Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
FEBS Lett ; 594(1): 31-42, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31423582

RESUMEN

Late endosomal/lysosomal adaptor and MAPK and mTOR activator (LAMTOR/Ragulator) is a scaffold protein complex that anchors and regulates multiprotein signaling units on late endosomes/lysosomes. To identify LAMTOR-modulated endolysosomal proteins, primary macrophages were derived from bone marrow of conditional knockout mice carrying a specific deletion of LAMTOR2 in the monocyte/macrophage cell lineage. Affymetrix-based transcriptomic analysis and quantitative iTRAQ-based organelle proteomic analysis of endosomes derived from macrophages were performed. Further analyses showed that LAMTOR could be a novel regulator of foam cell differentiation. The lipid droplet formation phenotype observed in macrophages was additionally confirmed in MEFs, where lipidomic analysis identified cholesterol esters as specifically downregulated in LAMTOR2 knockout cells. The data obtained indicate a function of LAMTOR2 in lipid metabolism.


Asunto(s)
Diferenciación Celular , Células Espumosas/metabolismo , Metabolismo de los Lípidos , Macrófagos/metabolismo , Proteínas/metabolismo , Animales , Células Cultivadas , Ésteres del Colesterol/metabolismo , Células Espumosas/citología , Gotas Lipídicas/metabolismo , Macrófagos/citología , Ratones , Proteínas/genética , Transcriptoma
2.
PLoS One ; 13(5): e0197674, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29787578

RESUMEN

Transgenic mouse models are indispensable tools to mimic human diseases and analyze the effectiveness of related new drugs. For a long time amyotrophic lateral sclerosis (ALS) research depended on only a few mouse models that exhibit a very strong and early phenotype, e.g. SOD1 mice, resulting in a short treatment time window. By now, several models are available that need to be characterized to highlight characteristics of each model. Here we further characterized the mThy1-hTDP-43 transgenic mouse model TAR6/6 that overexpresses wild type human TARDBP, also called TDP-43, under control of the neuronal Thy-1 promoter presented by Wils and colleagues, 2010, by using biochemical, histological and behavioral readouts. Our results show that TAR6/6 mice exhibit a strong TDP-43 expression in the hippocampus, spinal cord, hypothalamus and medulla oblongata. Apart from prominent protein expression in the nucleus, TDP-43 protein was found at lower levels in the cytosol of transgenic mice. Additionally, we detected insoluble TDP-43 in the cortex, motoneuron loss, and increased neuroinflammation in the central nervous system of TAR6/6 animals. Behavioral analyses revealed early motor deficits in the clasping- and wire suspension test as well as decreased anxiety in the elevated plus maze. Further motor tests showed differences at later time points compared to non-transgenic littermates, thus allowing the observation of onset and severity of such deficits. Together, TAR6/6 mice are a valuable tool to test new ALS/FTLD drugs that target TDP-43 expression and insolubility, neuroinflammation, motoneuron loss or other TDP-43 related downstream signaling pathways since these mice exhibit a later pathology as previously used ALS/FTLD mouse models.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Degeneración Lobar Frontotemporal/genética , Monoéster Fosfórico Hidrolasas/genética , Regulación hacia Arriba , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Núcleo Celular/metabolismo , Citosol/metabolismo , Modelos Animales de Enfermedad , Degeneración Lobar Frontotemporal/metabolismo , Degeneración Lobar Frontotemporal/fisiopatología , Hipocampo/metabolismo , Humanos , Hipotálamo/metabolismo , Bulbo Raquídeo/metabolismo , Ratones , Ratones Transgénicos , Neuronas Motoras/fisiología , Regiones Promotoras Genéticas , Médula Espinal/metabolismo
3.
Neurodegener Dis ; 15(4): 243-57, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26111745

RESUMEN

BACKGROUND: ß-Synuclein (ß-Syn) is a member of the highly homologous synuclein protein family. The most prominent family member, α-synuclein (α-Syn), abnormally accumulates in so-called Lewy bodies, one of the major pathological hallmarks of α-synucleinopathies. Notably, parts of the peptide backbone, called the nonamyloid component, are also found in amyloid plaques. However, ß-Syn seems to have beneficial effects by reducing α-Syn aggregation, and amyloid antiaggregatory activity has been described. OBJECTIVE: The aim of the study was to analyze if wild-type ß-Syn can counteract functional and pathological changes in a murine Alzheimer model over different time periods. METHODS: At the onset of pathology, lentiviral particles expressing human ß-Syn were injected into the hippocampus of transgenic mice overexpressing human amyloid precursor protein with Swedish and London mutations (APPSL). An empty vector served as the control. Behavioral analyses were performed 1, 3 and 6 months after injection followed by biochemical and histological examinations of brain samples. RESULTS: ß-Syn expression was locally concentrated and rather modest, but nevertheless changed its effect on APP expression and plaque load in a time- and concentration-dependent manner. Interestingly, the phosphorylation of glycogen synthase kinase 3 beta was enhanced in APPSL mice expressing human ß-Syn, but an inverse trend was observed in wild-type animals. CONCLUSION: The initially reported beneficial effects of ß-Syn could be partially reproduced, but locally elevated levels of ß-Syn might also cause neurodegeneration. To enlighten the controversial pathological mechanism of ß-Syn, further examinations considering the relationship between concentration and exposure time of ß-Syn are needed.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Hipocampo/metabolismo , Sinucleína beta/metabolismo , Animales , Ansiedad/prevención & control , Línea Celular Tumoral , Modelos Animales de Enfermedad , Encefalitis/metabolismo , Vectores Genéticos , Humanos , Lentivirus , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , alfa-Sinucleína/metabolismo , Sinucleína beta/administración & dosificación , Sinucleína beta/genética
4.
Molecules ; 20(3): 4492-515, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25764491

RESUMEN

We describe the multigram synthesis and in vivo efficacy studies of a donepezil‒huprine hybrid that has been found to display a promising in vitro multitarget profile of interest for the treatment of Alzheimer's disease (AD). Its synthesis features as the key step a novel multigram preparative chromatographic resolution of intermediate racemic huprine Y by chiral HPLC. Administration of this compound to transgenic CL4176 and CL2006 Caenorhabditis elegans strains expressing human Aß42, here used as simplified animal models of AD, led to a significant protection from the toxicity induced by Aß42. However, this protective effect was not accompanied, in CL2006 worms, by a reduction of amyloid deposits. Oral administration for 3 months to transgenic APPSL mice, a well-established animal model of AD, improved short-term memory, but did not alter brain levels of Aß peptides nor cortical and hippocampal amyloid plaque load. Despite the clear protective and cognitive effects of AVCRI104P4, the lack of Aß lowering effect in vivo might be related to its lower in vitro potency toward Aß aggregation and formation as compared with its higher anticholinesterase activities. Further lead optimization in this series should thus focus on improving the anti-amyloid/anticholinesterase activity ratio.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Aminoquinolinas/administración & dosificación , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Compuestos Heterocíclicos de 4 o más Anillos/administración & dosificación , Indanos/administración & dosificación , Piperidinas/administración & dosificación , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Aminoquinolinas/química , Aminoquinolinas/uso terapéutico , Precursor de Proteína beta-Amiloide/genética , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans , Modelos Animales de Enfermedad , Donepezilo , Células Hep G2 , Compuestos Heterocíclicos de 4 o más Anillos/química , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Humanos , Indanos/química , Indanos/uso terapéutico , Ratones , Estructura Molecular , Piperidinas/química , Piperidinas/uso terapéutico
5.
J Biol Chem ; 288(25): 18228-42, 2013 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23653355

RESUMEN

LAMTOR3 (MP1) and LAMTOR2 (p14) form a heterodimer as part of the larger Ragulator complex that is required for MAPK and mTOR1 signaling from late endosomes/lysosomes. Here, we show that loss of LAMTOR2 (p14) results in an unstable cytosolic monomeric pool of LAMTOR3 (MP1). Monomeric cytoplasmic LAMTOR3 is rapidly degraded in a proteasome-dependent but lysosome-independent manner. Mutational analyses indicated that the turnover of the protein is dependent on ubiquitination of several lysine residues. Similarly, other Ragulator subunits, LAMTOR1 (p18), LAMTOR4 (c7orf59), and LAMTOR5 (HBXIP), are degraded as well upon the loss of LAMTOR2. Thus the assembly of the Ragulator complex is monitored by cellular quality control systems, most likely to prevent aberrant signaling at the convergence of mTOR and MAPK caused by a defective Ragulator complex.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Multimerización de Proteína , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Western Blotting , Células Cultivadas , Embrión de Mamíferos/citología , Endosomas/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Células HEK293 , Células HeLa , Humanos , Ratones , Ratones Noqueados , Microscopía Confocal , Estabilidad Proteica , Proteínas/genética , Proteínas/metabolismo , Proteolisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ubiquitina/metabolismo
6.
J Hepatol ; 57(2): 337-43, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22521359

RESUMEN

BACKGROUND & AIMS: The mannose 6-phosphate/insulin-like growth factor II receptor (M6P/IGF2R), a multifunctional protein, plays a central role in intracellular targeting of lysosomal enzymes and control of insulin-like growth factor II (IGF-II) bioactivity. Importantly, the gene encoding this receptor is frequently inactivated in a wide range of malignant tumors including hepatocellular carcinomas. Thus, M6P/IGF2R is considered a putative liver tumor suppressor. The aim of this study was to establish the impact of the receptor on the invasive properties of liver cells. METHODS: Reconstitution experiments were performed by expression of wild type and mutant M6P/IGF2R in receptor-deficient FRL14 fetal rat liver cells. RNA interference was used to induce M6P/IGF2R downregulation in receptor-positive MIM-1-4 mouse hepatocytes. RESULTS: We show that the M6P/IGF2R status exerts a strong impact on the invasiveness of tumorigenic rodent liver cells. M6P/IGF2R-deficient fetal rat liver cells hypersecrete lysosomal cathepsins and penetrate extracellular matrix barriers in a cathepsin-dependent manner. Forced expression of M6P/IGF2R restores intracellular transport of cathepsins to lysosomes and concomitantly reduces the tumorigenicity and invasive potential of these cells. Conversely, M6P/IGF2R knock-down in receptor-positive mouse hepatocytes causes increased cathepsin secretion as well as enhanced cell motility and invasiveness. We also demonstrate that functional M6P-binding sites are important for the anti-invasive properties of M6P/IGF2R, whereas the capacity to bind IGF-II is dispensable for the anti-invasive activity of the receptor in liver cells. CONCLUSIONS: M6P/IGF2R restricts liver cell invasion by preventing the pericellular action of M6P-modified proteins.


Asunto(s)
Hepatocitos/patología , Neoplasias Hepáticas/patología , Manosafosfatos/metabolismo , Receptor IGF Tipo 2/fisiología , Animales , Línea Celular , Movimiento Celular , Proliferación Celular , Humanos , Leucina/análogos & derivados , Leucina/farmacología , Lisosomas/enzimología , Ratones , Invasividad Neoplásica , Unión Proteica , Ratas
7.
J Cell Sci ; 125(Pt 11): 2698-708, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22427693

RESUMEN

The outcome of an infection depends on the balance between host resistance and bacterial virulence. Here, we show that the late endosomal adaptor p14 (also known as LAMTOR2) is one of the components for cellular host defense against the intracellular pathogen Salmonella enterica serovar Typhimurium. During Salmonella infection, the complex of p14 and MP1 is required for the accurately timed transport of Salmonella through the endolysosomal system. Loss of p14 opens a time window that allows Salmonella to populate a replication niche, in which early and late antimicrobial effector systems, comprising NADPH phagocytic oxidase and inducible nitric oxide synthase, respectively, are inappropriately activated. Thus, p14 supports the accurate transport of Salmonella through the endolysosomal system, thereby limiting bacterial replication in both, professional phagocytes and in non-phagocytic cells in vitro, and helps mice to successfully battle Salmonella infection in vivo.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Interacciones Huésped-Patógeno/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Proteínas/metabolismo , Salmonelosis Animal/inmunología , Salmonelosis Animal/microbiología , Salmonella typhimurium/fisiología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Animales , Compartimento Celular , Susceptibilidad a Enfermedades , Endosomas/enzimología , Endosomas/ultraestructura , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas de Membrana de los Lisosomas/metabolismo , Macrófagos/metabolismo , Macrófagos/ultraestructura , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , NADPH Oxidasas/metabolismo , Fagocitosis , Transporte de Proteínas , Salmonella typhimurium/crecimiento & desarrollo , Vacuolas/metabolismo , Vacuolas/microbiología , Vacuolas/ultraestructura
8.
J Mol Neurosci ; 47(1): 192-203, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22351109

RESUMEN

The role of hyperphosphorylation of the microtubule-associated protein tau in the pathological processes of several neurodegenerative diseases is becoming better understood. Consequently, development of new compounds capable of preventing tau hyperphosphorylation is an increasingly hot topic. For this reason, dependable in vitro and in vivo models that reflect tau hyperphosphorylation in human diseases are needed. In this study, we generated and validated an in vitro model appropriate to test potential curative and preventive compound effects on tau phosphorylation. For this purpose, a stably transfected SH-SY5Y cell line was constructed over-expressing mutant human tau441 (SH-SY5Y-TMHT441). Analyses of expression levels and tau phosphorylation status in untreated cells confirmed relevance to human diseases. Subsequently, the effect of different established kinase inhibitors on tau phosphorylation (e.g., residues Thr231, Thr181, and Ser396) was examined. It was shown with several methods including immunosorbent assays and mass spectrometry that the phosphorylation pattern of tau in SH-SY5Y-TMHT441 cells can be reliably modulated by these compounds, specifically targeting JNK, GSK-3, CDK1/5, and CK1. These four protein kinases are known to be involved in in vivo tau phosphorylation and are therefore authentic indicators for the suitability of this new cell culture model for tauopathies.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Pruebas Genéticas/métodos , Farmacogenética/métodos , Proteínas tau/genética , Enfermedad de Alzheimer/patología , Animales , Línea Celular Tumoral , Diseño de Fármacos , Humanos , Ratones , Ratones Transgénicos , Mutagénesis/fisiología , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/genética , Neuroblastoma/patología , Fosforilación/efectos de los fármacos , Fosforilación/genética , Tauopatías/tratamiento farmacológico , Tauopatías/genética , Tauopatías/metabolismo , Transfección/métodos
9.
J Exp Med ; 208(13): 2675-89, 2011 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-22143885

RESUMEN

Despite intense investigation of intrinsic and extrinsic factors that regulate pluripotency, the process of initial fate commitment of embryonic stem (ES) cells is still poorly understood. We used a genome-wide short hairpin RNA screen in mouse ES cells to identify genes that are essential for initiation of differentiation. Knockdown of the scaffolding protein Mek binding protein 1 (Mp1, also known as Lamtor3 or Map2k1ip1) stimulated self-renewal of ES cells, blocked differentiation, and promoted proliferation. Fibroblast growth factor 4 (FGF4) signaling is required for initial fate commitment of ES cells. Knockdown of Mp1 inhibited FGF4-induced differentiation but did not alter FGF4-driven proliferation. This uncoupling of differentiation and proliferation was also observed when oncogenic Ras isoforms were overexpressed in ES cells. Knockdown of Mp1 redirected FGF4 signaling from differentiation toward pluripotency and up-regulated the pluripotency-related genes Esrrb, Rex1, Tcl1, and Sox2. We also found that human germ cell tumors (GCTs) express low amounts of Mp1 in the invasive embryonic carcinoma and seminoma histologies and higher amounts of Mp1 in the noninvasive carcinoma in situ precursor and differentiated components. Knockdown of Mp1 in invasive GCT cells resulted in resistance to differentiation, thereby showing a functional role for Mp1 both in normal differentiation of ES cells and in germ cell cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Diferenciación Celular/fisiología , Células Madre Embrionarias/metabolismo , Células Madre Pluripotentes/metabolismo , Interferencia de ARN , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Carcinoma in Situ/genética , Carcinoma in Situ/metabolismo , Línea Celular , Proliferación Celular , Células Madre Embrionarias/citología , Factor 4 de Crecimiento de Fibroblastos/genética , Factor 4 de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica/fisiología , Estudio de Asociación del Genoma Completo/métodos , Humanos , Ratones , Ratones Noqueados , Neoplasias de Células Germinales y Embrionarias/genética , Neoplasias de Células Germinales y Embrionarias/metabolismo , Células Madre Pluripotentes/citología , Transducción de Señal/fisiología
10.
Immunity ; 34(1): 61-74, 2011 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-21256055

RESUMEN

Erythropoietin (EPO) is the principal cytokine regulating erythropoiesis through its receptor, EPOR. Interestingly, EPORs are also found on immune cells with incompletely understood functions. Here, we show that EPO inhibits the induction of proinflammatory genes including tumor necrosis factor (TNF)-α and inducible nitric oxide (NO) synthase in activated macrophages, which is mechanistically attributable to blockage of nuclear factor (NF)-κB p65 activation by EPO. Accordingly, in systemic Salmonella infection, treatment of mice with EPO results in reduced survival and impaired pathogen clearance because of diminished formation of anti-microbial effector molecules such as TNF-α and NO. However, neutralization of endogenous EPO or genetic ablation of Epor promotes Salmonella elimination. In contrast, in chemically induced colitis, EPO-EPOR interaction decreases the production of NF-κB-inducible immune mediators, thus limiting tissue damage and ameliorating disease severity. These immune-modulatory effects of EPO may be of therapeutic relevance in infectious and inflammatory diseases.


Asunto(s)
Colitis/inmunología , Eritropoyetina/administración & dosificación , Macrófagos Peritoneales/efectos de los fármacos , FN-kappa B/metabolismo , Receptores de Eritropoyetina/metabolismo , Infecciones por Salmonella/inmunología , Salmonella/inmunología , Animales , Línea Celular , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Sulfato de Dextran/administración & dosificación , Humanos , Mediadores de Inflamación/metabolismo , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/inmunología , Óxido Nítrico/metabolismo , Receptores de Eritropoyetina/genética , Salmonella/patogenicidad , Infecciones por Salmonella/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Ácido Trinitrobencenosulfónico/administración & dosificación , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
11.
Nat Med ; 13(1): 38-45, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17195838

RESUMEN

Lysosome-related organelles have versatile functions, including protein and lipid degradation, signal transduction and protein secretion. The molecular elucidation of rare congenital diseases affecting endosomal-lysosomal biogenesis has given insights into physiological functions of the innate and adaptive immune system. Here, we describe a previously unknown human primary immunodeficiency disorder and provide evidence that the endosomal adaptor protein p14, previously characterized as confining mitogen-activated protein kinase (MAPK) signaling to late endosomes, is crucial for the function of neutrophils, B cells, cytotoxic T cells and melanocytes. Combining genetic linkage studies and transcriptional profiling analysis, we identified a homozygous point mutation in the 3' untranslated region (UTR) of p14 (also known as MAPBPIP), resulting in decreased protein expression. In p14-deficient cells, the distribution of late endosomes was severely perturbed, suggesting a previously unknown role for p14 in endosomal biogenesis. These findings have implications for understanding endosomal membrane dynamics, compartmentalization of cell signal cascades, and their role in immunity.


Asunto(s)
Complejo 4 de Proteína Adaptadora/metabolismo , Endosomas/metabolismo , Síndromes de Inmunodeficiencia/metabolismo , Complejo 4 de Proteína Adaptadora/deficiencia , Complejo 4 de Proteína Adaptadora/genética , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Linfocitos B/ultraestructura , Secuencia de Bases , Endosomas/ultraestructura , Salud de la Familia , Femenino , Genotipo , Factor Estimulante de Colonias de Granulocitos/farmacología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunoglobulina D/análisis , Inmunoglobulina M/análisis , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/patología , Recuento de Leucocitos , Desequilibrio de Ligamiento , Luciferasas/genética , Luciferasas/metabolismo , Masculino , Melanocitos/metabolismo , Melanocitos/ultraestructura , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Neutrófilos/metabolismo , Neutrófilos/ultraestructura , Mutación Puntual , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/ultraestructura , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/análisis
12.
J Cell Biol ; 175(6): 861-8, 2006 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-17178906

RESUMEN

The extracellular signal-regulated kinase (ERK) cascade regulates proliferation, differentiation, and survival in multicellular organisms. Scaffold proteins regulate intracellular signaling by providing critical spatial and temporal specificity. The scaffold protein MEK1 (mitogen-activated protein kinase and ERK kinase 1) partner (MP1) is localized to late endosomes by the adaptor protein p14. Using conditional gene disruption of p14 in mice, we now demonstrate that the p14-MP1-MEK1 signaling complex regulates late endosomal traffic and cellular proliferation. This function its essential for early embryogenesis and during tissue homeostasis, as revealed by epidermis-specific deletion of p14. These findings show that endosomal p14-MP1-MEK1 signaling has a specific and essential function in vivo and, therefore, indicate that regulation of late endosomal traffic by extracellular signals is required to maintain tissue homeostasis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proliferación Celular , Endosomas/metabolismo , MAP Quinasa Quinasa 1/metabolismo , Proteínas/fisiología , Transducción de Señal , Animales , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Células Epidérmicas , Epidermis/metabolismo , Receptores ErbB/metabolismo , Integrasas , Ratones , Ratones Noqueados , Proteínas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...