Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(22)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34039707

RESUMEN

Specified intestinal epithelial cells reprogram and contribute to the regeneration and renewal of the epithelium upon injury. Mutations that deregulate such renewal processes may contribute to tumorigenesis. Using intestinal organoids, we show that concomitant activation of Notch signaling and ablation of p53 induce a highly proliferative and regenerative cell state, which is associated with increased levels of Yap and the histone methyltransferase Mll1. The induced signaling system orchestrates high proliferation, self-renewal, and niche-factor-independent growth, and elevates the trimethylation of histone 3 at lysine 4 (H3K4me3). We demonstrate that Yap and Mll1 are also elevated in patient-derived colorectal cancer (CRC) organoids and control growth and viability. Our data suggest that Notch activation and p53 ablation induce a signaling circuitry involving Yap and the epigenetic regulator Mll1, which locks cells in a proliferative and regenerative state that renders them susceptible for tumorigenesis.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , N-Metiltransferasa de Histona-Lisina/fisiología , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Receptores Notch/metabolismo , Transducción de Señal , Factores de Transcripción/fisiología , Proteína p53 Supresora de Tumor/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Humanos , Mutación , Organoides/metabolismo , Factores de Transcripción/metabolismo
2.
J Biol Chem ; 294(35): 12992-13005, 2019 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-31296571

RESUMEN

Although Merlin's function as a tumor suppressor and regulator of mitogenic signaling networks such as the Ras/rac, Akt, and Hippo pathways is well-documented, in mammals as well as in insects, its role during cell cycle progression remains unclear. In this study, using a combination of approaches, including FACS analysis, time-lapse imaging, immunofluorescence microscopy, and co-immunoprecipitation, we show that Ser-518 of Merlin is a substrate of the Aurora protein kinase A during mitosis and that its phosphorylation facilitates the phosphorylation of a newly discovered site, Thr-581. We found that the expression in HeLa cells of a Merlin variant that is phosphorylation-defective on both sites leads to a defect in centrosomes and mitotic spindles positioning during metaphase and delays the transition from metaphase to anaphase. We also show that the dual mitotic phosphorylation not only reduces Merlin binding to microtubules but also timely modulates ezrin interaction with the cytoskeleton. Finally, we identify several point mutants of Merlin associated with neurofibromatosis type 2 that display an aberrant phosphorylation profile along with defective α-tubulin-binding properties. Altogether, our findings of an Aurora A-mediated interaction of Merlin with α-tubulin and ezrin suggest a potential role for Merlin in cell cycle progression.


Asunto(s)
Aurora Quinasa A/metabolismo , Mitosis , Neurofibromina 2/metabolismo , Aurora Quinasa A/antagonistas & inhibidores , Benzazepinas/farmacología , Células HEK293 , Células HeLa , Humanos , Mitosis/efectos de los fármacos , Mutación , Neurofibromina 2/antagonistas & inhibidores , Neurofibromina 2/genética , Nocodazol/farmacología , Fosforilación/efectos de los fármacos
3.
J Neurosci ; 37(12): 3127-3137, 2017 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-28193690

RESUMEN

Differentiation and maturation of oligodendrocyte progenitor cells (OPCs) involve the assembly and disassembly of actin microfilaments. However, how actin dynamics are regulated during this process remains poorly understood. Leucine-rich repeat and Ig-like domain-containing Nogo receptor interacting protein 1 (LINGO-1) is a negative regulator of OPC differentiation. We discovered that anti-LINGO-1 antibody-promoted OPC differentiation was accompanied by upregulation of cytoplasmic gelsolin (cGSN), an abundant actin-severing protein involved in the depolymerization of actin filaments. Treating rat OPCs with cGSN siRNA reduced OPC differentiation, whereas overexpression of cGSN promoted OPC differentiation in vitro and remyelination in vivo Furthermore, coexpression of cGSN and LINGO-1 blocked the inhibitory effect of LINGO-1. Our study demonstrates that cGSN works downstream of LINGO-1 signaling pathway, which enhances actin dynamics and is essential for OPC morphogenesis and differentiation. This finding may lead to novel therapeutic approaches for the treatment of demyelinating diseases such as multiple sclerosis (MS).SIGNIFICANCE STATEMENT Myelin loss and subsequent axon degeneration contributes to a variety of neurological diseases, such as multiple sclerosis (MS). Understanding the regulation of myelination by oligodendrocytes is therefore critical for developing therapies for the treatment of MS. We previously demonstrated that leucine-rich repeat and Ig-like domain-containing Nogo receptor interacting protein 1 (LINGO-1) is a negative regulator of oligodendrocyte differentiation and that anti-LINGO-1 promotes remyelination in preclinical animal models for MS and in a phase II acute optic neuritis clinical trial (RENEW). The mechanism by which LINGO-1 regulates oligodendrocyte differentiation is unknown. Here, we demonstrate that LINGO-1 regulates oligodendrocyte differentiation and maturation through the cytoplasmic gelsolin signaling pathway, providing new drug targets for the treatment of demyelination diseases.


Asunto(s)
Actinas/metabolismo , Diferenciación Celular/fisiología , Gelsolina/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Oligodendroglía/citología , Oligodendroglía/fisiología , Animales , Células Cultivadas , Citoplasma/metabolismo , Femenino , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología
4.
Proc Natl Acad Sci U S A ; 111(21): E2229-36, 2014 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-24825889

RESUMEN

In the majority of microsatellite-stable colorectal cancers (CRCs), an initiating mutation occurs in the adenomatous polyposis coli (APC) or ß-catenin gene, activating the ß-catenin/TCF pathway. The progression of resulting adenomas is associated with oncogenic activation of KRas and inactivation of the p53 and TGF-ß/Smad functions. Most established CRC cell lines contain mutations in the TGF-ß/Smad pathway, but little is known about the function of TGF-ß in the early phases of intestinal tumorigenesis. We used mouse and human ex vivo 3D intestinal organoid cultures and in vivo mouse models to study the effect of TGF-ß on the Lgr5(+) intestinal stem cells and their progeny in intestinal adenomas. We found that the TGF-ß-induced apoptosis in Apc-mutant organoids, including the Lgr5(+) stem cells, was mediated by up-regulation of the BH3-only proapoptotic protein Bcl-2-like protein 11 (Bim). BH3-mimetic compounds recapitulated the effect of Bim not only in the adenomas but also in human CRC organoids that had lost responsiveness to TGF-ß-induced apoptosis. However, wild-type intestinal crypts were markedly less sensitive to TGF-ß than Apc-mutant adenomas, whereas the KRas oncogene increased resistance to TGF-ß via the activation of the Erk1/2 kinase pathway, leading to Bim down-regulation. Our studies identify Bim as a critical mediator of TGF-ß-induced apoptosis in intestinal adenomas and show that the common progression mutations modify Bim levels and sensitivity to TGF-ß during intestinal adenoma development.


Asunto(s)
Adenoma/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/genética , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Intestinales/genética , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Proteína 11 Similar a Bcl2 , Western Blotting , Células Cultivadas , Cromatografía en Gel , Cartilla de ADN/genética , Citometría de Flujo , Humanos , Ratones , Análisis por Micromatrices , Organoides/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo
5.
Proc Natl Acad Sci U S A ; 110(15): E1380-9, 2013 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-23520048

RESUMEN

Efficient wound healing is required to maintain the integrity of the intestinal epithelial barrier because of its constant exposure to a large variety of environmental stresses. This process implies a partial cell depolarization and the acquisition of a motile phenotype that involves rearrangements of the actin cytoskeleton. Here we address how polarized enterocytes harboring actin-rich apical microvilli undergo extensive cell remodeling to drive injury repair. Using live imaging technologies, we demonstrate that enterocytes in vitro and in vivo rapidly depolarize their microvilli at the wound edge. Through its F-actin-severing activity, the microvillar actin-binding protein villin drives both apical microvilli disassembly in vitro and in vivo and promotes lamellipodial extension. Photoactivation experiments indicate that microvillar actin is mobilized at the lamellipodium, allowing optimal migration. Finally, efficient repair of colonic mechanical injuries requires villin severing of F-actin, emphasizing the importance of villin function in intestinal homeostasis. Thus, villin severs F-actin to ensure microvillus depolarization and enterocyte remodeling upon injury. This work highlights the importance of specialized apical pole disassembly for the repolarization of epithelial cells initiating migration.


Asunto(s)
Actinas/química , Enterocitos/citología , Proteínas de Microfilamentos/fisiología , Actinas/metabolismo , Animales , Apoptosis , Diferenciación Celular , Línea Celular , Movimiento Celular , Proliferación Celular , Endoscopía , Enterocitos/metabolismo , Femenino , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Microvellosidades/metabolismo , Fenotipo , Porcinos , Cicatrización de Heridas
6.
Traffic ; 13(5): 665-80, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22321127

RESUMEN

Myosin VI has been implicated in various steps of organelle dynamics. However, the molecular mechanism by which this myosin contributes to membrane traffic is poorly understood. Here, we report that myosin VI is associated with a lysosome-related organelle, the melanosome. Using an actin-based motility assay and video microscopy, we observed that myosin VI does not contribute to melanosome movements. Myosin VI expression regulates instead the organization of actin networks in the cytoplasm. Using a cell-free assay, we showed that myosin VI recruited actin at the surface of isolated melanosomes. Myosin VI is involved in the endocytic-recycling pathway, and this pathway contributes to the transport of a melanogenic enzyme to maturing melanosomes. We showed that depletion of myosin VI accumulated a melanogenic enzyme in enlarged melanosomes and increased their melanin content. We confirmed the requirement of myosin VI to regulate melanosome biogenesis by analysing the morphology of melanosomes in choroid cells from of the Snell's waltzer mice that do not express myosin VI. Together, our results provide new evidence that myosin VI regulates the organization of actin dynamics at the surface of a specialized organelle and unravel a novel function of this myosin in regulating the biogenesis of this organelle.


Asunto(s)
Actinas/metabolismo , Melanosomas/metabolismo , Cadenas Pesadas de Miosina/fisiología , Actinas/química , Animales , Membrana Celular/metabolismo , Coroides/citología , Citoplasma/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Ratones , Ratones Transgénicos , Microscopía Electrónica/métodos , Microscopía por Video/métodos , Microtúbulos/metabolismo , Modelos Biológicos , Cadenas Pesadas de Miosina/química , Miosinas/metabolismo , Pigmentación
7.
J Cell Sci ; 124(Pt 20): 3414-27, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22010197

RESUMEN

The Scar/Wave complex (SWC) generates lamellipodia through Arp2/3-dependent polymerisation of branched actin networks. In order to identify new SWC regulators, we conducted a screen in Drosophila cells combining proteomics with functional genomics. This screen identified Clathrin heavy chain (CHC) as a protein that binds to the SWC and whose depletion affects lamellipodium formation. This role of CHC in lamellipodium formation can be uncoupled from its role in membrane trafficking by several experimental approaches. Furthermore, CHC is detected in lamellipodia in the absence of the adaptor and accessory proteins of endocytosis. We found that CHC overexpression decreased membrane recruitment of the SWC, resulting in reduced velocity of protrusions and reduced cell migration. By contrast, when CHC was targeted to the membrane by fusion to a myristoylation sequence, we observed an increase in membrane recruitment of the SWC, protrusion velocity and cell migration. Together these data suggest that, in addition to its classical role in membrane trafficking, CHC brings the SWC to the plasma membrane, thereby controlling lamellipodium formation.


Asunto(s)
Clatrina/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Microfilamentos/metabolismo , Seudópodos/metabolismo , Animales , Movimiento Celular/genética , Extensiones de la Superficie Celular/metabolismo , Extensiones de la Superficie Celular/patología , Clatrina/genética , Drosophila , Proteínas de Drosophila/genética , Células HeLa , Humanos , Proteínas de Microfilamentos/genética , Unión Proteica/genética , Transporte de Proteínas/genética , Proteómica , Seudópodos/patología , Eliminación de Secuencia/genética , Transgenes/genética , Familia de Proteínas del Síndrome de Wiskott-Aldrich/genética , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo
8.
EMBO J ; 27(1): 38-50, 2008 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-18046454

RESUMEN

The remodeling of epithelial monolayers induced by hepatocyte growth factor (HGF) results in the reorganization of actin cytoskeleton and cellular junctions. We previously showed that the membrane-cytoskeleton linker ezrin plays a major role in HGF-induced morphogenic effects. Here we identified a novel partner of phosphorylated ezrin, the Fes kinase, that acts downstream of ezrin in HGF-mediated cell scattering. We found that Fes interacts directly, through its SH2 domain, with ezrin phosphorylated at tyrosine 477. We show that in epithelial cells, activated Fes localizes either to focal adhesions or cell-cell contacts depending on cell confluency. The recruitment and the activation of Fes to the cell-cell contacts in confluent cells depend on its interaction with ezrin. When this interaction is impaired, Fes remains in focal adhesions and as a consequence the cells show defective spreading and scattering in response to HGF stimulation. Altogether, these results provide a novel mechanism whereby ezrin/Fes interaction at cell-cell contacts plays an essential role in HGF-induced cell scattering and implicates Fes in the cross-talk between cell-cell and cell-matrix adhesion.


Asunto(s)
Movimiento Celular/fisiología , Proteínas del Citoesqueleto/fisiología , Factor de Crecimiento de Hepatocito/fisiología , Proteínas Proto-Oncogénicas c-fes/metabolismo , Animales , Adhesión Celular/fisiología , Comunicación Celular/fisiología , Uniones Célula-Matriz/fisiología , Activación Enzimática/fisiología , Células LLC-PK1 , Porcinos
9.
Exp Cell Res ; 317(19): 2740-7, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21745469

RESUMEN

Notch signaling has been recently shown to have a fundamental role in stem cell maintenance and control of proper homeostasis in the intestine of different species. Here, we briefly review the current literature on Notch signals in the intestine of Drosophila, Zebrafish and the mouse, and try to highlight conserved and divergent Notch functions across species. Notch signals show a remarkably conserved role in skewing cell fate choices in intestinal lineages throughout evolution. Genetic analysis demonstrates that loss of Notch signaling invariably leads to increased numbers of secretory cells and loss of enterocytes, while gain of Notch function will completely block secretory cell differentiation. Finally, we discuss the potential contribution of Notch signaling to the initiation of colorectal cancer by controlling the maintenance of the undifferentiated state of intestinal neoplastic cells and speculate on the therapeutic consequences of affecting cancer stem cells.


Asunto(s)
Drosophila , Homeostasis/genética , Intestinos/fisiología , Ratones , Receptores Notch/fisiología , Pez Cebra , Animales , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/patología , Drosophila/genética , Drosophila/metabolismo , Drosophila/fisiología , Homeostasis/fisiología , Humanos , Mucosa Intestinal/metabolismo , Neoplasias Intestinales/genética , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Ratones/genética , Ratones/metabolismo , Ratones/fisiología , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Especificidad de la Especie , Pez Cebra/genética , Pez Cebra/metabolismo , Pez Cebra/fisiología
10.
Proc Natl Acad Sci U S A ; 106(15): 6309-14, 2009 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-19251639

RESUMEN

Notch and Wnt signals play essential roles in intestinal development and homeostasis, yet how they integrate their action to affect intestinal morphogenesis is not understood. We examined the interplay between these two signaling pathways in vivo, by modulating Notch activity in mice carrying either a loss- or a gain-of-function mutation of Wnt signaling. We find that the dramatic proliferative effect that Notch signals have on early intestinal precursors requires normal Wnt signaling, whereas its influence on intestinal differentiation appears independent of Wnt. Analogous experiments in Drosophila demonstrate that the synergistic effects of Notch and Wnt are valid across species. We also demonstrate a striking synergy between Notch and Wnt signals that results in inducing the formation of intestinal adenomas, particularly in the colon, a region rarely affected in available mouse tumor models, but the primary target organ in human patients. These studies thus reveal a previously unknown oncogenic potential of Notch signaling in colorectal tumorigenesis that, significantly, is supported by the analysis of human tumors. Importantly, our experimental evidence raises the possibility that Notch activation might be an essential initial event triggering colorectal cancer.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Receptores Notch/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , Adenoma/genética , Adenoma/metabolismo , Adenoma/patología , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Diferenciación Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Humanos , Neoplasias Intestinales/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Receptores Notch/genética , Tasa de Supervivencia , Factores de Transcripción TCF/deficiencia , Factores de Transcripción TCF/genética , Factores de Transcripción TCF/metabolismo , Factor de Transcripción 4 , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
11.
Curr Opin Cell Biol ; 14(1): 104-9, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11792551

RESUMEN

The ERM (ezrin, radixin and moesin) family of proteins are linkers that tether actin microfilaments to the plasma membrane. Merlin, the NF2 tumor suppressor gene product, is highly homologous to ERM proteins. In ERM proteins and merlin, interdomain binding promotes auto-inhibition and homo-oligomerization or hetero-oligomerization. Recent studies have revealed that ERM proteins transduce growth signals, and have shed new light on how merlin links cell growth to the cytoskeleton.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Proteínas de Microfilamentos/fisiología , Neurofibromina 2/fisiología , Animales , Proteínas Sanguíneas/fisiología , División Celular , Proteínas del Citoesqueleto/fisiología , Proteínas de la Membrana/fisiología , Microvellosidades/ultraestructura , Modelos Biológicos , Neoplasias/etiología , Fosfoproteínas/fisiología , Transducción de Señal
12.
Nature ; 435(7044): 964-8, 2005 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-15959516

RESUMEN

The Notch signalling pathway plays a crucial role in specifying cellular fates in metazoan development by regulating communication between adjacent cells. Correlative studies suggested an involvement of Notch in intestinal development. Here, by modulating Notch activity in the mouse intestine, we directly implicate Notch signals in intestinal cell lineage specification. We also show that Notch activation is capable of amplifying the intestinal progenitor pool while inhibiting cell differentiation. We conclude that Notch activity is required for the maintenance of proliferating crypt cells in the intestinal epithelium.


Asunto(s)
Linaje de la Célula , Mucosa Intestinal/metabolismo , Intestinos/citología , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Animales , Apoptosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular , Proliferación Celular , Proteínas de Unión al ADN/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Integrasas/genética , Integrasas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Notch1 , Receptores de Superficie Celular/genética , Factor de Transcripción HES-1 , Factores de Transcripción/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
13.
C R Biol ; 344(1): 27-42, 2021 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-34213847

RESUMEN

This review provides an update on the different therapeutic approaches that have been used to treat SARS-CoV-2 infection, as well as, the resulting paradoxical inflammation disorders.


Cette revue fait le point sur les différentes approches thérapeutiques qui ont été suivies pour traiter l'infection à SARS-CoV-2, ainsi que les troubles liés à l'inflammation paradoxale qui en découlent.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Preparaciones Farmacéuticas , Antivirales/uso terapéutico , Humanos , SARS-CoV-2
14.
Traffic ; 9(4): 492-509, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18194411

RESUMEN

Important progress has been made during the past decade in the identification of molecular motors required in the distribution of early and late endosomes and the proper trafficking along the endocytic pathway. There is little direct evidence, however, that these motors drive movement of the endosomes. To evaluate the contributions of kinesin-1, dynein and kinesin-2 to the movement of early and late endosomes along microtubules, we made use of a cytosol-free motility assay using magnetically isolated early and late endosomes as well as biochemical analyses and live-cell imaging. By making use of specific antibodies, we confirmed that kinesin-1 and dynein move early endosomes and we found that kinesin-2 moves both early and late endosomes in the cell-free assay. Unexpectedly, dynein did not move late endosomes in the cell-free assay. We provide evidence from disruption of dynein function and latrunculin A treatment, suggesting that dynein regulates late endosome movement indirectly, possibly through a mechanism involving the actin cytoskeleton. These data provide new insights into the complex regulation of endosomes' motility and suggest that dynein is not the major motor required to move late endosomes toward the minus end of microtubules.


Asunto(s)
Endocitosis/fisiología , Endosomas/metabolismo , Proteínas de Microtúbulos/metabolismo , Microtúbulos/metabolismo , Proteínas Motoras Moleculares/metabolismo , Animales , Dineínas/metabolismo , Endosomas/ultraestructura , Aparato de Golgi/genética , Aparato de Golgi/metabolismo , Células HeLa , Humanos , Cinesinas/metabolismo , Proteínas de Membrana de los Lisosomas/genética , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/metabolismo , Magnetismo , Proteínas de Microtúbulos/genética , Proteínas Motoras Moleculares/genética , Nanopartículas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab5/genética , Proteínas de Unión al GTP rab5/metabolismo , Proteínas de Unión a GTP rab7
15.
Mol Cell Biol ; 27(21): 7551-9, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17785439

RESUMEN

The Wnt signaling pathway is deregulated in over 90% of human colorectal cancers. beta-Catenin, the central signal transducer of the Wnt pathway, can directly modulate gene expression by interacting with transcription factors of the TCF/LEF family. In the present study we investigate the role of Wnt signaling in the homeostasis of intestinal epithelium by using tissue-specific, inducible beta-catenin gene ablation in adult mice. Block of Wnt/beta-catenin signaling resulted in rapid loss of transient-amplifying cells and crypt structures. Importantly, intestinal stem cells were induced to terminally differentiate upon deletion of beta-catenin, resulting in a complete block of intestinal homeostasis and fatal loss of intestinal function. Transcriptional profiling of mutant crypt mRNA isolated by laser capture microdissection confirmed those observations and allowed us to identify genes potentially responsible for the functional preservation of intestinal stem cells. Our data demonstrate an essential requirement of Wnt/beta-catenin signaling for the maintenance of the intestinal epithelium in the adult organism. This challenges attempts to target aberrant Wnt signaling as a new therapeutic strategy to treat colorectal cancer.


Asunto(s)
Homeostasis , Intestinos/citología , Intestinos/fisiología , Células Madre/citología , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Biomarcadores/metabolismo , Muerte Celular , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Regulación hacia Abajo , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/ultraestructura , Intestinos/ultraestructura , Ratones , Ratones Mutantes , Transducción de Señal , Células Madre/metabolismo , Transcripción Genética , beta Catenina/deficiencia
16.
Mol Biol Cell ; 18(3): 827-38, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17182858

RESUMEN

Villin, an actin-binding protein associated with the actin bundles that support microvilli, bundles, caps, nucleates, and severs actin in a calcium-dependant manner in vitro. We hypothesized that the severing activity of villin is responsible for its reported role in enhancing cell plasticity and motility. To test this hypothesis, we chose a loss of function strategy and introduced mutations in villin based on sequence comparison with CapG. By pyrene-actin assays, we demonstrate that this mutant has a strongly reduced severing activity, whereas nucleation and capping remain unaffected. The bundling activity and the morphogenic effects of villin in cells are also preserved in this mutant. We thus succeeded in dissociating the severing from the three other activities of villin. The contribution of villin severing to actin dynamics is analyzed in vivo through the actin-based movement of the intracellular bacteria Shigella flexneri in cells expressing villin and its severing variant. The severing mutations abolish the gain of velocity induced by villin. To further analyze this effect, we reconstituted an in vitro actin-based bead movement in which the usual capping protein is replaced by either the wild type or the severing mutant of villin. Confirming the in vivo results, villin-severing activity enhances the velocity of beads by more than two-fold and reduces the density of actin in the comets. We propose a model in which, by severing actin filaments and capping their barbed ends, villin increases the concentration of actin monomers available for polymerization, a mechanism that might be paralleled in vivo when an enterocyte undergoes an epithelio-mesenchymal transition.


Asunto(s)
Actinas/metabolismo , Movimiento Celular , Proteínas de Microfilamentos/metabolismo , Proteínas de Capping de la Actina/metabolismo , Citoesqueleto de Actina/ultraestructura , Secuencia de Aminoácidos , Animales , Perros , Humanos , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/ultraestructura , Microesferas , Modelos Biológicos , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestructura , Mutación/genética , Conejos , Shigella flexneri/citología , Porcinos
17.
Mol Biol Cell ; 18(12): 4780-93, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17881735

RESUMEN

The mechanisms underlying functional interactions between ERM (ezrin, radixin, moesin) proteins and Rho GTPases are not well understood. Here we characterized the interaction between ezrin and a novel Rho guanine nucleotide exchange factor, PLEKHG6. We show that ezrin recruits PLEKHG6 to the apical pole of epithelial cells where PLEKHG6 induces the formation of microvilli and membrane ruffles. These morphological changes are inhibited by dominant negative forms of RhoG. Indeed, we found that PLEKHG6 activates RhoG and to a much lesser extent Rac1. In addition we show that ezrin forms a complex with PLEKHG6 and RhoG. Furthermore, we detected a ternary complex between ezrin, PLEKHG6, and the RhoG effector ELMO. We demonstrate that PLEKHG6 and ezrin are both required in macropinocytosis. After down-regulation of either PLEKHG6 or ezrin expression, we observed an inhibition of dextran uptake in EGF-stimulated A431 cells. Altogether, our data indicate that ezrin allows the local activation of RhoG at the apical pole of epithelial cells by recruiting upstream and downstream regulators of RhoG and that both PLEKHG6 and ezrin are required for efficient macropinocytosis.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Células Epiteliales/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Secuencia Conservada , Proteínas del Citoesqueleto/genética , Factores de Intercambio de Guanina Nucleótido/química , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Ratones , Datos de Secuencia Molecular , Unión Proteica , Alineación de Secuencia , Proteínas de Unión al GTP rho/genética
18.
Adv Cancer Res ; 100: 85-111, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18620093

RESUMEN

In this review, the major signal transduction pathways that have been shown to play an important role in intestinal homeostasis are highlighted. Each of them, the Wnt, Notch, Hedgehog, and Bone Morphogenetic Protein, as well as growth-factor regulated Receptor Tyrosine Kinases are depicted with a special emphasis through their involvement in stem cell maintenance and their role in intestinal tumorigenesis. Finally, we discuss recent data on the final steps of tumor progression, notably the formation of distant metastases. This multistep process is highly complex and still far from being understood while being of major importance for the survival of patients with digestive cancer.


Asunto(s)
Mucosa Intestinal/embriología , Neoplasias Intestinales/genética , Morfogénesis/genética , Transducción de Señal/genética , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/fisiología , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas Hedgehog/genética , Proteínas Hedgehog/fisiología , Humanos , Mucosa Intestinal/fisiología , Neoplasias Intestinales/patología , Modelos Biológicos , Morfogénesis/fisiología , Metástasis de la Neoplasia , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/fisiología , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores Notch/genética , Receptores Notch/fisiología , Proteínas Wnt/genética , Proteínas Wnt/fisiología , beta Catenina/genética , beta Catenina/fisiología
19.
J Cell Biol ; 165(2): 175-80, 2004 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-15096524

RESUMEN

The GTPase Rab13 regulates the assembly of functional epithelial tight junctions (TJs) through a yet unknown mechanism. Here, we show that expression of the GTP-bound form of Rab13 inhibits PKA-dependent phosphorylation and TJ recruitment of the vasodilator-stimulated phosphoprotein, an actin remodelling protein. We demonstrate that Rab13GTP directly binds to PKA and inhibits its activity. Interestingly, activation of PKA abrogates the inhibitory effect of Rab13 on the recruitment of vasodilator-stimulated phosphoprotein, ZO-1, and claudin1 to cell-cell junctions. Rab13 is, therefore, the first GTPase that controls PKA activity and provides an unexpected link between PKA signaling and the dynamics of TJ assembly.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Transducción de Señal/fisiología , Uniones Estrechas/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Línea Celular , Claudina-1 , Inhibidores Enzimáticos/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación , Subunidades de Proteína/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteínas de Unión al GTP rab/genética
20.
J Cell Biol ; 164(5): 653-9, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-14993232

RESUMEN

Ezrin, a membrane-actin cytoskeleton linker, which participates in epithelial cell morphogenesis, is held inactive in the cytoplasm through an intramolecular interaction. Phosphatidylinositol 4,5-bisphosphate (PIP2) binding and the phosphorylation of threonine 567 (T567) are involved in the activation process that unmasks both membrane and actin binding sites. Here, we demonstrate that ezrin binding to PIP2, through its NH2-terminal domain, is required for T567 phosphorylation and thus for the conformational activation of ezrin in vivo. Furthermore, we found that the T567D mutation mimicking T567 phosphorylation bypasses the need for PIP2 binding for unmasking both membrane and actin binding sites. However, PIP2 binding and T567 phosphorylation are both necessary for the correct apical localization of ezrin and for its role in epithelial cell morphogenesis. These results establish that PIP2 binding and T567 phosphorylation act sequentially to allow ezrin to exert its cellular functions.


Asunto(s)
Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatidilinositoles/metabolismo , Fosfoproteínas/metabolismo , Animales , Sitios de Unión , Línea Celular , Membrana Celular/metabolismo , Polaridad Celular , Tamaño de la Célula , Proteínas del Citoesqueleto , Células Epiteliales/citología , Células Epiteliales/metabolismo , Morfogénesis/fisiología , Fosforilación , Mutación Puntual , Unión Proteica , Treonina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA