Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell ; 185(16): 2918-2935.e29, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35803260

RESUMEN

Neoadjuvant immune checkpoint blockade has shown promising clinical activity. Here, we characterized early kinetics in tumor-infiltrating and circulating immune cells in oral cancer patients treated with neoadjuvant anti-PD-1 or anti-PD-1/CTLA-4 in a clinical trial (NCT02919683). Tumor-infiltrating CD8 T cells that clonally expanded during immunotherapy expressed elevated tissue-resident memory and cytotoxicity programs, which were already active prior to therapy, supporting the capacity for rapid response. Systematic target discovery revealed that treatment-expanded tumor T cell clones in responding patients recognized several self-antigens, including the cancer-specific antigen MAGEA1. Treatment also induced a systemic immune response characterized by expansion of activated T cells enriched for tumor-infiltrating T cell clonotypes, including both pre-existing and emergent clonotypes undetectable prior to therapy. The frequency of activated blood CD8 T cells, notably pre-treatment PD-1-positive KLRG1-negative T cells, was strongly associated with intra-tumoral pathological response. These results demonstrate how neoadjuvant checkpoint blockade induces local and systemic tumor immunity.


Asunto(s)
Neoplasias , Receptor de Muerte Celular Programada 1 , Linfocitos T CD8-positivos , Humanos , Inmunoterapia , Linfocitos Infiltrantes de Tumor , Terapia Neoadyuvante , Neoplasias/terapia , Microambiente Tumoral
2.
Sci Rep ; 6: 35298, 2016 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-27748415

RESUMEN

During tumor progression, alternative splicing gives rise to different Mena protein isoforms. We analyzed how Mena11a, an isoform enriched in epithelia and epithelial-like cells, affects Mena-dependent regulation of actin dynamics and cell behavior. While other Mena isoforms promote actin polymerization and drive membrane protrusion, we find that Mena11a decreases actin polymerization and growth factor-stimulated membrane protrusion at lamellipodia. Ectopic Mena11a expression slows mesenchymal-like cell motility, while isoform-specific depletion of endogenous Mena11a in epithelial-like tumor cells perturbs cell:cell junctions and increases membrane protrusion and overall cell motility. Mena11a can dampen membrane protrusion and reduce actin polymerization in the absence of other Mena isoforms, indicating that it is not simply an inactive Mena isoform. We identify a phosphorylation site within 11a that is required for some Mena11a-specific functions. RNA-seq data analysis from patient cohorts demonstrates that the difference between mRNAs encoding constitutive Mena sequences and those containing the 11a exon correlates with metastasis in colorectal cancer, suggesting that 11a exon exclusion contributes to invasive phenotypes and leads to poor clinical outcomes.


Asunto(s)
Citoesqueleto de Actina , Neoplasias Colorrectales/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Empalme Alternativo , Animales , Biomarcadores/metabolismo , Adhesión Celular , Comunicación Celular , Membrana Celular/metabolismo , Movimiento Celular , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Receptores ErbB/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Humanos , Pulmón/embriología , Pulmón/metabolismo , Células MCF-7 , Ratones , Fenotipo , Fosforilación , Seudópodos/patología , Alveolos Pulmonares/metabolismo , Piel/embriología , Piel/metabolismo , Resultado del Tratamiento , Regulación hacia Arriba , Cicatrización de Heridas
3.
Mol Biol Cell ; 27(20): 3085-3094, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27559126

RESUMEN

Directed cell migration, a key process in metastasis, arises from the combined influence of multiple processes, including chemotaxis-the directional movement of cells to soluble cues-and haptotaxis-the migration of cells on gradients of substrate-bound factors. However, it is unclear how chemotactic and haptotactic pathways integrate with each other to drive overall cell behavior. MenaINV has been implicated in metastasis by driving chemotaxis via dysregulation of phosphatase PTP1B and more recently in haptotaxis via interaction with integrin α5ß1. Here we find that MenaINV-driven haptotaxis on fibronectin (FN) gradients requires intact signaling between α5ß1 integrin and the epidermal growth factor receptor (EGFR), which is influenced by PTP1B. Furthermore, we show that MenaINV-driven haptotaxis and ECM reorganization both require the Rab-coupling protein RCP, which mediates α5ß1 and EGFR recycling. Finally, MenaINV promotes synergistic migratory response to combined EGF and FN in vitro and in vivo, leading to hyperinvasive phenotypes. Together our data demonstrate that MenaINV is a shared component of multiple prometastatic pathways that amplifies their combined effects, promoting synergistic cross-talk between RTKs and integrins.


Asunto(s)
Quimiotaxis/fisiología , Proteínas del Citoesqueleto/metabolismo , Animales , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular/fisiología , Proteínas del Citoesqueleto/fisiología , Receptores ErbB/metabolismo , Integrina alfa5beta1/metabolismo , Integrinas , Ratones , Proteínas de Microfilamentos/metabolismo , Metástasis de la Neoplasia/fisiopatología , Fosfoproteínas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Receptor Cross-Talk , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal/fisiología , Células Tumorales Cultivadas
4.
Cancer Discov ; 6(4): 382-99, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26984351

RESUMEN

UNLABELLED: Kinase inhibitor resistance often involves upregulation of poorly understood "bypass" signaling pathways. Here, we show that extracellular proteomic adaptation is one path to bypass signaling and drug resistance. Proteolytic shedding of surface receptors, which can provide negative feedback on signaling activity, is blocked by kinase inhibitor treatment and enhances bypass signaling. In particular, MEK inhibition broadly decreases shedding of multiple receptor tyrosine kinases (RTK), including HER4, MET, and most prominently AXL, an ADAM10 and ADAM17 substrate, thus increasing surface RTK levels and mitogenic signaling. Progression-free survival of patients with melanoma treated with clinical BRAF/MEK inhibitors inversely correlates with RTK shedding reduction following treatment, as measured noninvasively in blood plasma. Disrupting protease inhibition by neutralizing TIMP1 improves MAPK inhibitor efficacy, and combined MAPK/AXL inhibition synergistically reduces tumor growth and metastasis in xenograft models. Altogether, extracellular proteomic rewiring through reduced RTK shedding represents a surprising mechanism for bypass signaling in cancer drug resistance. SIGNIFICANCE: Genetic, epigenetic, and gene expression alterations often fail to explain adaptive drug resistance in cancer. This work presents a novel post-translational mechanism of such resistance: Kinase inhibitors, particularly targeting MAPK signaling, increase tumor cell surface receptor levels due to widely reduced proteolysis, allowing tumor signaling to circumvent intended drug action.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional , Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Línea Celular Tumoral , Membrana Celular/metabolismo , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/metabolismo , Melanoma/mortalidad , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Modelos Biológicos , Fosforilación , Proteolisis , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-jun , Proteínas Tirosina Quinasas Receptoras/sangre , Proteínas Tirosina Quinasas Receptoras/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Tirosina Quinasa del Receptor Axl
5.
Mol Biol Cell ; 26(21): 3867-78, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26337385

RESUMEN

During breast cancer progression, alternative mRNA splicing produces functionally distinct isoforms of Mena, an actin regulator with roles in cell migration and metastasis. Aggressive tumor cell subpopulations express Mena(INV), which promotes tumor cell invasion by potentiating EGF responses. However, the mechanism by which this occurs is unknown. Here we report that Mena associates constitutively with the tyrosine phosphatase PTP1B and mediates a novel negative feedback mechanism that attenuates receptor tyrosine kinase signaling. On EGF stimulation, complexes containing Mena and PTP1B are recruited to the EGFR, causing receptor dephosphorylation and leading to decreased motility responses. Mena also interacts with the 5' inositol phosphatase SHIP2, which is important for the recruitment of the Mena-PTP1B complex to the EGFR. When Mena(INV) is expressed, PTP1B recruitment to the EGFR is impaired, providing a mechanism for growth factor sensitization to EGF, as well as HGF and IGF, and increased resistance to EGFR and Met inhibitors in signaling and motility assays. In sum, we demonstrate that Mena plays an important role in regulating growth factor-induced signaling. Disruption of this attenuation by Mena(INV) sensitizes tumor cells to low-growth factor concentrations, thereby increasing the migration and invasion responses that contribute to aggressive, malignant cell phenotypes.


Asunto(s)
Proteínas de Microfilamentos/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Actinas/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Adhesión Celular/fisiología , Movimiento Celular/efectos de los fármacos , Proteínas del Citoesqueleto , Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Femenino , Humanos , Metástasis de la Neoplasia , Fosforilación , Isoformas de Proteínas , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal/efectos de los fármacos
6.
J Cell Biol ; 198(4): 657-76, 2012 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-22908313

RESUMEN

Mena is an Ena/VASP family actin regulator with roles in cell migration, chemotaxis, cell-cell adhesion, tumor cell invasion, and metastasis. Although enriched in focal adhesions, Mena has no established function within these structures. We find that Mena forms an adhesion-regulated complex with α5ß1 integrin, a fibronectin receptor involved in cell adhesion, motility, fibronectin fibrillogenesis, signaling, and growth factor receptor trafficking. Mena bound directly to the carboxy-terminal portion of the α5 cytoplasmic tail via a 91-residue region containing 13 five-residue "LERER" repeats. In fibroblasts, the Mena-α5 complex was required for "outside-in" α5ß1 functions, including normal phosphorylation of FAK and paxillin and formation of fibrillar adhesions. It also supported fibrillogenesis and cell spreading and controlled cell migration speed. Thus, fibroblasts require Mena for multiple α5ß1-dependent processes involving bidirectional interactions between the extracellular matrix and cytoplasmic focal adhesion proteins.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Adhesiones Focales/fisiología , Integrina alfa5/metabolismo , Integrina alfa5beta1/metabolismo , Transducción de Señal/fisiología , Animales , Proteínas del Citoesqueleto/genética , Matriz Extracelular/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/enzimología , Fibroblastos/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Ratones , Ratones Mutantes , Proteínas de Microfilamentos , Células 3T3 NIH , Embarazo , Transporte de Proteínas/fisiología , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...