Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 117(21): 11432-11443, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32381732

RESUMEN

The structure and mechanics of many connective tissues are dictated by a collagen-rich extracellular matrix (ECM), where collagen fibers provide topological cues that direct cell migration. However, comparatively little is known about how cells navigate the hyaluronic acid (HA)-rich, nanoporous ECM of the brain, a problem with fundamental implications for development, inflammation, and tumor invasion. Here, we demonstrate that glioblastoma cells adhere to and invade HA-rich matrix using microtentacles (McTNs), which extend tens of micrometers from the cell body and are distinct from filopodia. We observe these structures in continuous culture models and primary patient-derived tumor cells, as well as in synthetic HA matrix and organotypic brain slices. High-magnification and superresolution imaging reveals McTNs are dynamic, CD44-coated tubular protrusions containing microtubules and actin filaments, which respectively drive McTN extension and retraction. Molecular mechanistic studies reveal that McTNs are stabilized by an interplay between microtubule-driven protrusion, actomyosin-driven retraction, and CD44-mediated adhesion, where adhesive and cytoskeletal components are mechanistically coupled by an IQGAP1-CLIP170 complex. McTNs represent a previously unappreciated mechanism through which cells engage nanoporous HA matrix and may represent an important molecular target in physiology and disease.


Asunto(s)
Glioblastoma/patología , Receptores de Hialuranos/metabolismo , Actinas/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Femenino , Técnicas de Inactivación de Genes , Glioblastoma/metabolismo , Humanos , Ácido Hialurónico/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Miosinas/metabolismo , Proteínas de Neoplasias/metabolismo , Oligopéptidos/metabolismo , Técnicas de Cultivo de Órganos , Proteínas Activadoras de ras GTPasa/genética , Proteínas Activadoras de ras GTPasa/metabolismo
2.
Cancer Res ; 80(1): 69-78, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31641031

RESUMEN

The aggressive brain tumor glioblastoma (GBM) is characterized by rapid cellular infiltration of brain tissue, raising the possibility that disease progression could potentially be slowed by disrupting the machinery of cell migration. The LIM kinase isoforms LIMK1 and LIMK2 (LIMK1/2) play important roles in cell polarization, migration, and invasion and are markedly upregulated in GBM and many other infiltrative cancers. Yet, it remains unclear whether LIMK suppression could serve as a viable basis for combating GBM infiltration. In this study, we investigated effects of LIMK1/2 suppression on GBM invasion by combining GBM culture models, engineered invasion paradigms, and mouse xenograft models. While knockdown of either LIMK1 or LIMK2 only minimally influenced invasion in culture, simultaneous knockdown of both isoforms strongly reduced the invasive motility of continuous culture models and human GBM tumor-initiating cells (TIC) in both Boyden chamber and 3D hyaluronic acid spheroid invasion assays. Furthermore, LIMK1/2 functionally regulated cell invasiveness, in part, by disrupting polarized cell motility under confinement and cell chemotaxis. In an orthotopic xenograft model, TICs stably transduced with LIMK1/2 shRNA were implanted intracranially in immunocompromised mice. Tumors derived from LIMK1/2 knockdown TICs were substantially smaller and showed delayed growth kinetics and more distinct margins than tumors derived from control TICs. Overall, LIMK1/2 suppression increased mean survival time by 30%. These findings indicate that LIMK1/2 strongly regulate GBM invasive motility and tumor progression and support further exploration of LIMK1/2 as druggable targets. SIGNIFICANCE: Targeting the actin-binding proteins LIMK1 and LIMK2 significantly diminishes glioblastoma invasion and spread, suggesting the potential value of these proteins as therapeutic targets.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Quinasas Lim/metabolismo , Animales , Encéfalo/patología , Encéfalo/cirugía , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/cirugía , Línea Celular Tumoral , Quimiotaxis , Conjuntos de Datos como Asunto , Progresión de la Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Glioblastoma/mortalidad , Glioblastoma/cirugía , Humanos , Estimación de Kaplan-Meier , Quinasas Lim/genética , Masculino , Ratones , Clasificación del Tumor , Invasividad Neoplásica/patología , Cultivo Primario de Células , Pronóstico , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Factores de Tiempo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA