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










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 117(21): 11432-11443, 2020 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-32381732

RESUMO

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.


Assuntos
Glioblastoma/patologia , Receptores de Hialuronatos/metabolismo , Actinas/metabolismo , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Feminino , Técnicas de Inativação de Genes , Glioblastoma/metabolismo , Humanos , Ácido Hialurônico/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Miosinas/metabolismo , Proteínas de Neoplasias/metabolismo , Oligopeptídeos/metabolismo , Técnicas de Cultura de Órgãos , Proteínas Ativadoras de ras GTPase/genética , Proteínas Ativadoras de ras GTPase/metabolismo
2.
Cancer Res ; 80(1): 69-78, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31641031

RESUMO

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.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Quinases Lim/metabolismo , Animais , Encéfalo/patologia , Encéfalo/cirurgia , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/cirurgia , Linhagem Celular Tumoral , Quimiotaxia , Conjuntos de Dados como Assunto , Progressão da Doença , Feminino , Técnicas de Silenciamento de Genes , Glioblastoma/mortalidade , Glioblastoma/cirurgia , Humanos , Estimativa de Kaplan-Meier , Quinases Lim/genética , Masculino , Camundongos , Gradação de Tumores , Invasividade Neoplásica/patologia , Cultura Primária de Células , Prognóstico , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Fatores de Tempo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...