Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 147(2): 306-19, 2011 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-22000011

RESUMEN

Chronic myelogenous leukemia (CML) is caused by the constitutively active tyrosine kinase Bcr-Abl and treated with the tyrosine kinase inhibitor (TKI) imatinib. However, emerging TKI resistance prevents complete cure. Therefore, alternative strategies targeting regulatory modules of Bcr-Abl in addition to the kinase active site are strongly desirable. Here, we show that an intramolecular interaction between the SH2 and kinase domains in Bcr-Abl is both necessary and sufficient for high catalytic activity of the enzyme. Disruption of this interface led to inhibition of downstream events critical for CML signaling and, importantly, completely abolished leukemia formation in mice. Furthermore, disruption of the SH2-kinase interface increased sensitivity of imatinib-resistant Bcr-Abl mutants to TKI inhibition. An engineered Abl SH2-binding fibronectin type III monobody inhibited Bcr-Abl kinase activity both in vitro and in primary CML cells, where it induced apoptosis. This work validates the SH2-kinase interface as an allosteric target for therapeutic intervention.


Asunto(s)
Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Proteínas de Fusión bcr-abl/química , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/química , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Benzamidas , Células Cultivadas , Proteínas de Fusión bcr-abl/metabolismo , Humanos , Mesilato de Imatinib , Isoleucina/metabolismo , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/metabolismo , Pirimidinas/farmacología , Transducción de Señal , Dominios Homologos src
2.
Cell ; 138(4): 623-5, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19703390

RESUMEN

During metastasis, migrating breast cancer stem cells undergo a loss of polarity leading to an epithelial-to-mesenchymal transition (EMT). Gupta et al. (2009) use this attribute of cancer stem cells to develop a high-throughput screen, which successfully identifies small molecules that specifically inhibit cancer stem cell proliferation through the induction of differentiation.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Diferenciación Celular , Células Madre Neoplásicas/efectos de los fármacos , Animales , Células Epiteliales/citología , Humanos
3.
Nat Genet ; 39(6): 741-9, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17468757

RESUMEN

The mitogen-activated protein kinase (MAPK) p38alpha controls inflammatory responses and cell proliferation. Using mice carrying conditional Mapk14 (also known as p38alpha) alleles, we investigated its function in postnatal development and tumorigenesis. When we specifically deleted Mapk14 in the mouse embryo, fetuses developed to term but died shortly after birth, probably owing to lung dysfunction. Fetal hematopoietic cells and embryonic fibroblasts deficient in p38alpha showed increased proliferation resulting from sustained activation of the c-Jun N-terminal kinase (JNK)-c-Jun pathway. Notably, in chemical-induced liver cancer development, mice with liver-specific deletion of Mapk14 showed enhanced hepatocyte proliferation and tumor development that correlated with upregulation of the JNK-c-Jun pathway. Furthermore, inactivation of JNK or c-Jun suppressed the increased proliferation of Mapk14-deficient hepatocytes and tumor cells. These results demonstrate a new mechanism whereby p38alpha negatively regulates cell proliferation by antagonizing the JNK-c-Jun pathway in multiple cell types and in liver cancer development.


Asunto(s)
Proliferación Celular , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neoplasias Hepáticas Experimentales/metabolismo , Hígado/embriología , Proteína Quinasa 14 Activada por Mitógenos/fisiología , Proteínas Proto-Oncogénicas c-jun/metabolismo , Animales , Eritrocitos/citología , Eritrocitos/metabolismo , Eritrocitos/patología , Femenino , Perfilación de la Expresión Génica , Técnicas para Inmunoenzimas , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Hígado/citología , Hígado/metabolismo , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones , Ratones Noqueados , Proteína Quinasa 14 Activada por Mitógenos/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas c-jun/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-jun/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
4.
Breast Cancer Res ; 16(5): 433, 2014 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-25212966

RESUMEN

INTRODUCTION: Interleukin-like epithelial-to-mesenchymal transition inducer (ILEI) is an essential cytokine in tumor progression that is upregulated in several cancers, and its altered subcellular localization is a predictor of poor survival in human breast cancer. However, the regulation of ILEI activity and the molecular meaning of its altered localization remain elusive. METHODS: The influence of serum withdrawal, broad-specificity protease inhibitors, different serine proteases and plasminogen depletion on the size and amount of the secreted ILEI protein was investigated by Western blot analysis of EpRas cells. Proteases with ILEI-processing capacity were identified by carrying out an in vitro cleavage assay. Murine mammary tumor and metastasis models of EpC40 and 4T1 cells overexpressing different mutant forms of ILEI were used-extended with in vivo aprotinin treatment for the inhibition of ILEI-processing proteases-to test the in vivo relevance of proteolytic cleavage. Stable knockdown of urokinase plasminogen activator receptor (uPAR) in EpRas cells was performed to investigate the involvement of uPAR in ILEI secretion. The subcellular localization of the ILEI protein in tumor cell lines was analyzed by immunofluorescence. Immunohistochemistry for ILEI localization and uPAR expression was performed on two human breast cancer arrays, and ILEI and uPAR scores were correlated with the metastasis-free survival of patients. RESULTS: We demonstrate that secreted ILEI requires site-specific proteolytic maturation into its short form for its tumor-promoting function, which is executed by serine proteases, most efficiently by plasmin. Noncleaved ILEI is tethered to fibronectin-containing fibers of the extracellular matrix through a propeptide-dependent interaction. In addition to ILEI processing, plasmin rapidly increases ILEI secretion by mobilizing its intracellular protein pool in a uPAR-dependent manner. Elevated ILEI secretion correlates with an altered subcellular localization of the protein, most likely representing a shift into secretory vesicles. Moreover, altered subcellular ILEI localization strongly correlates with high tumor cell-associated uPAR protein expression, as well as with poor survival, in human breast cancer. CONCLUSIONS: Our findings point out extracellular serine proteases, in particular plasmin, and uPAR as valuable therapeutic targets against ILEI-driven tumor progression and emphasize the prognostic relevance of ILEI localization and a combined ILEI-uPAR marker analysis in human breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Citocinas/fisiología , Neoplasias Pulmonares/metabolismo , Proteínas de Neoplasias/fisiología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Animales , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Línea Celular Tumoral , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Femenino , Fibrinolisina/metabolismo , Humanos , Estimación de Kaplan-Meier , Elastasa de Leucocito/metabolismo , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/secundario , Ratones Desnudos , Trasplante de Neoplasias , Calicreína Plasmática/metabolismo , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Proteolisis
5.
Cancer Cell ; 10(3): 227-39, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16959614

RESUMEN

Erk/MAPK and TGFbeta signaling cause epithelial to mesenchymal transition (EMT) and metastasis in mouse mammary epithelial cells (EpH4) transformed with oncogenic Ras (EpRas). In trials to unravel underlying mechanisms, expression profiling for EMT-specific genes identified a secreted interleukin-related protein (ILEI), upregulated exclusively at the translational level. Stable overexpression of ILEI in EpH4 and EpRas cells caused EMT, tumor growth, and metastasis, independent of TGFbeta-R signaling and enhanced by Bcl2. RNAi-mediated knockdown of ILEI in EpRas cells before and after EMT (EpRasXT) prevented and reverted TGFbeta-dependent EMT, also abrogating metastasis formation. ILEI is overexpressed and/or altered in intracellular localization in multiple human tumors, an event strongly correlated to invasion/EMT, metastasis formation, and survival in human colon and breast cancer.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Citocinas/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células Madre Mesenquimatosas/citología , Proteínas de Neoplasias/metabolismo , Animales , Diferenciación Celular , Línea Celular , Citocinas/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia/patología , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Neoplasias/metabolismo , Neoplasias/patología , Pronóstico , Biosíntesis de Proteínas/genética , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Tasa de Supervivencia , Factores de Tiempo , Factor de Crecimiento Transformador beta/metabolismo
6.
Cancer Cell ; 10(1): 77-87, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16843267

RESUMEN

The tumor suppressor STAT1 is considered a key regulator of the surveillance of developing tumors. Here, we describe an unexpected tumor-promoting role for STAT1 in leukemia. STAT1(-/-) mice are partially protected from leukemia development, and STAT1(-/-) tumor cells induce leukemia in RAG2(-/-) and immunocompetent mice with increased latency. The low MHC class I protein levels of STAT1(-/-) tumor cells enable efficient NK cell lysis and account for the enhanced tumor clearance. Strikingly, STAT1(-/-) tumor cells acquire increased MHC class I expression upon leukemia progression. These findings define STAT1 as a tumor promoter in leukemia development. Furthermore, we describe the upregulation of MHC class I expression as a general mechanism that allows for the escape of hematopoietic malignancies from immune surveillance.


Asunto(s)
Leucemia Experimental/patología , Factor de Transcripción STAT1/fisiología , Animales , Linfocitos B/metabolismo , Linfocitos B/patología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/genética , Transformación Celular Neoplásica/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Genotipo , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Interferón gamma/genética , Interferón gamma/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Leucemia Experimental/genética , Leucemia Experimental/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Oncogénicas v-abl/genética , Proteínas Oncogénicas v-abl/metabolismo , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Fenotipo , Factor de Transcripción STAT1/deficiencia , Factor de Transcripción STAT1/genética , Células Madre/metabolismo , Células Madre/patología , Análisis de Supervivencia
7.
Nucleic Acids Res ; 40(1): 290-302, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21896617

RESUMEN

The majority of transcripts that harbor an internal ribosome entry site (IRES) are involved in cancer development via corresponding proteins. A crucial event in tumor progression referred to as epithelial to mesenchymal transition (EMT) allows carcinoma cells to acquire invasive properties. The translational activation of the extracellular matrix component laminin B1 (LamB1) during EMT has been recently reported suggesting an IRES-mediated mechanism. In this study, the IRES activity of LamB1 was determined by independent bicistronic reporter assays. Strong evidences exclude an impact of cryptic promoter or splice sites on IRES-driven translation of LamB1. Furthermore, no other LamB1 mRNA species arising from alternative transcription start sites or polyadenylation signals were detected that account for its translational control. Mapping of the LamB1 5'-untranslated region (UTR) revealed the minimal LamB1 IRES motif between -293 and -1 upstream of the start codon. Notably, RNA affinity purification showed that the La protein interacts with the LamB1 IRES. This interaction and its regulation during EMT were confirmed by ribonucleoprotein immunoprecipitation. In addition, La was able to positively modulate LamB1 IRES translation. In summary, these data indicate that the LamB1 IRES is activated by binding to La which leads to translational upregulation during hepatocellular EMT.


Asunto(s)
Regiones no Traducidas 5' , Autoantígenos/metabolismo , Transición Epitelial-Mesenquimal/genética , Laminina/genética , Biosíntesis de Proteínas , Ribonucleoproteínas/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Humanos , Laminina/biosíntesis , Ratones , Neoplasias/genética , Motivos de Nucleótidos , Empalme del ARN , ARN Mensajero/metabolismo , Transcripción Genética , Antígeno SS-B
8.
Proc Natl Acad Sci U S A ; 108(5): 1903-7, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21245337

RESUMEN

Tissue differentiation is an important process that involves major cellular membrane remodeling. We used Madin-Darby canine kidney cells as a model for epithelium formation and investigated the remodeling of the total cell membrane lipidome during the transition from a nonpolarized morphology to an epithelial morphology and vice versa. To achieve this, we developed a shotgun-based lipidomics workflow that enabled the absolute quantification of mammalian membrane lipidomes with minimal sample processing from low sample amounts. Epithelial morphogenesis was accompanied by a major shift from sphingomyelin to glycosphingolipid, together with an increase in plasmalogen, phosphatidylethanolamine, and cholesterol content, whereas the opposite changes took place during an epithelial-to-mesenchymal transition. Moreover, during polarization, the sphingolipids became longer, more saturated, and more hydroxylated as required to generate an apical membrane domain that serves as a protective barrier for the epithelial sheet.


Asunto(s)
Lípidos de la Membrana/metabolismo , Animales , Línea Celular , Perros , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Morfogénesis
9.
Cancer Cell ; 7(1): 87-99, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15652752

RESUMEN

Activation of Stat5 is frequently found in leukemias. To study the mechanism and role of Stat5 activation, we introduced a constitutively activated Stat5a mutant, cS5F, into murine bone marrow (BM) cells. BM transplantation with cS5F-transfected cells caused development of multilineage leukemias in lethally irradiated wild-type or nonirradiated Rag2(-/-) mice. The leukemic cells showed strongly enhanced levels of cS5F tetramers but unchanged cS5F dimer levels in a DNA binding assay. Moreover, Stat5a mutants engineered to form only dimers, but not tetramers, failed to induce leukemias. In addition, Stat5 tetramers were found to accumulate in excess compared to dimers in various human leukemias. These data suggest that Stat5 tetramers are associated with leukemogenesis.


Asunto(s)
Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Leucemia/metabolismo , Proteínas de la Leche/química , Proteínas de la Leche/metabolismo , Estructura Cuaternaria de Proteína , Transactivadores/química , Transactivadores/metabolismo , Animales , Biomarcadores , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Trasplante de Médula Ósea , Linaje de la Célula , Transformación Celular Neoplásica , Células Cultivadas , Proteínas de Unión al ADN/genética , Femenino , Prueba de Complementación Genética , Sustancias de Crecimiento/metabolismo , Humanos , Leucemia/genética , Leucemia/patología , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Proteínas de la Leche/genética , Mutación , Proteínas Nucleares , Oncogenes , Factor de Transcripción STAT5 , Bazo/metabolismo , Bazo/patología , Transactivadores/genética , Transfección , Proteínas Supresoras de Tumor
10.
Blood ; 116(9): 1548-58, 2010 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-20508164

RESUMEN

Stat5 transcription factors are essential gene regulators promoting proliferation, survival, and differentiation of all hematopoietic cell types. Mutations or fusions of oncogenic tyrosine kinases often result in constitutive Stat5 activation. We have modeled persistent Stat5 activity by using an oncogenic Stat5a variant (cS5). To analyze the hitherto unrecognized role of Stat5 serine phosphorylation in this context, we have generated cS5 constructs with mutated C-terminal serines 725 and 779, either alone or in combination. Genetic complementation assays in primary Stat5(null/null) mast cells and Stat5(DeltaN) T cells demonstrated reconstitution of proliferation with these mutants. Similarly, an in vivo reconstitution experiment of transduced Stat5(null/null) fetal liver cells transplanted into irradiated wild-type recipients revealed that these mutants exhibit biologic activity in lineage differentiation. By contrast, the leukemogenic potential of cS5 in bone marrow transplants decreased dramatically in cS5 single-serine mutants or was completely absent upon loss of both serine phosphorylation sites. Our data suggest that Stat5a serine phosphorylation is a prerequisite for cS5-mediated leukemogenesis. Hence, interference with Stat5a serine phosphorylation might provide a new therapeutic option for leukemia and myeloid dysplasias without affecting major functions of Stat5 in normal hematopoiesis.


Asunto(s)
Transformación Celular Neoplásica , Hematopoyesis/fisiología , Leucemia/patología , Factor de Transcripción STAT5/metabolismo , Serina/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Adulto , Anciano , Animales , Western Blotting , Trasplante de Médula Ósea , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Femenino , Feto , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Leucemia/genética , Leucemia/metabolismo , Trasplante de Hígado , Masculino , Mastocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Fosforilación , Células Precursoras de Linfocitos B/metabolismo , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT5/genética , Serina/genética , Linfocitos T/metabolismo , Proteínas Supresoras de Tumor/genética
11.
Curr Opin Cell Biol ; 17(5): 548-58, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16098727

RESUMEN

Epithelial-mesenchymal transitions (EMTs) occur as key steps during embryonic morphogenesis, and are now implicated in the progression of primary tumors towards metastases. Recent advances have fostered a more detailed understanding of molecular mechanisms and networks governing EMT in tumor progression. Besides TGFbeta and RTK/Ras signaling, autocrine factors and Wnt-, Notch-, Hedgehog- and NF-kappaB-dependent pathways were found to contribute to EMT. Repression of E-cadherin by transcriptional regulators such as Snail or Twist emerges as one critical step driving EMT, and this stage is currently being molecularly linked with many of the new players. Increasing evidence suggests that EMT plays a specific role in the migration of cells from a primary tumor into the circulation and may provide a rationale for developing more effective cancer therapies.


Asunto(s)
Epitelio/metabolismo , Mesodermo/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Movimiento Celular/fisiología , Polaridad Celular/fisiología , Proliferación Celular , Progresión de la Enfermedad , Epitelio/fisiología , Regulación Neoplásica de la Expresión Génica , Mesodermo/fisiología , Modelos Biológicos , Metástasis de la Neoplasia/fisiopatología , Transducción de Señal
12.
Am J Pathol ; 176(1): 472-81, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20008139

RESUMEN

Transforming growth factor-beta cooperates with oncogenic Ras to activate nuclear beta-catenin during the epithelial to mesenchymal transition of hepatocytes, a process relevant in the progression of hepatocellular carcinoma (HCC). In this study we investigated the role of beta-catenin in the differentiation of murine, oncogene-targeted hepatocytes and in 133 human HCC patients scheduled for orthotopic liver transplantation. Transforming growth factor-beta caused dissociation of plasma membrane E-cadherin/beta-catenin complexes and accumulation of nuclear beta-catenin in Ras-transformed, but otherwise normal hepatocytes in p19(ARF)-/- mice. Both processes were inhibited by Smad7-mediated disruption of transforming growth factor-beta signaling. Overexpression of constitutively active beta-catenin resulted in high levels of CK19 and M2-PK, whereas ablation of beta-catenin by axin overexpression caused strong expression of CK8 and CK18. Therefore, nuclear beta-catenin resulted in dedifferentiation of neoplastic hepatocytes to immature progenitor cells, whereas loss of nuclear beta-catenin led to a differentiated HCC phenotype. Poorly differentiated human HCC showed cytoplasmic redistribution or even loss of E-cadherin, suggesting epithelial to mesenchymal transition. Analysis of 133 HCC patient samples revealed that 58.6% of human HCC exhibited strong nuclear beta-catenin accumulation, which correlated with clinical features such as vascular invasion and recurrence of disease after orthotopic liver transplantation. These data suggest that activation of beta-catenin signaling causes dedifferentiation to malignant, immature hepatocyte progenitors and facilitates recurrence of human HCC after orthotopic liver transplantation.


Asunto(s)
Carcinoma Hepatocelular/patología , Núcleo Celular/metabolismo , Neoplasias Hepáticas/patología , Hígado/patología , Recurrencia Local de Neoplasia/metabolismo , Células Madre/patología , beta Catenina/metabolismo , Animales , Cadherinas/metabolismo , Carcinoma Hepatocelular/irrigación sanguínea , Carcinoma Hepatocelular/metabolismo , Diferenciación Celular , Membrana Celular/metabolismo , Epitelio/metabolismo , Epitelio/patología , Femenino , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Hígado/metabolismo , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/metabolismo , Trasplante de Hígado , Masculino , Mesodermo/metabolismo , Mesodermo/patología , Ratones , Neovascularización Patológica/complicaciones , Fenotipo , Transporte de Proteínas , Transducción de Señal , Proteína smad7/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
13.
Nat Cell Biol ; 6(9): 899-905, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15322555

RESUMEN

Continuously proliferating cells exactly double their mass during each cell cycle. Here we have addressed the controversial question of if and how cell size is sensed and regulated. We used erythroblasts that proliferate under the control of a constitutively active oncogene (v-ErbB) or under the control of physiological cytokines (stem cell factor, erythropoietin and v-ErbB inhibitor). The oncogene-driven cells proliferated 1.7 times faster and showed a 1.5-fold increase in cell volume. The two phenotypes could be converted into each other 24 h after altering growth factor signalling. The large cells had a higher rate of protein synthesis, together with a shortened G1 phase. Additional experiments with chicken erythroblasts and mouse fibroblasts, synchronized by centrifugal elutriation, provided further evidence that vertebrate cells can respond to cell size alterations (induced either through different growth factor signalling or DNA synthesis inhibitors) by compensatory shortening of the subsequent G1 phase. Taken together, these data suggest that an active size threshold mechanism exists in G1, which induces adjustment of cell-cycle length in the next cycle, thus ensuring maintenance of a proper balance between growth and proliferation rates in vertebrates.


Asunto(s)
Eritroblastos/citología , Fase G1/fisiología , Animales , División Celular , Fenómenos Fisiológicos Celulares , Tamaño de la Célula , Pollos , Humanos , Cinética , Ratones , Modelos Biológicos , Biosíntesis de Proteínas , Fase S/fisiología , Factores de Tiempo
14.
J Exp Med ; 196(10): 1347-53, 2002 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-12438425

RESUMEN

The Raf kinases are key signal transducers activated by mitogens or oncogenes. The best studied Raf isoform, Raf-1, was identified as an inhibitor of apoptosis by conventional and conditional gene ablation in mice. c-raf-1(-)(/)(-) embryos are growth retarded and anemic, and die at midgestation with anomalies in the placenta and fetal liver. Here, we show that Raf-1-deficient primary erythroblasts cannot be expanded in culture due to their accelerated differentiation into mature erythrocytes. In addition, Raf-1 expression is down-regulated in differentiating wild-type cells, whereas overexpression of activated Raf-1 delays differentiation. As recently described for human erythroid precursors, we find that caspase activation is necessary for the differentiation of murine fetal liver erythroblasts. Differentiation-associated caspase activation is accelerated in erythroid progenitors lacking Raf-1 and delayed by overexpression of the activated kinase. These results reveal an essential function of Raf-1 in erythropoiesis and demonstrate that the ability of Raf-1 to restrict caspase activation is biologically relevant in a context distinct from apoptosis.


Asunto(s)
Caspasas/metabolismo , Diferenciación Celular/fisiología , Activación Enzimática/fisiología , Eritrocitos/citología , Proteínas Proto-Oncogénicas c-raf/fisiología , Secuencia de Aminoácidos , Animales , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-raf/genética
15.
J Exp Med ; 199(6): 785-95, 2004 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15007095

RESUMEN

Regulation of survival, expansion, and differentiation of erythroid progenitors requires the well-controlled activity of signaling pathways induced by erythropoietin (Epo) and stem cell factor (SCF). In addition to qualitative regulation of signaling pathways, quantitative control may be essential to control appropriate cell numbers in peripheral blood. We demonstrate that Bruton's tyrosine kinase (Btk) is able to associate with the Epo receptor (EpoR) and Jak2, and is a substrate of Jak2. Deficiency of Btk results in reduced and delayed phosphorylation of the EpoR, Jak2, and downstream signaling molecules such as Stat5 and PLCgamma1 as well as in decreased responsiveness to Epo. As a result, expansion of erythroid progenitors lacking Btk is impaired at limiting concentrations of Epo and SCF. In addition, we show that SCF induces Btk to interact with TNF-related apoptosis-inducing ligand (TRAIL)-receptor 1 and that lack of Btk results in increased sensitivity to TRAIL-induced apoptosis. Together, our results indicate that Btk is a novel, quantitative regulator of Epo/SCF-dependent expansion and survival in erythropoiesis.


Asunto(s)
Células Precursoras Eritroides/fisiología , Eritropoyetina/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas , Transducción de Señal/fisiología , Factor de Células Madre/metabolismo , Agammaglobulinemia Tirosina Quinasa , Animales , Anticuerpos Monoclonales , Western Blotting , Células COS , Línea Celular , Chlorocebus aethiops , Eritropoyetina/fisiología , Citometría de Flujo , Hemoglobinas/metabolismo , Janus Quinasa 2 , Plásmidos/genética , Pruebas de Precipitina , Receptores de Eritropoyetina/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Células Madre/fisiología , Transfección
16.
Blood ; 112(9): 3878-88, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18694996

RESUMEN

Erythropoiesis strictly depends on signal transduction through the erythropoietin receptor (EpoR)-Janus kinase 2 (Jak2)-signal transducer and activator of transcription 5 (Stat5) axis, regulating proliferation, differentiation, and survival. The exact role of the transcription factor Stat5 in erythropoiesis remained puzzling, however, since the first Stat5-deficient mice carried a hypomorphic Stat5 allele, impeding full phenotypical analysis. Using mice completely lacking Stat5--displaying early lethality--we demonstrate that these animals suffer from microcytic anemia due to reduced expression of the antiapoptotic proteins Bcl-x(L) and Mcl-1 followed by enhanced apoptosis. Moreover, transferrin receptor-1 (TfR-1) cell surface levels on erythroid cells were decreased more than 2-fold on erythroid cells of Stat5(-/-) animals. This reduction could be attributed to reduced transcription of TfR-1 mRNA and iron regulatory protein 2 (IRP-2), the major translational regulator of TfR-1 mRNA stability in erythroid cells. Both genes were demonstrated to be direct transcriptional targets of Stat5. This establishes an unexpected mechanistic link between EpoR/Jak/Stat signaling and iron metabolism, processes absolutely essential for erythropoiesis and life.


Asunto(s)
Células Eritroides/metabolismo , Proteína 2 Reguladora de Hierro/metabolismo , Hierro/metabolismo , Receptores de Transferrina/metabolismo , Factor de Transcripción STAT5/metabolismo , Anemia Ferropénica/genética , Anemia Ferropénica/metabolismo , Anemia Ferropénica/patología , Animales , Apoptosis , Transporte Biológico Activo , Pérdida del Embrión , Células Eritroides/patología , Femenino , Deficiencias de Hierro , Hígado/embriología , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Embarazo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción STAT5/deficiencia , Factor de Transcripción STAT5/genética
17.
Blood ; 112(7): 2750-60, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18625885

RESUMEN

Stem cell factor (SCF)-induced activation of phosphoinositide-3-kinase (PI3K) is required for transient amplification of the erythroblast compartment. PI3K stimulates the activation of mTOR (target of rapamycin) and subsequent release of the cap-binding translation initiation factor 4E (eIF4E) from the 4E-binding protein 4EBP, which controls the recruitment of structured mRNAs to polysomes. Enhanced expression of eIF4E renders proliferation of erythroblasts independent of PI3K. To investigate which mRNAs are selectively recruited to polysomes, we compared SCF-dependent gene expression between total and polysome-bound mRNA. This identified 111 genes primarily subject to translational regulation. For 8 of 9 genes studied in more detail, the SCF-induced polysome recruitment of transcripts exceeded 5-fold regulation and was PI3K-dependent and eIF4E-sensitive, whereas total mRNA was not affected by signal transduction. One of the targets, Immunoglobulin binding protein 1 (Igbp1), is a regulatory subunit of protein phosphatase 2A (Pp2a) sustaining mTOR signaling. Constitutive expression of Igbp1 impaired erythroid differentiation, maintained 4EBP and p70S6k phosphorylation, and enhanced polysome recruitment of multiple eIF4E-sensitive mRNAs. Thus, PI3K-dependent polysome recruitment of Igbp1 acts as a positive feedback mechanism on translation initiation underscoring the important regulatory role of selective mRNA recruitment to polysomes in the balance between proliferation and maturation of erythroblasts.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Eritroides/citología , Células Eritroides/efectos de los fármacos , Retroalimentación Fisiológica/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Factor de Células Madre/farmacología , Proliferación Celular/efectos de los fármacos , Análisis por Conglomerados , Activación Enzimática/efectos de los fármacos , Eritroblastos/citología , Eritroblastos/efectos de los fármacos , Eritropoyetina/farmacología , Factor 4E Eucariótico de Iniciación/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Polirribosomas/efectos de los fármacos , Polirribosomas/enzimología , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Serina-Treonina Quinasas TOR , Factor de Crecimiento Transformador beta/farmacología
18.
Blood ; 111(9): 4511-22, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18239084

RESUMEN

Erythropoiesis requires erythropoietin (Epo) and stem cell factor (SCF) signaling via their receptors EpoR and c-Kit. EpoR, like many other receptors involved in hematopoiesis, acts via the kinase Jak2. Deletion of EpoR or Janus kinase 2 (Jak2) causes embryonic lethality as a result of defective erythropoiesis. The contribution of distinct EpoR/Jak2-induced signaling pathways (mitogen-activated protein kinase, phosphatidylinositol 3-kinase, signal transducer and activator of transcription 5 [Stat5]) to functional erythropoiesis is incompletely understood. Here we demonstrate that expression of a constitutively activated Stat5a mutant (cS5) was sufficient to relieve the proliferation defect of Jak2(-/-) and EpoR(-/-) cells in an Epo-independent manner. In addition, tamoxifen-induced DNA binding of a Stat5a-estrogen receptor (ER)* fusion construct enabled erythropoiesis in the absence of Epo. Furthermore, c-Kit was able to enhance signaling through the Jak2-Stat5 axis, particularly in lymphoid and myeloid progenitors. Although abundance of hematopoietic stem cells was 2.5-fold reduced in Jak2(-/-) fetal livers, transplantation of Jak2(-/-)-cS5 fetal liver cells into irradiated mice gave rise to mature erythroid and myeloid cells of donor origin up to 6 months after transplantation. Cytokine- and c-Kit pathways do not function independently of each other in hematopoiesis but cooperate to attain full Jak2/Stat5 activation. In conclusion, activated Stat5 is a critical downstream effector of Jak2 in erythropoiesis/myelopoiesis, and Jak2 functionally links cytokine- with c-Kit-receptor tyrosine kinase signaling.


Asunto(s)
Eritropoyesis , Janus Quinasa 2 , Receptores de Eritropoyetina , Factor de Transcripción STAT5/metabolismo , Animales , Células Cultivadas , Citocinas/metabolismo , Ratones , Ratones Noqueados , Mielopoyesis , Proteínas Proto-Oncogénicas c-kit/metabolismo
19.
Mol Cell Biol ; 27(10): 3839-3854, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17353275

RESUMEN

The cooperation of stem cell factor (SCF) and erythropoietin (Epo) is required to induce renewal divisions in erythroid progenitors, whereas differentiation to mature erythrocytes requires the presence of Epo only. Epo and SCF activate common signaling pathways such as the activation of protein kinase B (PKB) and the subsequent phosphorylation and inactivation of Foxo3a. In contrast, only Epo activates Stat5. Both Foxo3a and Stat5 promote erythroid differentiation. To understand the interplay of SCF and Epo in maintaining the balance between renewal and differentiation during erythroid development, we investigated differential Foxo3a target regulation by Epo and SCF. Expression profiling revealed that a subset of Foxo3a targets was not inhibited but was activated by Epo. One of these genes was Cited2. Transcriptional control of Epo/Foxo3a-induced Cited2 was studied and compared with that of the Epo-repressed Foxo3a target Btg1. We show that in response to Epo, the allegedly growth-inhibitory factor Foxo3a associates with the allegedly growth-stimulatory factor Stat5 in the nucleus, which is required for Epo-induced Cited2 expression. In contrast, Btg1 expression is controlled by the cooperation of Foxo3a with cyclic AMP- and Jun kinase-dependent Creb family members. Thus, Foxo3a not only is an effector of PKB but also integrates distinct signals to regulate gene expression in erythropoiesis.


Asunto(s)
Eritropoyesis/fisiología , Eritropoyetina/metabolismo , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Factor de Células Madre/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular/fisiología , Células Cultivadas , Análisis por Conglomerados , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Perfilación de la Expresión Génica , Humanos , Ratones , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Alineación de Secuencia , Transducción de Señal/fisiología , Transactivadores/genética , Transactivadores/metabolismo
20.
J Clin Invest ; 116(6): 1561-70, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16741576

RESUMEN

Metastasis is the major cause of cancer morbidity, but strategies for direct interference with invasion processes are lacking. Dedifferentiated, late-stage tumor cells secrete multiple factors that represent attractive targets for therapeutic intervention. Here we show that metastatic potential of oncogenic mammary epithelial cells requires an autocrine PDGF/PDGFR loop, which is established as a consequence of TGF-beta-induced epithelial-mesenchymal transition (EMT), a faithful in vitro correlate of metastasis. The cooperation of autocrine PDGFR signaling with oncogenic Ras hyperactivates PI3K and is required for survival during EMT. Autocrine PDGFR signaling also contributes to maintenance of EMT, possibly through activation of STAT1 and other distinct pathways. Inhibition of PDGFR signaling interfered with EMT and caused apoptosis in murine and human mammary carcinoma cell lines. Consequently, overexpression of a dominant-negative PDGFR or application of the established cancer drug STI571 interfered with experimental metastasis in mice. Similarly, in mouse mammary tumor virus-Neu (MMTV-Neu) transgenic mice, TGF-beta enhanced metastasis of mammary tumors, induced EMT, and elevated PDGFR signaling. Finally, expression of PDGFRalpha and -beta correlated with invasive behavior in human mammary carcinomas. Thus, autocrine PDGFR signaling plays an essential role during cancer progression, suggesting a novel application of STI571 to therapeutically interfere with metastasis.


Asunto(s)
Comunicación Autocrina , Neoplasias de la Mama , Neoplasias Mamarias Experimentales , Metástasis de la Neoplasia , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/fisiología , Animales , Antineoplásicos/metabolismo , Apoptosis , Benzamidas , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Diferenciación Celular/fisiología , Línea Celular Tumoral , Activación Enzimática , Células Epiteliales/citología , Células Epiteliales/fisiología , Femenino , Humanos , Mesilato de Imatinib , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Virus del Tumor Mamario del Ratón/genética , Virus del Tumor Mamario del Ratón/metabolismo , Mesodermo/fisiología , Ratones , Ratones Desnudos , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/metabolismo , Piperazinas/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo , Pirimidinas/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Proteínas ras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA