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(34): 13898-903, 2013 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-23918390

RESUMEN

Mechanotransduction in the mammalian auditory system depends on mechanosensitive channels in the hair bundles that project from the apical surface of the sensory hair cells. Individual stereocilia within each bundle contain a core of tightly packed actin filaments, whose length is dynamically regulated during development and in the adult. We show that the actin-binding protein epidermal growth factor receptor pathway substrate 8 (Eps8)L2, a member of the Eps8-like protein family, is a newly identified hair bundle protein that is localized at the tips of stereocilia of both cochlear and vestibular hair cells. It has a spatiotemporal expression pattern that complements that of Eps8. In the cochlea, whereas Eps8 is essential for the initial elongation of stereocilia, Eps8L2 is required for their maintenance in adult hair cells. In the absence of both proteins, the ordered staircase structure of the hair bundle in the cochlea decays. In contrast to the early profound hearing loss associated with an absence of Eps8, Eps8L2 null-mutant mice exhibit a late-onset, progressive hearing loss that is directly linked to a gradual deterioration in hair bundle morphology. We conclude that Eps8L2 is required for the long-term maintenance of the staircase structure and mechanosensory function of auditory hair bundles. It complements the developmental role of Eps8 and is a candidate gene for progressive age-related hearing loss.


Asunto(s)
Células Ciliadas Auditivas/patología , Pérdida Auditiva/genética , Proteínas de Microfilamentos/deficiencia , Análisis de Varianza , Animales , Audiometría de Respuesta Evocada , Células Ciliadas Auditivas/fisiología , Células Ciliadas Auditivas/ultraestructura , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/genética , Microscopía Electrónica , Técnicas de Placa-Clamp
2.
PLoS Biol ; 9(4): e1001048, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21526224

RESUMEN

Hair cells of the mammalian cochlea are specialized for the dynamic coding of sound stimuli. The transduction of sound waves into electrical signals depends upon mechanosensitive hair bundles that project from the cell's apical surface. Each stereocilium within a hair bundle is composed of uniformly polarized and tightly packed actin filaments. Several stereociliary proteins have been shown to be associated with hair bundle development and function and are known to cause deafness in mice and humans when mutated. The growth of the stereociliar actin core is dynamically regulated at the actin filament barbed ends in the stereociliary tip. We show that Eps8, a protein with actin binding, bundling, and barbed-end capping activities in other systems, is a novel component of the hair bundle. Eps8 is localized predominantly at the tip of the stereocilia and is essential for their normal elongation and function. Moreover, we have found that Eps8 knockout mice are profoundly deaf and that IHCs, but not OHCs, fail to mature into fully functional sensory receptors. We propose that Eps8 directly regulates stereocilia growth in hair cells and also plays a crucial role in the physiological maturation of mammalian cochlear IHCs. Together, our results indicate that Eps8 is critical in coordinating the development and functionality of mammalian auditory hair cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Extensiones de la Superficie Celular/metabolismo , Cóclea/fisiología , Proteínas del Citoesqueleto/metabolismo , Células Ciliadas Auditivas/metabolismo , Estimulación Acústica , Potenciales de Acción , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Calcio/metabolismo , Canales de Calcio/metabolismo , Extensiones de la Superficie Celular/ultraestructura , Cóclea/citología , Cóclea/crecimiento & desarrollo , Proteínas del Citoesqueleto/genética , Sordera/genética , Potenciales Evocados Auditivos del Tronco Encefálico , Exocitosis , Eliminación de Gen , Células Ciliadas Auditivas/ultraestructura , Mecanotransducción Celular , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Canales de Potasio/metabolismo
4.
PLoS Biol ; 8(6): e1000387, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20532239

RESUMEN

Actin capping and cross-linking proteins regulate the dynamics and architectures of different cellular protrusions. Eps8 is the founding member of a unique family of capping proteins capable of side-binding and bundling actin filaments. However, the structural basis through which Eps8 exerts these functions remains elusive. Here, we combined biochemical, molecular, and genetic approaches with electron microscopy and image analysis to dissect the molecular mechanism responsible for the distinct activities of Eps8. We propose that bundling activity of Eps8 is mainly mediated by a compact four helix bundle, which is contacting three actin subunits along the filament. The capping activity is mainly mediated by a amphipathic helix that binds within the hydrophobic pocket at the barbed ends of actin blocking further addition of actin monomers. Single-point mutagenesis validated these modes of binding, permitting us to dissect Eps8 capping from bundling activity in vitro. We further showed that the capping and bundling activities of Eps8 can be fully dissected in vivo, demonstrating the physiological relevance of the identified Eps8 structural/functional modules. Eps8 controls actin-based motility through its capping activity, while, as a bundler, is essential for proper intestinal morphogenesis of developing Caenorhabditis elegans.


Asunto(s)
Actinas/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Espectrometría de Masas , Microscopía Electrónica , Modelos Moleculares , Unión Proteica , Termodinámica
5.
PLoS Biol ; 7(6): e1000138, 2009 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-19564905

RESUMEN

The regulation of filopodia plays a crucial role during neuronal development and synaptogenesis. Axonal filopodia, which are known to originate presynaptic specializations, are regulated in response to neurotrophic factors. The structural components of filopodia are actin filaments, whose dynamics and organization are controlled by ensembles of actin-binding proteins. How neurotrophic factors regulate these latter proteins remains, however, poorly defined. Here, using a combination of mouse genetic, biochemical, and cell biological assays, we show that genetic removal of Eps8, an actin-binding and regulatory protein enriched in the growth cones and developing processes of neurons, significantly augments the number and density of vasodilator-stimulated phosphoprotein (VASP)-dependent axonal filopodia. The reintroduction of Eps8 wild type (WT), but not an Eps8 capping-defective mutant, into primary hippocampal neurons restored axonal filopodia to WT levels. We further show that the actin barbed-end capping activity of Eps8 is inhibited by brain-derived neurotrophic factor (BDNF) treatment through MAPK-dependent phosphorylation of Eps8 residues S624 and T628. Additionally, an Eps8 mutant, impaired in the MAPK target sites (S624A/T628A), displays increased association to actin-rich structures, is resistant to BDNF-mediated release from microfilaments, and inhibits BDNF-induced filopodia. The opposite is observed for a phosphomimetic Eps8 (S624E/T628E) mutant. Thus, collectively, our data identify Eps8 as a critical capping protein in the regulation of axonal filopodia and delineate a molecular pathway by which BDNF, through MAPK-dependent phosphorylation of Eps8, stimulates axonal filopodia formation, a process with crucial impacts on neuronal development and synapse formation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factor Neurotrófico Derivado del Encéfalo/farmacología , Proteínas del Citoesqueleto/metabolismo , Neuronas/efectos de los fármacos , Seudópodos/fisiología , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Axones/metabolismo , Axones/fisiología , Línea Celular , Células Cultivadas , Proteínas del Citoesqueleto/genética , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/citología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neuronas/citología , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Seudópodos/efectos de los fármacos , Seudópodos/metabolismo , Interferencia de ARN , Ratas , Transfección
6.
Exp Cell Res ; 317(6): 757-69, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21281626

RESUMEN

Stable expression of the tyrosine kinase receptor ErbB4 confers increased migratory behavior to the neuronal progenitor cell line ST14A, in response to neuregulin 1 (NRG1) stimulation. We used gene expression profiling analysis to identify transcriptional changes associated with higher migratory activity caused by the activation of a specific ErbB4 isoform, and found constitutive up-regulation of the epidermal growth factor receptor pathway substrate 8 (Eps8), a multimodular regulator of actin dynamics. We confirmed the increase of Eps8, both at the mRNA and at the protein level, in stable clones expressing two different ErbB4 isoforms, both characterized by high migratory activity. Using Transwell assays and experimental manipulation of Eps8 expression level, we demonstrated that Eps8 synergizes with ErbB4 to increase both basal and ligand induced cell migration, whereas siRNA mediated Eps8 silencing strongly impairs cell motility and NRG1 induced actin cytoskeleton remodeling. By transient knockdown of Eps8 through in vivo siRNA electroporation, followed by explant primary cultures, we demonstrated that Eps8 down-regulation affects migration of normal neuronal precursors. In conclusion, our data demonstrate that Eps8 is a key regulator of motility of neuronal progenitor cells expressing ErbB4, both in basal conditions and in response to external motogenic cues.


Asunto(s)
Receptores ErbB/metabolismo , Células-Madre Neurales , Neurregulina-1/metabolismo , Proteínas/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Western Blotting , Línea Celular Transformada , Movimiento Celular/fisiología , Ratones , Análisis por Micromatrices , Células-Madre Neurales/fisiología , Isoformas de Proteínas/fisiología , Ratas , Receptor ErbB-4 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Nature ; 426(6966): 579-84, 2003 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-14654845

RESUMEN

The assembly of 80S ribosomes requires joining of the 40S and 60S subunits, which is triggered by the formation of an initiation complex on the 40S subunit. This event is rate-limiting for translation, and depends on external stimuli and the status of the cell. Here we show that 60S subunits are activated by release of eIF6 (also termed p27BBP). In the cytoplasm, eIF6 is bound to free 60S but not to 80S. Furthermore, eIF6 interacts in the cytoplasm with RACK1, a receptor for activated protein kinase C (PKC). RACK1 is a major component of translating ribosomes, which harbour significant amounts of PKC. Loading 60S subunits with eIF6 caused a dose-dependent translational block and impairment of 80S formation, which were reversed by expression of RACK1 and stimulation of PKC in vivo and in vitro. PKC stimulation led to eIF6 phosphorylation, and mutation of a serine residue in the carboxy terminus of eIF6 impaired RACK1/PKC-mediated translational rescue. We propose that eIF6 release regulates subunit joining, and that RACK1 provides a physical and functional link between PKC signalling and ribosome activation.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Péptidos/metabolismo , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Ribosomas/química , Ribosomas/metabolismo , Animales , Proteínas Portadoras/genética , Línea Celular , Citoplasma/metabolismo , Factores Eucarióticos de Iniciación , Técnica del Anticuerpo Fluorescente , Humanos , Proteínas de Filamentos Intermediarios/genética , Fosforilación , Unión Proteica , Biosíntesis de Proteínas , Proteína Quinasa C/metabolismo , Receptores de Cinasa C Activada
8.
Life Sci Alliance ; 2(1)2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30692166

RESUMEN

EPS15 and its homologous EPS15L1 are endocytic accessory proteins. Studies in mammalian cell lines suggested that EPS15 and EPS15L1 regulate endocytosis in a redundant manner. However, at the organismal level, it is not known to which extent the functions of the two proteins overlap. Here, by exploiting various constitutive and conditional null mice, we report redundant and nonredundant functions of the two proteins. EPS15L1 displays a unique nonredundant role in the nervous system, whereas both proteins are fundamental during embryo development as shown by the embryonic lethality of -Eps15/Eps15L1-double KO mice. At the cellular level, the major process redundantly regulated by EPS15 and EPS15L1 is the endocytosis of the transferrin receptor, a pathway that sustains the development of red blood cells and controls iron homeostasis. Consequently, hematopoietic-specific conditional Eps15/Eps15L1-double KO mice display traits of microcytic hypochromic anemia, due to a cell-autonomous defect in iron internalization.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Endocitosis/fisiología , Anemia Hipocrómica/genética , Animales , Escala de Evaluación de la Conducta , Desarrollo Embrionario/fisiología , Eritrocitos/metabolismo , Fibroblastos/metabolismo , Técnicas de Inactivación de Genes , Genes Letales/fisiología , Hipocampo/citología , Hierro/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Receptores de Transferrina/metabolismo , Homología de Secuencia de Aminoácido , Homología Estructural de Proteína , Sinapsis/metabolismo
9.
Mol Biol Cell ; 15(1): 91-8, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14565974

RESUMEN

Sos-1, a guanine nucleotide exchange factor (GEF), eps8 and Abi1, two signaling proteins, and the lipid kinase phosphoinositide 3-kinase (PI3-K), assemble in a multimolecular complex required for Rac activation leading to actin cytoskeletal remodeling. Consistently, eps8 -/- fibroblasts fail to form membrane ruffles in response to growth factor stimulation. Surprisingly, eps8 null mice are healthy, fertile, and display no overt phenotype, suggesting the existence of functional redundancy within this pathway. Here, we describe the identification and characterization of a family of eps8-related proteins, comprising three novel gene products, named eps8L1, eps8L2, and eps8L3. Eps8Ls display collinear topology and 27-42% identity to eps8. Similarly to eps8, eps8Ls interact with Abi1 and Sos-1; however, only eps8L1 and eps8L2 activate the Rac-GEF activity of Sos-1, and bind to actin in vivo. Consistently, eps8L1 and eps8L2, but not eps8L3, localize to PDGF-induced, F-actin-rich ruffles and restore receptor tyrosine kinase (RTK)-mediated actin remodeling when expressed in eps8 -/- fibroblasts. Thus, the eps8Ls define a novel family of proteins responsible for functional redundancy in the RTK-activated signaling pathway leading to actin remodeling. Finally, the patterns of expression of eps8 and eps8L2 in mice are remarkably overlapping, thus providing a likely explanation for the lack of overt phenotype in eps8 null mice.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas del Citoesqueleto/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas/metabolismo , Proteína SOS1/metabolismo , Actinas/metabolismo , Animales , Northern Blotting , Extensiones de la Superficie Celular/metabolismo , Células Cultivadas , Clonación Molecular , Citoesqueleto/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Hibridación in Situ , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Unión Proteica/fisiología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Proteínas/genética , Proteínas Proto-Oncogénicas c-akt , Homología de Secuencia , Transducción de Señal , Distribución Tisular
10.
Nat Commun ; 6: 8261, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26383020

RESUMEN

Insulin regulates glycaemia, lipogenesis and increases mRNA translation. Cells with reduced eukaryotic initiation factor 6 (eIF6) do not increase translation in response to insulin. The role of insulin-regulated translation is unknown. Here we show that reduction of insulin-regulated translation in mice heterozygous for eIF6 results in normal glycaemia, but less blood cholesterol and triglycerides. eIF6 controls fatty acid synthesis and glycolysis in a cell autonomous fashion. eIF6 acts by exerting translational control of adipogenic transcription factors like C/EBPß, C/EBPδ and ATF4 that have G/C rich or uORF sequences in their 5' UTR. The outcome of the translational activation by eIF6 is a reshaping of gene expression with increased levels of lipogenic and glycolytic enzymes. Finally, eIF6 levels modulate histone acetylation and amounts of rate-limiting fatty acid synthase (Fasn) mRNA. Since obesity, type 2 diabetes, and cancer require a Fasn-driven lipogenic state, we propose that eIF6 could be a therapeutic target for these diseases.


Asunto(s)
Resistencia a la Insulina/genética , Metabolismo de los Lípidos/genética , Factores de Iniciación de Péptidos/genética , Biosíntesis de Proteínas/genética , ARN Mensajero/metabolismo , Transcripción Genética/genética , Células 3T3 , Acetilación , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Adipocitos/metabolismo , Adipogénesis/genética , Animales , Western Blotting , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteína delta de Unión al Potenciador CCAAT/genética , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Electroforesis en Gel de Poliacrilamida , Ácido Graso Sintasas/genética , Ácido Graso Sintasas/metabolismo , Ácidos Grasos , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Glucógeno/metabolismo , Glucólisis/genética , Células HEK293 , Hepatocitos/metabolismo , Código de Histonas , Humanos , Ácido Láctico/metabolismo , Lipogénesis/genética , Hígado/diagnóstico por imagen , Hígado/metabolismo , Células Madre Mesenquimatosas , Ratones , Oxidación-Reducción , Factores de Iniciación de Péptidos/metabolismo , Radiografía , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Neuroreport ; 13(9): 1189-93, 2002 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-12151767

RESUMEN

Truncated trkB.T1 is a splice variant of the neurotrophin receptor trkB. In spite of its abundance, and ability to bind and internalize BDNF, it is not clear whether it can transmit BDNF signaling. We tested this hypothesis by searching for proteins binding the evolutionarily conserved cyto-domain of trkB.T1, and by studying BDNF-induced changes of gene expression through DNA microarrays. Cells bearing trkB.T1 receptors presented morphological changes. However, no cytoplasmic interactors of trkB.T1 were found. In addition, BDNF-dependent modulation of gene expression was detected in cells bearing trkB.TK but not trkB.T1 receptors. These results suggest that the main function of trkB.T1 is to regulate local availability of neurotrophins and that it is unable to sense changes in BDNF availability.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/farmacología , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Sistema Nervioso/crecimiento & desarrollo , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Receptor trkB/efectos de los fármacos , Células 3T3 , Empalme Alternativo/efectos de los fármacos , Empalme Alternativo/fisiología , Animales , Tamaño de la Célula/efectos de los fármacos , Tamaño de la Célula/fisiología , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Genes/efectos de los fármacos , Genes/fisiología , Ratones , Sistema Nervioso/efectos de los fármacos , Sistema Nervioso/metabolismo , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Isoformas de Proteínas/efectos de los fármacos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína/fisiología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Receptor trkB/genética , Receptor trkB/metabolismo , Receptores de Superficie Celular/efectos de los fármacos , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Transcripción Genética/efectos de los fármacos , Transcripción Genética/fisiología , Transducción Genética
12.
Curr Biol ; 23(23): 2355-64, 2013 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-24239119

RESUMEN

BACKGROUND: Integrin-mediated adhesion of cells to the extracellular matrix (ECM) relies on the dynamic formation of focal adhesions (FAs), which are biochemical and mechanosensitive platforms composed of a large variety of cytosolic and transmembrane proteins. During migration, there is a constant turnover of ECM contacts that initially form as nascent adhesions at the leading edge, mature into FAs as actomyosin tension builds up, and are then disassembled at the cell rear, thus allowing for cell detachment. Although the mechanisms of FA assembly have largely been defined, the molecular circuitry that regulates their disassembly still remains elusive. RESULTS: Here, we show that RN-tre, a GTPase-activating protein (GAP) for Rabs including Rab5 and Rab43, is a novel regulator of FA dynamics and cell migration. RN-tre localizes to FAs and to a pool of Rab5-positive vesicles mainly associated with FAs undergoing rapid remodeling. We found that RN-tre inhibits endocytosis of ß1, but not ß3, integrins and delays the turnover of FAs, ultimately impairing ß1-dependent, but not ß3-dependent, chemotactic cell migration. All of these effects are mediated by its GAP activity and rely on Rab5. CONCLUSIONS: Our findings identify RN-tre as the Rab5-GAP that spatiotemporally controls FA remodeling during chemotactic cell migration.


Asunto(s)
Movimiento Celular/genética , Endocitosis/genética , Proteínas Activadoras de GTPasa/metabolismo , Proteínas de Unión al GTP rab5/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Adhesión Celular/fisiología , Células Cultivadas , Doxiciclina/farmacología , Endocitosis/fisiología , Adhesiones Focales/metabolismo , Proteínas Activadoras de GTPasa/genética , Integrina beta1/metabolismo , Integrina beta3/metabolismo , Ratones , Ratones Noqueados , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal/fisiología , Proteínas de Unión al GTP rab/biosíntesis , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab5/biosíntesis
13.
PLoS One ; 7(11): e50818, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23226392

RESUMEN

Eps15 is an endocytic adaptor protein involved in clathrin and non-clathrin mediated endocytosis. In Caenorhabditis elegans and Drosophila melanogaster lack of Eps15 leads to defects in synaptic vesicle recycling and synapse formation. We generated Eps15-KO mice to investigate its function in mammals. Eps15-KO mice are born at the expected Mendelian ratio and are fertile. Using a large-scale phenotype screen covering more than 300 parameters correlated to human disease, we found that Eps15-KO mice did not show any sign of disease or neural deficits. Instead, altered blood parameters pointed to an immunological defect. By competitive bone marrow transplantation we demonstrated that Eps15-KO hematopoietic precursor cells were more efficient than the WT counterparts in repopulating B220⁺ bone marrow cells, CD19⁻ thymocytes and splenic marginal zone (MZ) B cells. Eps15-KO mice showed a 2-fold increase in MZ B cell numbers when compared with controls. Using reverse bone marrow transplantation, we found that Eps15 regulates MZ B cell numbers in a cell autonomous manner. FACS analysis showed that although MZ B cells were increased in Eps15-KO mice, transitional and pre-MZ B cell numbers were unaffected. The increase in MZ B cell numbers in Eps15 KO mice was not dependent on altered BCR signaling or Notch activity. In conclusion, in mammals, the endocytic adaptor protein Eps15 is a regulator of B-cell lymphopoiesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Linfocitos B/citología , Recuento de Células , Endocitosis , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Trasplante de Médula Ósea , Femenino , Técnicas de Inactivación de Genes , Masculino , Ratones , Receptores Notch/metabolismo , Timo/citología , Timo/inmunología
14.
PLoS One ; 5(3): e9468, 2010 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-20209148

RESUMEN

BACKGROUND: In a variety of organisms, including mammals, caloric restriction improves metabolic status and lowers the incidence of chronic-degenerative diseases, ultimately leading to increased lifespan. METHODOLOGY/PRINCIPAL FINDINGS: Here we show that knockout mice for Eps8, a regulator of actin dynamics, display reduced body weight, partial resistance to age- or diet-induced obesity, and overall improved metabolic status. Alteration in the liver gene expression profile, in behavior and metabolism point to a calorie restriction-like phenotype in Eps8 knockout mice. Additionally, and consistent with a calorie restricted metabolism, Eps8 knockout mice show increased lifespan. The metabolic alterations in Eps8 knockout mice correlated with a significant reduction in intestinal fat absorption presumably caused by a 25% reduction in intestinal microvilli length. CONCLUSIONS/SIGNIFICANCE: Our findings implicate actin dynamics as a novel variable in the determination of longevity. Additionally, our observations suggest that subtle differences in energy balance can, over time, significantly affect bodyweight and metabolic status in mice.


Asunto(s)
Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas del Citoesqueleto/fisiología , Mucosa Intestinal/metabolismo , Células 3T3-L1 , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Peso Corporal , Células CACO-2 , Restricción Calórica , Proteínas del Citoesqueleto/genética , Metabolismo Energético , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microvellosidades/metabolismo
15.
Cell ; 127(1): 213-26, 2006 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-17018287

RESUMEN

Dynamic modulation of the actin cytoskeleton is critical for synaptic plasticity, abnormalities of which are thought to contribute to mental illness and addiction. Here we report that mice lacking Eps8, a regulator of actin dynamics, are resistant to some acute intoxicating effects of ethanol and show increased ethanol consumption. In the brain, the N-methyl-D-aspartate (NMDA) receptor is a major target of ethanol. We show that Eps8 is localized to postsynaptic structures and is part of the NMDA receptor complex. Moreover, in Eps8 null mice, NMDA receptor currents and their sensitivity to inhibition by ethanol are abnormal. In addition, Eps8 null neurons are resistant to the actin-remodeling activities of NMDA and ethanol. We propose that proper regulation of the actin cytoskeleton is a key determinant of cellular and behavioral responses to ethanol.


Asunto(s)
Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Conducta Animal/efectos de los fármacos , Depresores del Sistema Nervioso Central/farmacología , Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Etanol/farmacología , Proteínas Adaptadoras Transductoras de Señales/genética , Consumo de Bebidas Alcohólicas , Animales , Conducta Animal/fisiología , Células Cultivadas , Cerebelo/citología , Proteínas del Citoesqueleto/genética , Citoesqueleto/efectos de los fármacos , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal/fisiología
16.
Am J Physiol Regul Integr Comp Physiol ; 282(4): R1219-26, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11893628

RESUMEN

During avian embryonic development, terminal erythroid differentiation occurs in the circulation. Some of the key events, such as the induction of erythroid 2,3-bisphosphoglycerate (2,3-BPG), carbonic anhydrase (CAII), and pyrimidine 5'-nucleotidase (P5N) synthesis are oxygen dependent (Baumann R, Haller EA, Schöning U, and Weber M, Dev Biol 116: 548-551, 1986; Dragon S and Baumann R, Am J Physiol Regulatory Integrative Comp Physiol 280: R870-R878, 2001; Dragon S, Carey C, Martin K, and Baumann R, J Exp Biol 202: 2787-2795, 1999; Dragon S, Glombitza S, Götz R, and Baumann R, Am J Physiol Regulatory Integrative Comp Physiol 271: R982-R989, 1996; Dragon S, Hille R, Götz R, and Baumann R, Blood 91: 3052-3058, 1998; Million D, Zillner P, and Baumann R, Am J Physiol Regulatory Integrative Comp Physiol 261: R1188-R1196, 1991) in an indirect way: hypoxia stimulates the release of norepinephrine (NE)/adenosine into the circulation (Dragon et al., J Exp Biol 202: 2787-2795, 1999; Dragon et al., Am J Physiol Regulatory Integrative Comp Physiol 271: R982-R989, 1996). This leads via erythroid beta-adrenergic/adenosine A(2) receptor activation to a cAMP signal inducing several proteins in a transcription-dependent manner (Dragon et al., Am J Physiol Regulatory Integrative Comp Physiol 271: R982-R989, 1996; Dragon et al., Blood 91: 3052-3058, 1998; Glombitza S, Dragon S, Berghammer M, Pannermayr M, and Baumann R, Am J Physiol Regulatory Integrative Comp Physiol 271: R973-R981, 1996). To understand how the cAMP-dependent processes are initiated, we screened an erythroid cDNA library for cAMP-regulated genes. We detected three genes that were strongly upregulated (>5-fold) by cAMP in definitive and primitive red blood cells. They are homologous to the mammalian Tob, Ifr1, and Fos proteins. In addition, the genes are induced in the intact embryo during short-term hypoxia. Because the genes are regulators of proliferation and differentiation in other cell types, we suggest that cAMP might promote general differentiating processes in erythroid cells, thereby allowing adaptive modulation of the latest steps of erythroid differentiation during developmental hypoxia.


Asunto(s)
Proteínas Portadoras/genética , AMP Cíclico/metabolismo , Eritrocitos/fisiología , Hipoxia/metabolismo , Proteínas Inmediatas-Precoces/genética , Proteínas de la Membrana/genética , Proteínas Proto-Oncogénicas c-fos/genética , Animales , Embrión de Pollo , Eritropoyesis/fisiología , Expresión Génica/fisiología , Sistemas de Lectura Abierta/genética , ARN Mensajero/análisis , Receptores Adrenérgicos beta/metabolismo , Receptores Purinérgicos P1/metabolismo
17.
Biochem Biophys Res Commun ; 295(2): 295-9, 2002 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-12150946

RESUMEN

p27(BBP)/eIF6 is an evolutionarily conserved protein necessary for ribosome biogenesis which was cloned in mammals for its ability to bind the cytodomain of beta 4 integrin. In cultured cells, a conspicuous fraction of p27(BBP)/eIF6 is associated with the intermediate filaments/nuclear matrix (IF/NM) cytoskeleton. The mechanism of this association is not known. Here we show that in epidermis p27(BBP)/eIF6 is naturally associated with IF/NM. To analyze the intrinsic capability of p27(BBP)/eIF6 to generate cytoskeletal networks, the properties of the pure, recombinant, untagged protein were studied. Recombinant p27(BBP)/eIF6 binds beta 4 integrin. Upon dialysis against IF buffer, p27(BBP)/eIF6 forms polymers which, strikingly, have a morphology identical to NM filaments. Cross-linking experiments suggested that polymerization is favored by the formation of disulphide bridges. These data suggest that p27(BBP)/eIF6 is associated with the cytoskeleton, and contributes to formation of NM filaments. These findings help to settle the controversy on nuclear matrix.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Matriz Nuclear/metabolismo , Factores de Iniciación de Péptidos , Animales , Western Blotting , Ratones , Matriz Nuclear/ultraestructura , Proteínas Recombinantes/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA