Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
FASEB J ; 28(1): 327-36, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24056085

RESUMEN

Memo is a widely expressed 33-kDa protein required for heregulin (HRG)-, epidermal growth factor (EGF)-, and fibroblast growth factor (FGF)-induced cell motility. Studies in mouse embryonic fibroblasts, wild-type or knockout for Memo, were performed to further investigate the role of Memo downstream of FGFR. We demonstrated that Memo associates with the FGFR signalosome and is necessary for optimal activation of signaling. To uncover Memo's physiological role, Memo conditional-knockout mice were generated. These animals showed a reduced life span, increased insulin sensitivity, small stature, graying hair, alopecia, kyphosis, loss of subcutaneous fat, and loss of spermatozoa in the epididymis. Memo-knockout mice also have elevated serum levels of active vitamin D, 1,25-dihydroxyvitamin D3 (1,25(OH)2D), and calcium compared to control littermates expressing Memo. In summary, the results from in vivo and in vitro models support the hypothesis that Memo is a novel regulator of FGFR signaling with a role in controlling 1,25(OH)2D production and normal calcium homeostasis.


Asunto(s)
Proteínas de Hierro no Heme/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Animales , Calcitriol/sangre , Calcio/sangre , Células Cultivadas , Inmunohistoquímica , Inmunoprecipitación , Ratones , Proteínas de Hierro no Heme/genética , Vitamina D/sangre
2.
Breast Cancer Res ; 9(5): R63, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17897439

RESUMEN

BACKGROUND: De-regulation of the wingless and integration site growth factor (WNT) signaling pathway via mutations in APC and Axin, proteins that target beta-catenin for destruction, have been linked to various types of human cancer. These genetic alterations rarely, if ever, are observed in breast tumors. However, various lines of evidence suggest that WNT signaling may also be de-regulated in breast cancer. Most breast tumors show hypermethylation of the promoter region of secreted Frizzled-related protein 1 (sFRP1), a negative WNT pathway regulator, leading to downregulation of its expression. As a consequence, WNT signaling is enhanced and may contribute to proliferation of human breast tumor cells. We previously demonstrated that, in addition to the canonical WNT/beta-catenin pathway, WNT signaling activates the extracellular signal-regulated kinase 1/2 (ERK1/2) pathway in mouse mammary epithelial cells via epidermal growth factor receptor (EGFR) transactivation. METHODS: Using the WNT modulator sFRP1 and short interfering RNA-mediated Dishevelled (DVL) knockdown, we interfered with autocrine WNT signaling at the ligand-receptor level. The impact on proliferation was measured by cell counting, YOPRO, and the MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl-tetrazolium bromide) assay; beta-catenin, EGFR, ERK1/2 activation, and PARP (poly [ADP-ribose]polymerase) cleavages were assessed by Western blotting after treatment of human breast cancer cell lines with conditioned media, purified proteins, small-molecule inhibitors, or blocking antibodies. RESULTS: Phospho-DVL and stabilized beta-catenin are present in many breast tumor cell lines, indicating autocrine WNT signaling activity. Interfering with this loop decreases active beta-catenin levels, lowers ERK1/2 activity, blocks proliferation, and induces apoptosis in MDA-MB-231, BT474, SkBr3, JIMT-1, and MCF-7 cells. The effects of WNT signaling are mediated partly by EGFR transactivation in human breast cancer cells in a metalloprotease- and Src-dependent manner. Furthermore, Wnt1 rescues estrogen receptor-positive (ER+) breast cancer cells from the anti-proliferative effects of 4-hydroxytamoxifen (4-HT) and this activity can be blocked by an EGFR tyrosine kinase inhibitor. CONCLUSION: Our data show that interference with autocrine WNT signaling in human breast cancer reduces proliferation and survival of human breast cancer cells and rescues ER+ tumor cells from 4-HT by activation of the canonical WNT pathway and EGFR transactivation. These findings suggest that interference with WNT signaling at the ligand-receptor level in combination with other targeted therapies may improve the efficiency of breast cancer treatments.


Asunto(s)
Comunicación Autocrina , Neoplasias de la Mama/patología , Proliferación Celular , Receptores ErbB/metabolismo , Transducción de Señal , Activación Transcripcional , Proteína Wnt1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis , Western Blotting , Neoplasias de la Mama/metabolismo , Proteínas de Ciclo Celular/metabolismo , Colágeno Tipo XI/metabolismo , Medios de Cultivo Condicionados , Proteínas Dishevelled , Activación Enzimática , Antagonistas de Estrógenos/farmacología , Humanos , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Interferente Pequeño/farmacología , Receptores de Estrógenos/metabolismo , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología , beta Catenina/metabolismo , Familia-src Quinasas/metabolismo
3.
Sci Signal ; 7(329): ra56, 2014 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-24917593

RESUMEN

Memo is an evolutionarily conserved protein with a critical role in cell motility. We found that Memo was required for migration and invasion of breast cancer cells in vitro and spontaneous lung metastasis from breast cancer cell xenografts in vivo. Biochemical assays revealed that Memo is a copper-dependent redox enzyme that promoted a more oxidized intracellular milieu and stimulated the production of reactive oxygen species (ROS) in cellular structures involved in migration. Memo was also required for the sustained production of the ROS O2- by NADPH (reduced form of nicotinamide adenine dinucleotide phosphate) oxidase 1 (NOX1) in breast cancer cells. Memo abundance was increased in >40% of the primary breast tumors tested, was correlated with clinical parameters of aggressive disease, and was an independent prognostic factor of early distant metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Movimiento Celular , Cobre/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Hierro no Heme/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Xenoinjertos , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Endogámicos NOD , Ratones SCID , NADP/genética , NADP/metabolismo , NADPH Oxidasa 1 , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Proteínas de Hierro no Heme/genética , Superóxidos/metabolismo
4.
EMBO Mol Med ; 5(9): 1335-50, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23868506

RESUMEN

We show that elevated levels of Ret receptor are found in different sub-types of human breast cancers and that high Ret correlates with decreased metastasis-free survival. The role of Ret in ER+ breast cancer models was explored combining in vitro and in vivo approaches. Our analyses revealed that ligand-induced Ret activation: (i) stimulates migration of breast cancer cells; (ii) rescues cells from anti-proliferative effects of endocrine treatment and (iii) stimulates expression of cytokines in the presence of endocrine agents. Indeed, we uncovered a positive feed-forward loop between the inflammatory cytokine IL6 and Ret that links them at the expression and the functional level. In vivo inhibition of Ret in a metastatic breast cancer model inhibits tumour outgrowth and metastatic potential. Ret inhibition blocks the feed-forward loop by down-regulating Ret levels, as well as decreasing activity of Fak, an integrator of IL6-Ret signalling. Our results suggest that Ret kinase should be considered as a novel therapeutic target in subsets of breast cancer.


Asunto(s)
Neoplasias de la Mama/fisiopatología , Movimiento Celular , Proliferación Celular , Metástasis de la Neoplasia/fisiopatología , Proteínas Proto-Oncogénicas c-ret/biosíntesis , Receptores de Estrógenos/metabolismo , Animales , Neoplasias de la Mama/mortalidad , Modelos Animales de Enfermedad , Femenino , Humanos , Interleucina-6/metabolismo , Ratones , Análisis por Micromatrices , Análisis de Supervivencia
5.
J Cell Sci ; 122(Pt 6): 787-97, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19223396

RESUMEN

Heregulin (HRG) activates ErbB2-ErbB3 heterodimers thereby stimulating many cellular responses, including motility. Memo and PLCgamma1 interact with ErbB2 autophosphorylation sites and are essential for HRG-induced chemotaxis. By tracing HRG-stimulated cell migration in Dunn chambers, we found that Memo- or PLCgamma1 knockdown (KD) strongly impairs cell directionality. Memo has no obvious enzymatic activity and was discovered via its ability to complex with ErbB2. Using the yeast two-hybrid approach to gain insight into Memo function, an interaction between Memo and cofilin, a regulator of actin dynamics, was uncovered. The interaction was confirmed in vitro using recombinant proteins and in vivo in co-immunoprecipitation experiments where Memo was detected in complexes with cofilin, ErbB2 and PLCgamma1. Interestingly, in Memo KD cells, HRG-induced PLCgamma1 phosphorylation was decreased, suggesting that Memo regulates PLCgamma1 activation. Furthermore, HRG-induced recruitment of GFP-cofilin to lamellipodia is impaired in Memo and in PLCgamma1 KD cells, suggesting that both proteins lie upstream of cofilin in models of ErbB2-driven tumor-cell migration. Finally, in vitro F-actin binding and depolymerization assays showed that Memo enhances cofilin depolymerizing and severing activity. In summary, these data indicate that Memo also regulates actin dynamics by interacting with cofilin and enhancing its function.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Movimiento Celular , Neoplasias/enzimología , Neoplasias/patología , Proteínas de Hierro no Heme/metabolismo , Fosfolipasa C gamma/metabolismo , Receptor ErbB-2/metabolismo , Actinas/metabolismo , Bioensayo , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Neurregulina-1/farmacología , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Seudópodos/efectos de los fármacos , Seudópodos/enzimología , Proteínas Recombinantes de Fusión/metabolismo
6.
J Biol Chem ; 283(5): 2734-40, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18045866

RESUMEN

Memo (mediator of ErbB2-driven cell motility) is a 297-amino-acid protein recently shown to co-precipitate with the C terminus of ErbB2 and be required for ErbB2-driven cell motility. Memo is not homologous to any known signaling proteins, and how it mediates ErbB2 signals is not known. To provide a molecular basis for understanding Memo function, we have determined and report here the 2.1A crystal structure of human Memo and show it be homologous to class III nonheme iron-dependent dioxygenases, a structural class that now includes a zinc-binding protein of unknown function. No metal binding or enzymatic activity can be detected for Memo, but Memo does bind directly to a specific ErbB2-derived phosphopeptide encompassing Tyr-1227 using its vestigial enzymatic active site. Memo thus represents a new class of phosphotyrosine-binding protein.


Asunto(s)
Proteínas de Hierro no Heme/química , Proteínas de Hierro no Heme/metabolismo , Receptor ErbB-2/metabolismo , Secuencia de Aminoácidos , Sitios de Unión/genética , Movimiento Celular/fisiología , Cristalografía por Rayos X , Humanos , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intracelular , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas de Hierro no Heme/genética , Fosfopéptidos/química , Fosfopéptidos/genética , Fosfopéptidos/metabolismo , Fosfotirosina/química , Fosfotirosina/metabolismo , Unión Proteica , Conformación Proteica , Receptor ErbB-2/química , Receptor ErbB-2/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA