Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Proc Natl Acad Sci U S A ; 110(5): 1815-20, 2013 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-23319610

RESUMEN

Breast tumors lacking expression of human epidermal growth factor receptor 2 (HER2) and the estrogen and the progesterone receptors (triple negative; TNBC) are more aggressive than other disease subtypes, and no molecular targeted agents are currently available for their treatment. Because TNBC commonly displays EGF receptor (EGFR) expression, and combinations of monoclonal antibodies to EGFR effectively inhibit other tumor models, we addressed the relevance of this strategy to treatment of TNBC. Unlike a combination of the clinically approved monoclonal antibodies, cetuximab and panitumumab, which displaced each other and displayed no cooperative effects, several other combinations resulted in enhanced inhibition of TNBC's cell growth both in vitro and in animals. The ability of certain antibody mixtures to remove EGFR from the cell surface and to promote its intracellular degradation correlated with the inhibitory potential. However, unlike EGF-induced sorting of EGFR to lysosomal degradation, the antibody-induced pathway displayed independence from the intrinsic kinase activity and dimer formation ability of EGFR, and it largely avoided the recycling route. In conclusion, although TNBC clinical trials testing EGFR inhibitors reported lack of benefit, our results offer an alternative strategy that combines noncompetitive antibodies to achieve robust degradation of EGFR and tumor inhibition.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Receptores ErbB/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cetuximab , Receptores ErbB/metabolismo , Femenino , Células HeLa , Humanos , Immunoblotting , Ratones , Ratones Desnudos , Panitumumab , Proteolisis/efectos de los fármacos , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Carga Tumoral/efectos de los fármacos
2.
Nat Cell Biol ; 9(8): 961-9, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17643115

RESUMEN

Cell migration driven by the epidermal growth factor receptor (EGFR) propels morphogenesis and involves reorganization of the actin cytoskeleton. Although de novo transcription precedes migration, transcript identity remains largely unknown. Through their actin-binding domains, tensins link the cytoskeleton to integrin-based adhesion sites. Here we report that EGF downregulates tensin-3 expression, and concomitantly upregulates cten, a tensin family member that lacks the actin-binding domain. Knockdown of cten or tensin-3, respectively, impairs or enhances mammary cell migration. Furthermore, cten displaces tensin-3 from the cytoplasmic tail of integrin beta1, thereby instigating actin fibre disassembly. In invasive breast cancer, cten expression correlates not only with high EGFR and HER2, but also with metastasis to lymph nodes. Moreover, treatment of inflammatory breast cancer patients with an EGFR/HER2 dual-specificity kinase inhibitor significantly downregulated cten expression. In conclusion, a transcriptional tensin-3-cten switch may contribute to the metastasis of mammary cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Movimiento Celular/fisiología , Factor de Crecimiento Epidérmico/metabolismo , Proteínas de Microfilamentos/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Inhibidores Enzimáticos/metabolismo , Receptores ErbB , Femenino , Humanos , Proteínas de Microfilamentos/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Tensinas
3.
FASEB J ; 26(4): 1582-92, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22198386

RESUMEN

The signaling pathways that commit cells to migration are incompletely understood. We employed human mammary cells and two stimuli: epidermal growth factor (EGF), which induced cellular migration, and serum factors, which stimulated cell growth. In addition to strong activation of ERK by EGF, and AKT by serum, early transcription remarkably differed: while EGF induced early growth response-1 (EGR1), and this was required for migration, serum induced c-Fos and FosB to enhance proliferation. We demonstrate that induction of EGR1 involves ERK-mediated down-regulation of microRNA-191 and phosphorylation of the ETS2 repressor factor (ERF) repressor, which subsequently leaves the nucleus. Unexpectedly, knockdown of ERF inhibited migration, which implies migratory roles for exported ERF molecules. On the other hand, chromatin immunoprecipitation identified a subset of direct EGR1 targets, including EGR1 autostimulation and SERPINB2, whose transcription is essential for EGF-induced cell migration. In summary, EGR1 and the EGF-ERK-ERF axis emerge from our study as major drivers of growth factor-induced mammary cell migration.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glándulas Mamarias Humanas/citología , Proteínas Represoras/metabolismo , Transducción de Señal/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Femenino , Perfilación de la Expresión Génica , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Análisis por Micromatrices , Proteoma/análisis , Proteínas Represoras/genética , Transducción de Señal/fisiología , Técnicas del Sistema de Dos Híbridos
4.
Nat Biotechnol ; 41(6): 788-793, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36593397

RESUMEN

Spatial transcriptomics and proteomics provide complementary information that independently transformed our understanding of complex biological processes. However, experimental integration of these modalities is limited. To overcome this, we developed Spatial PrOtein and Transcriptome Sequencing (SPOTS) for high-throughput simultaneous spatial transcriptomics and protein profiling. Compared with unimodal measurements, SPOTS substantially improves signal resolution and cell clustering and enhances the discovery power in differential gene expression analysis across tissue regions.


Asunto(s)
Perfilación de la Expresión Génica , Transcriptoma , Transcriptoma/genética , Proteínas , Proteómica , Análisis por Conglomerados
5.
bioRxiv ; 2023 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-37961223

RESUMEN

Tumor-infiltrating macrophages support critical steps in tumor progression, and their accumulation in the tumor microenvironment (TME) is associated with adverse outcomes and therapeutic resistance across human cancers. In the TME, macrophages adopt diverse phenotypic alterations, giving rise to heterogeneous immune activation states and induction of cell cycle. While the transcriptional profiles of these activation states are well-annotated across human cancers, the underlying signals that regulate macrophage heterogeneity and accumulation remain incompletely understood. Here, we leveraged a novel ex vivo organotypic TME (oTME) model of breast cancer, in vivo murine models, and human samples to map the determinants of functional heterogeneity of TME macrophages. We identified a subset of F4/80highSca-1+ self-renewing macrophages maintained by type-I interferon (IFN) signaling and requiring physical contact with cancer-associated fibroblasts. We discovered that the contact-dependent self-renewal of TME macrophages is mediated via Notch4, and its inhibition abrogated tumor growth of breast and ovarian carcinomas in vivo, as well as lung dissemination in a PDX model of triple-negative breast cancer (TNBC). Through spatial multi-omic profiling of protein markers and transcriptomes, we found that the localization of macrophages further dictates functionally distinct but reversible phenotypes, regardless of their ontogeny. Whereas immune-stimulatory macrophages (CD11C+CD86+) populated the tumor epithelial nests, the stroma-associated macrophages (SAMs) were proliferative, immunosuppressive (Sca-1+CD206+PD-L1+), resistant to CSF-1R depletion, and associated with worse patient outcomes. Notably, following cessation of CSF-1R depletion, macrophages rebounded primarily to the SAM phenotype, which was associated with accelerated growth of mammary tumors. Our work reveals the spatial determinants of macrophage heterogeneity in breast cancer and highlights the disruption of macrophage self-renewal as a potential new therapeutic strategy.

6.
Adv Sci (Weinh) ; 9(9): e2105696, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35092356

RESUMEN

Macrophage infiltration in mammary tumors is associated with enhanced tumor progression, metastasis, and poor clinical outcome, and considered as target for therapeutic intervention. By using different genetic mouse models, the authors show that ablation of the tyrosine kinase PYK2, either in breast cancer cells, only in the tumor microenvironment, or in both, markedly reduces the number of infiltrating tumor macrophages and concomitantly inhibits tumor angiogenesis and tumor growth. Strikingly, PYK2 ablation only in macrophages is sufficient to induce similar effects. These phenotypic changes are associated with reduced monocyte recruitment and a substantial decrease in tumor-associated macrophages (TAMs). Mechanistically, the authors show that PYK2 mediates mutual communication between breast cancer cells and macrophages through critical effects on key receptor signaling. Specifically, PYK2 ablation inhibits Notch1 signaling and consequently reduces CCL2 secretion by breast cancer cells, and concurrently reduces the levels of CCR2, CXCR4, IL-4Rα, and Stat6 activation in macrophages. These bidirectional effects modulate monocyte recruitment, macrophage polarization, and tumor angiogenesis. The expression of PYK2 is correlated with infiltrated macrophages in breast cancer patients, and its effects on macrophage infiltration and pro-tumorigenic phenotype suggest that PYK2 targeting can be utilized as an effective strategy to modulate TAMs and possibly sensitize breast cancer to immunotherapy.


Asunto(s)
Neoplasias de la Mama , Macrófagos , Animales , Carcinogénesis , Comunicación Celular , Femenino , Quinasa 2 de Adhesión Focal/metabolismo , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Microambiente Tumoral
7.
Sci Rep ; 9(1): 857, 2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30696910

RESUMEN

Iron deposits are a phenotypic trait of tumor-associated macrophages (TAMs). Histological iron imaging and contrast-agent free magnetic resonance imaging (MRI) can detect these deposits, but their presence  in human cancer, and correlation with immunotherapeutic response is largely untested. Here, primarily using these iron imaging approaches, we evaluated the spatial distribution of polarized macrophage populations containing high endogenous levels of iron in preclinical murine models and human breast cancer, and used them as metabolic biomarkers to correlate TAM infiltration with response to immunotherapy in preclinical trials. Macrophage-targeted inhibition of the colony stimulating factor 1 receptor (CSF1R) by immunotherapy was confirmed to inhibit macrophage accumulation and slow mammary tumor growth in mouse models while also reducing hemosiderin iron-laden TAM accumulation as measured by both iron histology and in vivo iron MRI (FeMRI). Spatial profiling of TAM iron deposit infiltration defined regions of maximal accumulation and response to the CSF1R inhibitor, and revealed differences between microenvironments of human cancer according to levels of polarized macrophage iron accumulation in stromal margins. We therefore demonstrate that iron deposition serves as an endogenous metabolic imaging biomarker of TAM infiltration in breast cancer that has high translational potential for evaluation of immunotherapeutic response.


Asunto(s)
Antineoplásicos/uso terapéutico , Biomarcadores Farmacológicos/metabolismo , Neoplasias de la Mama/inmunología , Inmunoterapia/métodos , Hierro/metabolismo , Macrófagos/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/terapia , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Diagnóstico por Imagen , Femenino , Hemosiderina/metabolismo , Humanos , Espacio Intracelular , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Microambiente Tumoral
8.
PLoS One ; 12(9): e0184765, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28898277

RESUMEN

Iron-deposition is a metabolic biomarker of macrophages in both normal and pathological situations, but the presence of iron in tumor and metastasis-associated macrophages is not known. Here we mapped and quantified hemosiderin-laden macrophage (HLM) deposits in murine models of metastatic breast cancer using iron and macrophage histology, and in vivo MRI. Iron MRI detected high-iron pixel clusters in mammary tumors, lung metastasis, and brain metastasis as well as liver and spleen tissue known to contain the HLMs. Iron histology showed these regions to contain clustered macrophages identified by their common iron status and tissue-intrinsic association with other phenotypic macrophage markers. The in vivo MRI and ex vivo histological images were further processed to determine the frequencies and sizes of the iron deposits, and measure the number of HLMs in each deposit to estimate the in vivo MRI sensitivity for these cells. Hemosiderin accumulation is a macrophage biomarker and intrinsic contrast source for cellular MRI associated with the innate function of macrophages in iron metabolism systemically, and in metastatic cancer.


Asunto(s)
Hemosiderina/metabolismo , Hierro/metabolismo , Macrófagos/metabolismo , Neoplasias Mamarias Experimentales/diagnóstico por imagen , Animales , Gránulos Citoplasmáticos/metabolismo , Gránulos Citoplasmáticos/patología , Femenino , Macrófagos/patología , Imagen por Resonancia Magnética , Neoplasias Mamarias Experimentales/patología , Ratones , Metástasis de la Neoplasia
9.
Nat Cell Biol ; 19(8): 974-987, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28737771

RESUMEN

Obesity is associated with chronic, low-grade inflammation, which can disrupt homeostasis within tissue microenvironments. Given the correlation between obesity and relative risk of death from cancer, we investigated whether obesity-associated inflammation promotes metastatic progression. We demonstrate that obesity causes lung neutrophilia in otherwise normal mice, which is further exacerbated by the presence of a primary tumour. The increase in lung neutrophils translates to increased breast cancer metastasis to this site, in a GM-CSF- and IL5-dependent manner. Importantly, weight loss is sufficient to reverse this effect, and reduce serum levels of GM-CSF and IL5 in both mouse models and humans. Our data indicate that special consideration of the obese patient population is critical for effective management of cancer progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Interleucina-5/metabolismo , Neoplasias Pulmonares/metabolismo , Pulmón/metabolismo , Infiltración Neutrófila , Neutrófilos/metabolismo , Obesidad/metabolismo , Neumonía/metabolismo , Adiposidad , Traslado Adoptivo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Técnicas de Cocultivo , Dieta con Restricción de Grasas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/sangre , Interleucina-5/sangre , Pulmón/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Neutrófilos/patología , Neutrófilos/trasplante , Obesidad/complicaciones , Obesidad/dietoterapia , Obesidad/patología , Neumonía/etiología , Neumonía/patología , Neumonía/prevención & control , Transducción de Señal , Factores de Tiempo , Microambiente Tumoral , Pérdida de Peso
10.
Cancer Res ; 76(14): 4249-58, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27221706

RESUMEN

Abnormal architectures of collagen fibers in the extracellular matrix (ECM) are hallmarks of many invasive diseases, including cancer. Targeting specific stages of collagen assembly in vivo presents a great challenge due to the involvement of various crosslinking enzymes in the multistep, hierarchical process of ECM build-up. Using advanced microscopic tools, we monitored stages of fibrillary collagen assembly in a native fibroblast-derived 3D matrix system and identified anti-lysyl oxidase-like 2 (LOXL2) antibodies that alter the natural alignment and width of endogenic fibrillary collagens without affecting ECM composition. The disrupted collagen morphologies interfered with the adhesion and invasion properties of human breast cancer cells. Treatment of mice bearing breast cancer xenografts with the inhibitory antibodies resulted in disruption of the tumorigenic collagen superstructure and in reduction of primary tumor growth. Our approach could serve as a general methodology to identify novel therapeutics targeting fibrillary protein organization to treat ECM-associated pathologies. Cancer Res; 76(14); 4249-58. ©2016 AACR.


Asunto(s)
Neoplasias de la Mama/patología , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Aminoácido Oxidorreductasas/antagonistas & inhibidores , Aminoácido Oxidorreductasas/fisiología , Animales , Anticuerpos Monoclonales/inmunología , Línea Celular Tumoral , Proliferación Celular , Proteínas de la Matriz Extracelular/análisis , Femenino , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Microambiente Tumoral
11.
Sci Signal ; 8(368): ra29, 2015 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-25783158

RESUMEN

Growth factors promote tumor growth and metastasis. We found that epidermal growth factor (EGF) induced a set of 22 microRNAs (miRNAs) before promoting the migration of mammary cells. These miRNAs were more abundant in human breast tumors relative to the surrounding tissue, and their abundance varied among breast cancer subtypes. One of these miRNAs, miR-15b, targeted the 3' untranslated region of MTSS1 (metastasis suppressor protein 1). Although xenografts in which MTSS1 was knocked down grew more slowly in mice initially, longer-term growth was unaffected. Knocking down MTSS1 increased migration and Matrigel invasion of nontransformed mammary epithelial cells. Overexpressing MTSS1 in an invasive cell line decreased cell migration and invasiveness, decreased the formation of invadopodia and actin stress fibers, and increased the formation of cellular junctions. In tissues from breast cancer patients with the aggressive basal subtype, an inverse correlation occurred with the high expression of miRNA-15b and the low expression of MTSS1. Furthermore, low abundance of MTSS1 correlated with poor patient prognosis. Thus, growth factor-inducible miRNAs mediate mechanisms underlying the progression of cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Movimiento Celular , Factor de Crecimiento Epidérmico/metabolismo , MicroARNs/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas de Neoplasias/metabolismo , Animales , Neoplasias de la Mama/genética , Línea Celular Tumoral , Factor de Crecimiento Epidérmico/genética , Femenino , Xenoinjertos , Humanos , Ratones , Ratones SCID , MicroARNs/genética , Proteínas de Microfilamentos/genética , Proteínas de Neoplasias/genética , Trasplante de Neoplasias
12.
EMBO Mol Med ; 7(3): 299-314, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25678558

RESUMEN

Dissemination of primary tumor cells depends on migratory and invasive attributes. Here, we identify Navigator-3 (NAV3), a gene frequently mutated or deleted in human tumors, as a regulator of epithelial migration and invasion. Following induction by growth factors, NAV3 localizes to the plus ends of microtubules and enhances their polarized growth. Accordingly, NAV3 depletion trimmed microtubule growth, prolonged growth factor signaling, prevented apoptosis and enhanced random cell migration. Mathematical modeling suggested that NAV3-depleted cells acquire an advantage in terms of the way they explore their environment. In animal models, silencing NAV3 increased metastasis, whereas ectopic expression of the wild-type form, unlike expression of two, relatively unstable oncogenic mutants from human tumors, inhibited metastasis. Congruently, analyses of > 2,500 breast and lung cancer patients associated low NAV3 with shorter survival. We propose that NAV3 inhibits breast cancer progression by regulating microtubule dynamics, biasing directionally persistent rather than random migration, and inhibiting locomotion of initiated cells.


Asunto(s)
Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/patología , Movimiento Celular , Proteínas de la Membrana/metabolismo , Metástasis de la Neoplasia/patología , Proteínas del Tejido Nervioso/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Ratones
13.
Sci Signal ; 8(360): ra7, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25605973

RESUMEN

Amplified HER2, which encodes a member of the epidermal growth factor receptor (EGFR) family, is a target of effective therapies against breast cancer. In search for similarly targetable genomic aberrations, we identified copy number gains in SYNJ2, which encodes the 5'-inositol lipid phosphatase synaptojanin 2, as well as overexpression in a small fraction of human breast tumors. Copy gain and overexpression correlated with shorter patient survival and a low abundance of the tumor suppressor microRNA miR-31. SYNJ2 promoted cell migration and invasion in culture and lung metastasis of breast tumor xenografts in mice. Knocking down SYNJ2 impaired the endocytic recycling of EGFR and the formation of cellular lamellipodia and invadopodia. Screening compound libraries identified SYNJ2-specific inhibitors that prevented cell migration but did not affect the related neural protein SYNJ1, suggesting that SYNJ2 is a potentially druggable target to block cancer cell migration.


Asunto(s)
Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica/genética , Metástasis de la Neoplasia/genética , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/fisiopatología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Descubrimiento de Drogas , Receptores ErbB/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Dosificación de Gen , Humanos , Procesamiento de Imagen Asistido por Computador , Immunoblotting , Inmunohistoquímica , Ratones , Ratones SCID , Microscopía Electrónica de Rastreo , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Podosomas/genética , Podosomas/fisiología , Seudópodos/genética , Seudópodos/fisiología , ARN Interferente Pequeño/genética , Estadísticas no Paramétricas
14.
Nat Commun ; 5: 5437, 2014 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-25421715

RESUMEN

Cells cope with replication-blocking lesions via translesion DNA synthesis (TLS). TLS is carried out by low-fidelity DNA polymerases that replicate across lesions, thereby preventing genome instability at the cost of increased point mutations. Here we perform a two-stage siRNA-based functional screen for mammalian TLS genes and identify 17 validated TLS genes. One of the genes, NPM1, is frequently mutated in acute myeloid leukaemia (AML). We show that NPM1 (nucleophosmin) regulates TLS via interaction with the catalytic core of DNA polymerase-η (polη), and that NPM1 deficiency causes a TLS defect due to proteasomal degradation of polη. Moreover, the prevalent NPM1c+ mutation that causes NPM1 mislocalization in ~30% of AML patients results in excessive degradation of polη. These results establish the role of NPM1 as a key TLS regulator, and suggest a mechanism for the better prognosis of AML patients carrying mutations in NPM1.


Asunto(s)
Daño del ADN , Replicación del ADN , Leucemia Mieloide Aguda/metabolismo , Proteínas Nucleares/metabolismo , Línea Celular , Daño del ADN/efectos de la radiación , Reparación del ADN , Replicación del ADN/efectos de la radiación , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Humanos , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/genética , Proteínas Nucleares/genética , Nucleofosmina , Unión Proteica , Rayos Ultravioleta
15.
Nat Commun ; 5: 5073, 2014 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-25278152

RESUMEN

Signal transduction by receptor tyrosine kinases (RTKs) and nuclear receptors for steroid hormones is essential for body homeostasis, but the cross-talk between these receptor families is poorly understood. We observed that glucocorticoids inhibit signalling downstream of EGFR, an RTK. The underlying mechanism entails suppression of EGFR's positive feedback loops and simultaneous triggering of negative feedback loops that normally restrain EGFR. Our studies in mice reveal that the regulation of EGFR's feedback loops by glucocorticoids translates to circadian control of EGFR signalling: EGFR signals are suppressed by high glucocorticoids during the active phase (night-time in rodents), while EGFR signals are enhanced during the resting phase. Consistent with this pattern, treatment of animals bearing EGFR-driven tumours with a specific kinase inhibitor was more effective if administered during the resting phase of the day, when glucocorticoids are low. These findings support a circadian clock-based paradigm in cancer therapy.


Asunto(s)
Receptores ErbB/metabolismo , Glucocorticoides/metabolismo , Neoplasias/patología , Transducción de Señal , Animales , Línea Celular Tumoral , Movimiento Celular , Ritmo Circadiano , Progresión de la Enfermedad , Femenino , Humanos , Ligandos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oscilometría , Receptores de Glucocorticoides/metabolismo , Resultado del Tratamiento
16.
Cell Adh Migr ; 7(1): 33-7, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23076209

RESUMEN

Unlike the well-characterized checkpoints of the cell cycle, which establish commitment to cell division, signaling pathways and gene expression programs that commit cells to migration are incompletely understood. Apparently, several molecular switches are activated in response to an extracellular cue, such as the epidermal growth factor (EGF), and they simultaneously confer distinct features of an integrated motile phenotype. Here we review such early (transcription-independent) and late switches, in light of a novel ERK-ERF-EGR1 switch we recently reported in the FASEB Journal. The study employed human mammary cells and two stimuli: EGF, which induced mammary cell migration, and serum factors, which stimulated cell growth. By contrasting the underlying pathways we unveiled a cascade that allows the active form of the ERK mitogen-activated protein kinase (MAPK) cascade to export the ERF repressor from the nucleus, thereby permitting tightly balanced stimulation of an EGR1-centered gene expression program.

17.
PLoS One ; 8(12): e80566, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24324612

RESUMEN

Signal-induced transcript isoform variation (TIV) includes alternative promoter usage as well as alternative splicing and alternative polyadenylation of mRNA. To assess the phenotypic relevance of signal-induced TIV, we employed exon arrays and breast epithelial cells, which migrate in response to the epidermal growth factor (EGF). We show that EGF rapidly--within one hour--induces widespread TIV in a significant fraction of the transcriptome. Importantly, TIV characterizes many genes that display no differential expression upon stimulus. In addition, similar EGF-dependent changes are shared by a panel of mammary cell lines. A functional screen, which utilized isoform-specific siRNA oligonucleotides, indicated that several isoforms play essential, non-redundant roles in EGF-induced mammary cell migration. Taken together, our findings highlight the importance of TIV in the rapid evolvement of a phenotypic response to extracellular signals.


Asunto(s)
Factor de Crecimiento Epidérmico/farmacología , Células Epiteliales/efectos de los fármacos , Exones , Variación Genética , ARN Mensajero/genética , Transcriptoma , Empalme Alternativo , Línea Celular , Movimiento Celular/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Humanos , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/efectos de los fármacos , Glándulas Mamarias Humanas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Poliadenilación , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA