Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Sci Transl Med ; 14(661): eaax8933, 2022 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-36070364

RESUMEN

Brain metastasis is a complication of increasing incidence in patients with breast cancer at advanced disease stage. It is a severe condition characterized by a rapid decline in quality of life and poor prognosis. There is a critical clinical need to develop effective therapies to prevent and treat brain metastases. Here, we describe a unique and robust spontaneous preclinical model of breast cancer metastasis to the brain (4T1-BM2) in mice that has been instrumental in uncovering molecular mechanisms guiding metastatic dissemination and colonization of the brain. Key experimental findings were validated in the additional murine D2A1-BM2 model and in human MDA231-BrM2 model. Gene expression analyses and functional studies, coupled with clinical transcriptomic and histopathological investigations, identified connexins (Cxs) and focal adhesion kinase (FAK) as master molecules orchestrating breast cancer colonization of the brain. Cx31 promoted homotypic tumor cell adhesion, heterotypic tumor-astrocyte interaction, and FAK phosphorylation. FAK signaling prompted NF-κB activation inducing Lamc2 expression and laminin 332 (laminin 5) deposition, α6 integrin-mediated adhesion, and sustained survival and growth within brain parenchyma. In the MDA231-BrM2 model, the human homologous molecules CX43, LAMA4, and α3 integrin were involved. Systemic treatment with FAK inhibitors reduced brain metastasis progression. In conclusion, we report a spontaneous model of breast cancer metastasis to the brain and identified Cx-mediated FAK-NF-κB signaling as a mechanism promoting cell-autonomous and microenvironmentally controlled cell survival for brain colonization. Considering the limited therapeutic options for brain metastatic disease in cancer patients, we propose FAK as a therapeutic candidate to further pursue in the clinic.


Asunto(s)
Neoplasias Encefálicas , Neoplasias de la Mama , Animales , Encéfalo/metabolismo , Neoplasias de la Mama/genética , Conexinas/metabolismo , Femenino , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Melanoma , Ratones , FN-kappa B/metabolismo , Calidad de Vida , Neoplasias Cutáneas , Melanoma Cutáneo Maligno
2.
Cancer Res ; 81(3): 594-605, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33526470

RESUMEN

Early detection and adjuvant therapies have significantly improved survival of patients with breast cancer over the past three decades. In contrast, management of metastatic disease remains unresolved. Brain metastasis is a late complication frequently observed among patients with metastatic breast cancer, whose poor prognosis calls for novel and more effective therapies. Here, we report that active hypoxia inducible factor-1 (HIF1) signaling and loss of the miRNA let-7d concur to promote brain metastasis in a recently established model of spontaneous breast cancer metastasis from the primary site to the brain (4T1-BM2), and additionally in murine and human experimental models of breast cancer brain metastasis (D2A1-BM2 and MDA231-BrM2). Active HIF1 and let-7d loss upregulated expression of platelet-derived growth factor (PDGF) B/A in murine and human brain metastatic cells, respectively, while either individual silencing of HIF1α and PDGF-A/B or let-7d overexpression suppressed brain metastasis formation in the tested models. Let-7d silencing upregulated HIF1α expression and HIF1 activity, indicating a regulatory hierarchy of the system. The clinical relevance of the identified targets was supported by human gene expression data analyses. Treatment of mice with nilotinib, a kinase inhibitor impinging on PDGF receptor (PDGFR) signaling, prevented formation of spontaneous brain metastases in the 4T1-BM2 model and reduced growth of established brain metastases in mouse and human models. These results identify active HIF1 signaling and let-7d loss as coordinated events promoting breast cancer brain metastasis through increased expression of PDGF-A/B. Moreover, they identify PDGFR inhibition as a potentially actionable therapeutic strategy for patients with brain metastatis. SIGNIFICANCE: These findings show that loss of miRNA let-7d and active HIF1 signaling promotes breast cancer brain metastasis via PDGF and that pharmacologic inhibition of PDGFR suppresses brain metastasis, suggesting novel therapeutic opportunities. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/3/594/F1.large.jpg.See related article by Thies et al., p. 606.


Asunto(s)
Neoplasias de la Mama , MicroARNs , Animales , Encéfalo , Neoplasias de la Mama/genética , Línea Celular Tumoral , Humanos , Factor 1 Inducible por Hipoxia , Ratones , MicroARNs/genética , Factor de Crecimiento Derivado de Plaquetas/genética
3.
Nat Commun ; 10(1): 5755, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31848340

RESUMEN

Autophagy perturbation represents an emerging therapeutic strategy in cancer. Although LATS1 and LATS2 kinases, core components of the mammalian Hippo pathway, have been shown to exert tumor suppressive activities, here we report a pro-survival role of LATS1 but not LATS2 in hepatocellular carcinoma (HCC) cells. Specifically, LATS1 restricts lethal autophagy in HCC cells induced by sorafenib, the standard of care for advanced HCC patients. Notably, autophagy regulation by LATS1 is independent of its kinase activity. Instead, LATS1 stabilizes the autophagy core-machinery component Beclin-1 by promoting K27-linked ubiquitination at lysine residues K32 and K263 on Beclin-1. Consequently, ubiquitination of Beclin-1 negatively regulates autophagy by promoting inactive dimer formation of Beclin-1. Our study highlights a functional diversity between LATS1 and LATS2, and uncovers a scaffolding role of LATS1 in mediating a cross-talk between the Hippo signaling pathway and autophagy.


Asunto(s)
Autofagia/inmunología , Carcinoma Hepatocelular/patología , Supervivencia Celular/inmunología , Neoplasias Hepáticas/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Autofagia/efectos de los fármacos , Beclina-1/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/mortalidad , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Conjuntos de Datos como Asunto , Supervivencia sin Enfermedad , Resistencia a Antineoplásicos/inmunología , Vía de Señalización Hippo , Humanos , Estimación de Kaplan-Meier , Hígado/patología , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/mortalidad , Lisina/metabolismo , Ratones , Ratones Noqueados , Organoides , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Estabilidad Proteica , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Sorafenib/farmacología , Sorafenib/uso terapéutico , Proteínas Supresoras de Tumor/inmunología , Ubiquitinación , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Cereb Blood Flow Metab ; 37(8): 2894-2909, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28273717

RESUMEN

Activated leukocyte cell adhesion molecule (ALCAM) has been proposed to mediate leukocyte migration across the blood-brain barrier (BBB) in multiple sclerosis or experimental autoimmune encephalomyelitis (EAE). Here, we confirmed vascular ALCAM expression in human brain tissue samples in situ and on two different human in vitro BBB models. Antibody-mediated inhibition of ALCAM reduced diapedesis of human CD4+ Th1 but not of Th17 cells across the human BBB in vitro. In accordance to human Th1 cells, mouse Th1 cells showed reduced diapedesis across an ALCAM-/- in vitro BBB model under static but no longer under flow conditions. In contrast to the limited role of ALCAM in T cell extravasation across the BBB, we found a contribution of ALCAM to rolling, adhesion, and diapedesis of human CD14+ monocytes across the human BBB under flow and static conditions. Taken together, our study highlights the potential differences in the CNS expression of ALCAM in mouse and human and supports a prominent role for ALCAM in the multi-step extravasation of monocytes across the BBB.


Asunto(s)
Antígenos CD/metabolismo , Barrera Hematoencefálica/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas Fetales/metabolismo , Monocitos/inmunología , Linfocitos T/inmunología , Migración Transendotelial y Transepitelial/inmunología , Animales , Antígenos CD/genética , Barrera Hematoencefálica/inmunología , Moléculas de Adhesión Celular Neuronal/genética , Células Cultivadas , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Proteínas Fetales/genética , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/metabolismo , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Linfocitos T/metabolismo , Migración Transendotelial y Transepitelial/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA