fibronectin fibrillogenesis, and production of a stiff ECM with oriented fibers. Snail1 expression in human tumor-derived CAF was associated with an ability to organize the ECM. In coculture, a relatively smaller number of Snail1-expressing CAF were capable of imposing an anisotropic ECM architecture, compared with nonactivated fibroblasts. Pathologically, human breast cancers with Snail1(+) CAF tended to exhibit desmoplastic areas with anisotropic fibers, lymph node involvement, and poorer outcomes. Snail1 involvement in driving an ordered ECM was further confirmed in wound-healing experiments in mice, with Snail1 depletion preventing the anisotropic organization of granulation tissue and delaying wound healing. Overall, our results showed that inhibiting Snail1 function in CAF could prevent tumor-driven ECM reorganization and cancer invasion.


Asunto(s)
Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Factores de Transcripción/biosíntesis , Animales , Humanos , Ratones , Ratones Noqueados , Metástasis de la Neoplasia , Factores de Transcripción de la Familia Snail , Microambiente Tumoral , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
6.
Biochem J ; 435(3): 563-8, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21361876

RESUMEN

Fibronectins are cell-secreted glycoproteins that modulate cell attachment, spreading, migration, morphology, differentiation and oncogenic transformation. Fibronectin expression is activated during EMT (epithelial-mesenchymal transition) and is a hallmark of mesenchymal cells. It is shown in the present study that a transcription factor previously unrelated with EMT, TFCP2c/LSF/LBP-1c, was translocated to the nucleus and bound to the fibronectin promoter upon EMT induction by Snail1. Consequently, the interference of TFCP2c/LSF/LBP-1c's activity prevented fibronectin expression. Moreover, TFCP2c/LSF/LBP-1c was detected in nuclei of embryonic dermal mesenchymal cells adjacent to the hair bud, a cell population that expresses endogenous nuclear Snail1 and fibronectin. Therefore we indicate a new molecular role for TFCP2c/LSF/LBP-1c in fibronectin expression.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fibronectinas/metabolismo , Regulación de la Expresión Génica/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Línea Celular , Embrión de Mamíferos/metabolismo , Fibronectinas/genética , Humanos , Ratones , Regiones Promotoras Genéticas , Unión Proteica , Transporte de Proteínas/fisiología , Factores de Transcripción de la Familia Snail
7.
J Cell Sci ; 124(Pt 24): 4161-71, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22223884

RESUMEN

Snail1 is a transcriptional repressor of E-cadherin that triggers epithelial-mesenchymal transition (EMT). Here, we report assisted Snail1 interaction with the promoter of a typical mesenchymal gene, fibronectin (FN1), both in epithelial cells undergoing EMT and in fibroblasts. Together with Snail1, the p65 subunit of NF-κB and PARP1 bound to the FN1 promoter. We detected nuclear interaction of these proteins and demonstrated the requirement of all three for FN1 transcription. Moreover, other genes involved in cell movement mimic FN1 expression induced by Snail1 or TGF-ß1 treatment and recruit p65NF-κB and Snail1 to their promoters. The molecular cooperation between Snail1 and NF-κB in transcription activation provides a new insight into how Snail1 can modulate a variety of cell programs.


Asunto(s)
Fibronectinas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Factor de Transcripción ReIA/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional , Animales , Sitios de Unión , Línea Celular , Núcleo Celular/metabolismo , Proteínas de la Matriz Extracelular/genética , Fibronectinas/biosíntesis , Humanos , Mesodermo/citología , Ratones , Poli(ADP-Ribosa) Polimerasa-1 , Regiones Promotoras Genéticas , Factores de Transcripción de la Familia Snail
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA