Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Rev Mol Cell Biol ; 14(6): 393-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23636498

RESUMEN

The past few years have marked significant anniversaries in signal transduction, including the identification of classic growth factors and morphogens, the notion of protein modification through phosphorylation and the characterization of protein interaction domains. Here, six researchers reflect on the context in which these discoveries were made, and how our concept of cell signalling has evolved during the past three decades.


Asunto(s)
Investigación Biomédica/historia , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Procesamiento Proteico-Postraduccional , Transducción de Señal , Animales , Investigación Biomédica/métodos , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Fosforilación
2.
J Mammary Gland Biol Neoplasia ; 25(1): 13-26, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32080788

RESUMEN

Ret receptor tyrosine kinase is a proto-oncogene that participates in development of various cancers. Several independent studies have recently identified Ret as a key player in breast cancer. Although Ret overexpression and function have been under investigation, mainly in estrogen receptor positive breast cancer, a more comprehensive analysis of the impact of recurring Ret alterations in breast cancer is needed. This review consolidates the current knowledge of Ret alterations and their potential effects in breast cancer. We discuss and integrate data on Ret changes in different breast cancer subtypes and potential function in progression, as well as the participation of distinct Ret network signaling partners in these processes. We propose that it will be essential to define a shared molecular feature of tumors with alteration in Ret receptor, be this at the genetic level or via overexpression in order to design effective therapies to target the Ret pathway. Here we review experimental evidence from basic research and pre-clinical studies concentrating on Ret alterations as potential biomarkers for recurrence, and we discuss the possibility that targeting the Ret pathway might in the future become a treatment for breast cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica , Mutación , Proteínas Proto-Oncogénicas c-ret/metabolismo , Animales , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-ret/genética
3.
PLoS Biol ; 14(9): e2000314, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27684370

RESUMEN

Cancer research has become a global enterprise, and the number of researchers, as well as the cost for their activities, has skyrocketed. The budget for the National Cancer Institute of the United States National Institutes of Health alone was US$5.2 billion in 2015. Since most of the research is funded by public money, it is perfectly legitimate to ask if these large expenses are worth it. In this brief commentary, we recapitulate some of the breakthroughs that mark the history of breast cancer research over the past decades and emphasize the resulting benefits for afflicted women. In 1971, only 40% of women diagnosed with breast cancer would live another 10 years. Today, nearly 80% of women reach that significant milestone in most developed countries. This dramatic change has afforded breast cancer patients many productive years and a better quality of life. Progress resulted largely from advances in the understanding of the molecular details of the disease and their translation into innovative, rationally designed therapies. These developments are founded on the revolution in molecular and cellular biology, an entirely new array of methods and technologies, the enthusiasm, optimism, and diligence of scientists and clinicians, and the considerable funding efforts from public and private sources. We were lucky to be able to spend our productive years in a period of scientific upheaval in which methods and concepts were revolutionized and that allowed us to contribute, within the global scientific community, to the progress in basic science and clinical practice.

4.
Mol Cell ; 41(1): 5-7, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21211718

RESUMEN

In a recent issue of Molecular Cell, Kazanietz and colleagues (Sosa et al., 2010) show that P-Rex1, a Rac1 GEF, is overexpressed in ER+ and/or ErbB2+ breast cancers, suggesting that P-Rex1 might be a convergence node downstream of these receptors and an attractive therapeutic target.

5.
Dev Biol ; 415(2): 278-295, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-26746790

RESUMEN

The cranial base is a component of the neurocranium and has a central role in the structural integration of the face, brain and vertebral column. Consequently, alteration in the shape of the human cranial base has been intimately linked with primate evolution and defective development is associated with numerous human facial abnormalities. Here we describe a novel recessive mutant mouse strain that presented with a domed head and fully penetrant cleft secondary palate coupled with defects in the formation of the underlying cranial base. Mapping and non-complementation studies revealed a specific mutation in Memo1 - a gene originally associated with cell migration. Expression analysis of Memo1 identified robust expression in the perichondrium and periosteum of the developing cranial base, but only modest expression in the palatal shelves. Fittingly, although the palatal shelves failed to elevate in Memo1 mutants, expression changes were modest within the shelves themselves. In contrast, the cranial base, which forms via endochondral ossification had major reductions in the expression of genes responsible for bone formation, notably matrix metalloproteinases and markers of the osteoblast lineage, mirrored by an increase in markers of cartilage and extracellular matrix development. Concomitant with these changes, mutant cranial bases showed an increased zone of hypertrophic chondrocytes accompanied by a reduction in both vascular invasion and mineralization. Finally, neural crest cell-specific deletion of Memo1 caused a failure of anterior cranial base ossification indicating a cell autonomous role for MEMO1 in the development of these neural crest cell derived structures. However, palate formation was largely normal in these conditional mutants, suggesting a non-autonomous role for MEMO1 in palatal closure. Overall, these findings assign a new function to MEMO1 in driving endochondral ossification in the cranium, and also link abnormal development of the cranial base with more widespread effects on craniofacial shape relevant to human craniofacial dysmorphology.


Asunto(s)
Fisura del Paladar/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Desarrollo Maxilofacial/fisiología , Osteogénesis/fisiología , Hueso Paladar/embriología , Base del Cráneo/embriología , Animales , Cartílago/embriología , Cartílago/patología , Fisura del Paladar/embriología , Etilnitrosourea , Exones , Regulación del Desarrollo de la Expresión Génica , Genes Recesivos , Humanos , Masculino , Mesodermo/citología , Mesodermo/embriología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutagénesis , Cresta Neural/citología , Cresta Neural/embriología , Hueso Paladar/metabolismo , Hueso Paladar/patología , Mutación Puntual , Base del Cráneo/metabolismo , Base del Cráneo/patología , Especificidad de la Especie
6.
Int J Cancer ; 137(8): 1842-54, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25868708

RESUMEN

Tenascin-W is a matricellular protein with a dynamically changing expression pattern in development and disease. In adults, tenascin-W is mostly restricted to stem cell niches, and is also expressed in the stroma of solid cancers. Here, we analyzed its expression in the bone microenvironment of breast cancer metastasis. Osteoblasts were isolated from tumor-free or tumor-bearing bones of mice injected with MDA-MB231-1833 breast cancer cells. We found a fourfold upregulation of tenascin-W in the osteoblast population of tumor-bearing mice compared to healthy mice, indicating that tenascin-W is supplied by the bone metastatic niche. Transwell and co-culture studies showed that human bone marrow stromal cells (BMSCs) express tenascin-W protein after exposure to factors secreted by MDA-MB231-1833 breast cancer cells. To study tenascin-W gene regulation, we identified and analyzed the tenascin-W promoter as well as three evolutionary conserved regions in the first intron. 5'RACE analysis of mRNA from human breast cancer, glioblastoma and bone tissue showed a single tenascin-W transcript with a transcription start site at a noncoding first exon followed by exon 2 containing the ATG translation start. Site-directed mutagenesis of a SMAD4-binding element in proximity of the TATA box strongly impaired promoter activity. TGFß1 induced tenascin-W expression in human BMSCs through activation of the TGFß1 receptor ALK5, while glucocorticoids were inhibitory. Our experiments show that tenascin-W acts as a niche component for breast cancer metastasis to bone by supporting cell migration and cell proliferation of the cancer cells.


Asunto(s)
Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Neoplasias de la Mama/patología , Osteoblastos/metabolismo , Tenascina/genética , Factor de Crecimiento Transformador beta/metabolismo , Animales , Células de la Médula Ósea/citología , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Femenino , Humanos , Ratones , Trasplante de Neoplasias , Osteoblastos/patología , Transducción de Señal , Células del Estroma/citología , Células del Estroma/metabolismo , Tenascina/metabolismo , Microambiente Tumoral , Regulación hacia Arriba
7.
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
9.
Growth Factors ; 32(2): 74-81, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24641597

RESUMEN

The epidermal growth factor receptor (EGFR) undergoes a conformational change in response to ligand binding. The ligand-induced changes in cell surface aggregation and mobility have a profound effect on the function of all the family members. Ligand also activates the EGFR intracellular kinase, stimulating proliferation and cell survival. The EGFR family are often activated, overexpressed or mutated in cancer cells and therapeutic drugs (including antibodies) can slow the progress of some cancers. This article provides a brief, annotated summary of the presentations and discussion which occurred at the Epidermal Growth Factor Receptor - Future Directions Conference held in Jerusalem in November 2013.


Asunto(s)
Receptores ErbB/metabolismo , Receptores ErbB/uso terapéutico , Neoplasias/tratamiento farmacológico , Animales , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Unión Proteica , Conformación Proteica , Transporte de Proteínas , Receptor ErbB-2/metabolismo , Transducción de Señal
10.
Cancer Cell ; 10(1): 7-11, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16843261

RESUMEN

Aberrant ERBB receptor activity contributes to the development of many human cancers. Receptor overexpression, kinase domain (KD) mutations, and autocrine ligand production contribute to ERBB activation in human tumors. ERBB-targeted tyrosine kinase inhibitors (TKIs) and monoclonal antibodies are used in cancer treatment; however, clinical hurdles, including patient selection and TKI resistance, need to be overcome in order to optimize therapy. This minireview will discuss recent findings on possible mechanisms leading to ERBB-targeted therapy resistance and potential means to overcome them.


Asunto(s)
Proteínas ADAM/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas ADAM/metabolismo , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/metabolismo , Receptor ErbB-3/antagonistas & inhibidores , Receptor ErbB-3/metabolismo , Receptor ErbB-4
11.
Nat Rev Cancer ; 5(5): 341-54, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15864276

RESUMEN

ERBB receptor tyrosine kinases have important roles in human cancer. In particular, the expression or activation of epidermal growth factor receptor and ERBB2 are altered in many epithelial tumours, and clinical studies indicate that they have important roles in tumour aetiology and progression. Accordingly, these receptors have been intensely studied to understand their importance in cancer biology and as therapeutic targets, and many ERBB inhibitors are now used in the clinic. We will discuss the significance of these receptors as clinical targets, in particular the molecular mechanisms underlying response.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/etiología , Receptor ErbB-2/fisiología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Sistemas de Liberación de Medicamentos , Resistencia a Antineoplásicos , Humanos , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/inmunología , Transducción de Señal/efectos de los fármacos
12.
Breast Cancer Res ; 15(5): 315, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24172042

RESUMEN

Epidermal growth factor receptor (EGFR) is highly expressed in triple-negative breast cancer (TNBC), and elevated levels correlate with poor prognosis. In analogy with the paradigm of oncogene addiction, blocking EGFR in TNBC was expected to have clinical efficacy ­ but this has not been the case. Reasons for these results have remained elusive. Recently, Meyer and colleagues showed interplay between EGFR and the epithelial-to-mesenchymal transition-associated AXL receptor in TNBC cells, which might provide some clues.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptores ErbB/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal , Femenino , Humanos
13.
Breast Cancer Res ; 15(1): R8, 2013 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-23343422

RESUMEN

INTRODUCTION: Targeting receptor tyrosine kinases (RTKs) with kinase inhibitors is a clinically validated anti-cancer approach. However, blocking one signaling pathway is often not sufficient to cause tumor regression and the effectiveness of individual inhibitors is often short-lived. As alterations in fibroblast growth factor receptor (FGFR) activity have been implicated in breast cancer, we examined in breast cancer models with autocrine FGFR activity the impact of targeting FGFRs in vivo with a selective kinase inhibitor in combination with an inhibitor of PI3K/mTOR or with a pan-ErbB inhibitor. METHODS: Using 4T1 or 67NR models of basal-like breast cancer, tumor growth was measured in mice treated with an FGFR inhibitor (dovitinib/TKI258), a PI3K/mTOR inhibitor (NVP-BEZ235) or a pan-ErbB inhibitor (AEE788) individually or in combination. To uncover mechanisms underlying inhibitor action, signaling pathway activity was examined in tumor lysates and transcriptome analysis carried out to identify pathways upregulated by FGFR inhibition. Anti-phosphotyrosine receptor antibody arrays (P-Tyr RTK) were also used to screen 4T1 tumors. RESULTS: The combination of dovitinib + NVP-BEZ235 causes tumor stasis and strong down-regulation of the FRS2/Erk and PI3K/Akt/mTOR signaling pathways. P-Tyr RTK arrays identified high levels of P-EGFR and P-ErbB2 in 4T1 tumors. Testing AEE788 in the tumor models revealed that the combination of dovitinib + AEE788 resulted in blockade of the PI3K/Akt/mTOR pathway, prolonged tumor stasis and in the 4T1 model, a significant decrease in lung metastasis. The results show that in vivo these breast cancer models become dependent upon co-activation of FGFR and ErbB receptors for PI3K pathway activity. CONCLUSIONS: The work presented here shows that in the breast cancer models examined, the combination of dovitinib + NVP-BEZ235 or dovitinib + AEE788 results in strong inhibition of tumor growth and a block in metastatic spread. Only these combinations strongly down-regulate the FGFR/FRS2/Erk and PI3K/Akt/mTOR signaling pathways. The resultant decrease in mitosis and increase in apoptosis was consistently stronger in the dovitinib + AEE788 treatment-group, suggesting that targeting ErbB receptors has broader downstream effects compared to targeting only PI3K/mTOR. Considering that sub-classes of human breast tumors co-express ErbB receptors and FGFRs, these results have implications for targeted therapy.


Asunto(s)
Neoplasias de la Mama/genética , Proliferación Celular/efectos de los fármacos , Receptores ErbB/genética , Receptores de Factores de Crecimiento de Fibroblastos/genética , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Receptores ErbB/antagonistas & inhibidores , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Imidazoles/administración & dosificación , Ratones , Terapia Molecular Dirigida , Fosfatidilinositol 3-Quinasas/administración & dosificación , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Proto-Oncogénicas c-akt/genética , Purinas/administración & dosificación , Quinolinas/administración & dosificación , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/genética , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Cell Commun Signal ; 17(3): 705-722, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36434320

RESUMEN

Memo1 deletion in mice causes premature aging and an unbalanced metabolism partially resembling Fgf23 and Klotho loss-of-function animals. We report a role for Memo's redox function in renal FGF23-Klotho signaling using mice with postnatally induced Memo deficiency in the whole body (cKO). Memo cKO mice showed impaired FGF23-driven renal ERK phosphorylation and transcriptional responses. FGF23 actions involved activation of oxidation-sensitive protein phosphotyrosyl phosphatases in the kidney. Redox proteomics revealed excessive thiols of Rho-GDP dissociation inhibitor 1 (Rho-GDI1) in Memo cKO, and we detected a functional interaction between Memo's redox function and oxidation at Rho-GDI1 Cys79. In isolated cellular systems, Rho-GDI1 did not directly affect FGF23-driven cell signaling, but we detected disturbed Rho-GDI1 dependent small Rho-GTPase protein abundance and activity in the kidney of Memo cKO mice. Collectively, this study reveals previously unknown layers in the regulation of renal FGF23 signaling and connects Memo with the network of small Rho-GTPases.

15.
Breast Cancer Res ; 14(5): R131, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-23062209

RESUMEN

INTRODUCTION: The tyrosine kinase receptors HER2 and HER3 play an important role in breast cancer. The HER2/HER3 heterodimer is a critical oncogenic unit associated with reduced relapse-free and decreased overall survival. While signaling cascades downstream of HER2 and HER3 have been studied extensively at the level of post-translational modification, little is known about the effects of HER2/HER3 overexpression and activation on gene expression in breast cancer. We have now defined the genetic landscape induced by activation of the HER2/HER3 unit in mammary cells, and have identified interleukin (IL)8 and CXCR1 as potential therapeutic targets for the treatment of HER2/HER3-overexpressing breast cancers. METHODS: Three-dimensional (3D) cultures, invasion and migration assays were used to determine the effects of HER2 and HER3 co-expression and activation. Gene expression analysis was performed to identify the gene network induced by HER2/HER3 in 3D cultures. Bioinformatic analysis and neutralizing antibodies were used to identify key mediators of HER2/HER3-evoked invasion. RESULTS: Co-expression of the tyrosine kinase receptors HER2 and HER3 induced migration and invasion of MCF10A cells. Microarray analysis of these cells revealed a specific "HER2/HER3 signature" comprising 80 upregulated transcripts, with IL8 being the highest (11-fold upregulation). Notably, examination of public datasets revealed high levels of IL8 transcripts in HER2-enriched as well as basal-like primary breast tumors, two subtypes characterized by a particularly poor prognosis. Moreover, IL8 expression correlated with high tumor grade and ER-negative status. Importantly, treatment with IL8-neutralizing antibodies prevented invasion of MCF10A-HER2/HER3 and BT474 cells in 3D cultures, highlighting the importance of IL8 autocrine signaling upon HER2/HER3 activation. CONCLUSIONS: Our findings demonstrate that HER2 and HER3 co-expression induces IL8 autocrine signaling, leading to the invasion of mammary cells. Agents targeting IL8 or its receptor CXCR1 may be useful for the treatment of HER2/HER3/IL8-positive breast cancers with invasive traits.


Asunto(s)
Comunicación Autocrina , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Expresión Génica , Interleucina-8/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-3/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Conjuntos de Datos como Asunto , Femenino , Perfilación de la Expresión Génica , Humanos , Modelos Biológicos , Clasificación del Tumor , Neurregulina-1/farmacología , Transducción de Señal , Esferoides Celulares , Células Tumorales Cultivadas
16.
Cancer Cell ; 5(4): 299-301, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15093533

RESUMEN

In cancer cells, the ErbB2 receptor tyrosine kinase can be activated in two ways: by overexpression or by ligand-mediated stimulation of another ErbB receptor. The ErbB2-targeting antibody trastuzumab (Herceptin) is used for treatment of metastatic breast cancer patients whose tumors overexpress ErbB2. A new structural study in this issue of Cancer Cell reveals how targeting ErbB2 with another antibody, pertuzumab (Omnitarg), prevents ligand-induced dimerization of ErbB2 with the other ErbB receptors. Pertuzumab's novel mode of action might offer additional therapeutic opportunities for treatment of tumors expressing ligand-activated ErbB2.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias de la Mama/terapia , Receptor ErbB-2/metabolismo , Anticuerpos Monoclonales Humanizados , Neoplasias de la Mama/metabolismo , Dimerización , Femenino , Humanos , Trastuzumab
17.
Dis Model Mech ; 15(3)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35044452

RESUMEN

RET is a receptor tyrosine kinase with oncogenic potential in the mammary epithelium. Several receptors with oncogenic activity in the breast are known to participate in specific developmental stages. We found that RET is differentially expressed during mouse mammary gland development: RET is present in lactation and its expression dramatically decreases in involution, the period during which the lactating gland returns to a quiescent state after weaning. Based on epidemiological and pre-clinical findings, involution has been described as tumor promoting. Using the Ret/MTB doxycycline-inducible mouse transgenic system, we show that sustained expression of RET in the mammary epithelium during the post-lactation transition to involution is accompanied by alterations in tissue remodeling and an enhancement of cancer potential. Following constitutive Ret expression, we observed a significant increase in neoplastic lesions in the post-involuting versus the virgin mammary gland. Furthermore, we show that abnormal RET overexpression during lactation promotes factors that prime involution, including premature activation of Stat3 signaling and, using RNA sequencing, an acute-phase inflammatory signature. Our results demonstrate that RET overexpression negatively affects the normal post-lactation transition.


Asunto(s)
Glándulas Mamarias Humanas , Neoplasias , Animales , Femenino , Humanos , Lactancia/fisiología , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Glándulas Mamarias Humanas/metabolismo , Ratones , Neoplasias/patología , Proteínas Proto-Oncogénicas c-ret/genética , Proteínas Proto-Oncogénicas c-ret/metabolismo , Factor de Transcripción STAT3/metabolismo
18.
Nat Cell Biol ; 6(6): 515-22, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15156151

RESUMEN

Clinical studies have revealed that cancer patients whose tumours have increased ErbB2 expression tend to have more aggressive, metastatic disease, which is associated with parameters predicting a poor outcome. The molecular basis underlying ErbB2-dependent cell motility and metastases formation, however, still remains poorly understood. In this study, we show that activation of a set of signalling molecules, including MAPK, phosphatidylinositol-3-OH kinase (PI(3)K) and Src, is required for Neu/ErbB2-dependent lamellipodia formation and for motility of breast carcinoma cells. Stimulation of these molecules, however, failed to induce efficient cell migration in the absence of Neu/ErbB2 phosphorylation at Tyr 1201 or Tyr 1227. We describe a novel molecule, Memo (mediator of ErbB2-driven cell motility), that interacts with a phospho-Tyr 1227-containing peptide, most probably through the Shc adaptor protein. After Neu/ErbB2 activation, Memo-defective cells form actin fibres and grow lamellipodia, but fail to extend microtubules towards the cell cortex. Our data suggest that Memo controls cell migration by relaying extracellular chemotactic signals to the microtubule cytoskeleton.


Asunto(s)
Proteínas Portadoras/metabolismo , Movimiento Celular/fisiología , Proteínas de la Membrana/metabolismo , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , Receptor ErbB-2/genética , Transducción de Señal/genética , Citoesqueleto de Actina/metabolismo , Humanos , Proteínas de la Membrana/aislamiento & purificación , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Seudópodos/metabolismo , Receptor ErbB-2/metabolismo , Células Tumorales Cultivadas , Familia-src Quinasas/metabolismo
19.
J Cell Biol ; 171(4): 717-28, 2005 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-16301336

RESUMEN

Integrin-mediated adhesion regulates the development and function of a range of tissues; however, little is known about its role in glandular epithelium. To assess the contribution of beta1 integrin, we conditionally deleted its gene in luminal epithelia during different stages of mouse mammary gland development and in cultured primary mammary epithelia. Loss of beta1 integrin in vivo resulted in impaired alveologenesis and lactation. Cultured beta1 integrin-null cells displayed abnormal focal adhesion function and signal transduction and could not form or maintain polarized acini. In vivo, epithelial cells became detached from the extracellular matrix but remained associated with each other and did not undergo overt apoptosis. beta1 integrin-null mammary epithelial cells did not differentiate in response to prolactin stimulation because of defective Stat5 activation. In mice where beta1 integrin was deleted after the initiation of differentiation, fewer defects in alveolar morphology occurred, yet major deficiencies were also observed in milk protein and milk fat production and Stat5 activation, indicating a permissive role for beta1 integrins in prolactin signaling. This study demonstrates that beta1 integrin is critical for the alveolar morphogenesis of a glandular epithelium and for maintenance of its differentiated function. Moreover, it provides genetic evidence for the cooperation between integrin and cytokine signaling pathways.


Asunto(s)
Células Epiteliales/citología , Epitelio/metabolismo , Integrina beta1/genética , Integrina beta1/fisiología , Glándulas Mamarias Animales/metabolismo , Animales , Western Blotting , Adhesión Celular , Diferenciación Celular , Células Cultivadas , Cruzamientos Genéticos , Citocinas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Eliminación de Gen , Regulación de la Expresión Génica , Integrinas/metabolismo , Lactancia , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Modelos Genéticos , Prolactina/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Factores de Tiempo
20.
Clin Cancer Res ; 15(11): 3733-9, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19458057

RESUMEN

PURPOSE: The epidermal growth factor receptor (ERBB1) and related family member HER-2/neu (ERBB2) are often overexpressed in aggressive breast cancers and their overexpression is correlated with poor prognosis. Clinical studies using ERBB inhibitors have focused on tumor growth effects, but ERBBs can contribute to malignancy independent of their effects on tumor growth. Our studies were designed to evaluate the effect of ERBB inhibition on tumor cell motility and intravasation in vivo using clinically relevant small-molecule inhibitors. EXPERIMENTAL DESIGN: Using in vivo mouse models of breast cancer, we test the effects of ERBB1 and ERBB2 inhibitors AC480 and lapatinib, ERBB1 inhibitor gefitinib, and ERBB2 inhibitor AG825 on in vivo tumor cell invasive properties in mammary fat pad tumors. RESULTS: ERBB1 and ERBB2 inhibition rapidly (within 3 h) inhibits both tumor cell motility and intravasation. Using gefitinib, ERBB1 inhibition rapidly inhibits tumor cell motility and invasion but not intravasation, whereas ERBB2 inhibition by AG825 rapidly blocks intravasation. CONCLUSIONS: ERBB1 and ERBB2 inhibition can rapidly block tumor cell invasive properties. In addition, we differentiate for the first time the contributions of ERBB1 and ERBB2 to the key metastatic properties of in vivo tumor cell invasion and intravasation. These experiments temporally and molecularly separate two key stages in tumor cell entry into blood vessels: invasion and intravasation. These results indicate that ERBB inhibition should be considered for blocking other tumor cell malignant properties besides growth.


Asunto(s)
Receptores ErbB/metabolismo , Neoplasias Mamarias Experimentales/patología , Receptor ErbB-2/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Benzotiazoles/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/fisiología , Femenino , Gefitinib , Humanos , Lapatinib , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones SCID , Invasividad Neoplásica , Metástasis de la Neoplasia , Fosforilación/efectos de los fármacos , Quinazolinas/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/fisiología , Tirfostinos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA