Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
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.
Med Sci (Paris) ; 34(1): 63-71, 2018 Jan.
Artículo en Francés | MEDLINE | ID: mdl-29384098

RESUMEN

Genetic and most likely epigenetic alterations occurring during tumor progression and metastatic process lead to a broad deregulation of major cellular functions. However, the molecular mechanisms involved are still poorly understood. To understand them, the cell, the basic unit of life, remains more than ever the essential level to integrate the functional impact of genetics and epigenetics processes in the light of the global economy of the normal and cancerous cell, and of its interactions with its microenvironment.


Asunto(s)
Investigación Biomédica/tendencias , Fenómenos Fisiológicos Celulares/fisiología , Neoplasias/etiología , Neoplasias/terapia , Animales , Epigénesis Genética/fisiología , Humanos , Neoplasias/diagnóstico , Integración de Sistemas
4.
Neoplasia ; 18(1): 10-24, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26806348

RESUMEN

The Hippo signaling network is a key regulator of cell fate. In the recent years, it was shown that its implication in cancer goes well beyond the sole role of YAP transcriptional activity and its regulation by the canonical MST/LATS kinase cascade. Here we show that the motin family member AMOTL1 is an important effector of Hippo signaling in breast cancer. AMOTL1 connects Hippo signaling to tumor cell aggressiveness. We show that both canonical and noncanonical Hippo signaling modulates AMOTL1 levels. The tumor suppressor Merlin triggers AMOTL1 proteasomal degradation mediated by the NEDD family of ubiquitin ligases through direct interaction. In parallel, YAP stimulates AMOTL1 expression. The loss of Merlin expression and the induction of Yap activity that are frequently observed in breast cancers thus result in elevated AMOTL1 levels. AMOTL1 expression is sufficient to trigger tumor cell migration and stimulates proliferation by activating c-Src. In a large cohort of human breast tumors, we show that AMOTL1 protein levels are upregulated during cancer progression and that, importantly, the expression of AMOTL1 in lymph node metastasis appears predictive of the risk of relapse. Hence we uncover an important mechanism by which Hippo signaling promotes breast cancer progression by modulating the expression of AMOTL1.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteínas de la Membrana/metabolismo , Neurofibromina 2/metabolismo , Angiomotinas , Animales , Neoplasias de la Mama/genética , Proteínas de Ciclo Celular , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal/genética , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Xenoinjertos , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Neurofibromina 2/genética , Proteínas Nucleares/metabolismo , Unión Proteica , Proteolisis , Transducción de Señal , Factores de Transcripción/metabolismo , Familia-src Quinasas/metabolismo
5.
Nat Commun ; 5: 5005, 2014 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-25295490

RESUMEN

Epithelial-to-mesenchymal transition-like (EMT-like) is a critical process allowing initiation of metastases during tumour progression. Here, to investigate its role in intestinal cancer, we combine computational network-based and experimental approaches to create a mouse model with high metastatic potential. Construction and analysis of this network map depicting molecular mechanisms of EMT regulation based on the literature suggests that Notch activation and p53 deletion have a synergistic effect in activating EMT-like processes. To confirm this prediction, we generate transgenic mice by conditionally activating the Notch1 receptor and deleting p53 in the digestive epithelium (NICD/p53(-/-)). These mice develop metastatic tumours with high penetrance. Using GFP lineage tracing, we identify single malignant cells with mesenchymal features in primary and metastatic tumours in vivo. The development of such a model that recapitulates the cellular features observed in invasive human colorectal tumours is appealing for innovative drug discovery.


Asunto(s)
Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/fisiología , Tracto Gastrointestinal/fisiología , Metástasis de la Neoplasia/fisiopatología , Receptor Notch1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Secuencia de Bases , Linaje de la Célula , Cartilla de ADN/genética , Exoma/genética , Tracto Gastrointestinal/metabolismo , Genotipo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Transducción de Señal/genética , Transducción de Señal/fisiología
6.
Eur J Cell Biol ; 93(10-12): 388-95, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25269996

RESUMEN

While absent from normal epithelia, an actin bundling protein, fascin, becomes expressed in invasive carcinoma of different origins. It is highly enriched at the tumors' invasive front suggesting that it could play a role in cancer invasion. Multiple studies have shown that fascin, through its role in formation of cellular protrusions such as filopodia and invadopodia, enhances cancer cell migration and invasion in vitro. However, the role of fascin in vivo remains unknown. We have generated a compound transgenic mouse model that allows expression of fascin in the intestinal epithelium in the Apc-mutated background. Conditional expression of fascin led to decrease in mice survival and increase in tumor burden compared to control animals. Induction of fascin expression in adult tumor-bearing animals accelerated tumor progression and led to formation of invasive adenocarcinoma. Altogether, our study shows that fascin can promote tumor progression in vivo, but also unravels an unexpected role of fascin in tumor initiation.


Asunto(s)
Adenocarcinoma/metabolismo , Proteínas Portadoras/genética , Neoplasias Colorrectales/metabolismo , Proteínas de Microfilamentos/genética , Adenocarcinoma/patología , Animales , Proteínas Portadoras/metabolismo , Proliferación Celular/genética , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia
7.
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
8.
Neuro Oncol ; 16(9): 1196-209, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24558021

RESUMEN

BACKGROUND: Inactivation of the NF2 gene predisposes to neurofibromatosis type II and the development of schwannomas. In vitro studies have shown that loss of NF2 leads to the induction of mitogenic signaling mediated by receptor tyrosine kinases (RTKs), MAP kinase, AKT, or Hippo pathways. The goal of our study was to evaluate the expression and activity of these signaling pathways in human schwannomas in order to identify new potential therapeutic targets. METHODS: Large sets of human schwannomas, totaling 68 tumors, were analyzed using complementary proteomic approaches. RTK arrays identified the most frequently activated RTKs. The correlation between the expression and activity of signaling pathways and proliferation of tumor cells using Ki67 marker was investigated by reverse-phase protein array (RRPA). Finally, immunohistochemistry was used to evaluate the expression pattern of signaling effectors in the tumors. RESULTS: We showed that Her2, Her3, PDGFRß, Axl, and Tie2 are frequently activated in the tumors. Furthermore, RRPA demonstrated that Ki67 levels are linked to YAP, p-Her3, and PDGFRß expression levels. In addition, Her2, Her3, and PDGFRß are transcriptional targets of Yes-associated protein (YAP) in schwannoma cells in culture. Finally, we observed that the expression of these signaling effectors is very variable between tumors. CONCLUSIONS: Tumor cell proliferation in human schwannomas is linked to a signaling network controlled by the Hippo effector YAP. Her2, Her3, PDGFRß, Axl, and Tie2, as well as YAP, represent potentially valuable therapeutic targets. However, the variability of their expression between tumors may result in strong differences in the response to targeted therapy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias Encefálicas/metabolismo , Neurilemoma/metabolismo , Neurofibromatosis 2/metabolismo , Fosfoproteínas/metabolismo , Transducción de Señal , Proliferación Celular , Femenino , Humanos , Masculino , Proteómica , Factores de Transcripción , Proteínas Señalizadoras YAP
9.
Eur J Cell Biol ; 91(11-12): 930-7, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22939225

RESUMEN

During metastasis, cancer cells breach the basement membrane and migrate through the stroma mostly composed of a network of collagen I fibers. Cell migration on 2D is initiated by protrusion of the cell membrane followed by formation of adhesions that link the actin cytoskeleton to the extracellular matrix (ECM). Cells then move forwards by exerting traction forces on the adhesions at its front and by disassembling adhesions at the rear. In 2D, only the ventral surface of a migrating cell is in contact with the ECM, where cell-matrix adhesions are assembled. In 3D matrices, even though the whole surface of a migrating cell is available for interacting with the ECM, it is unclear whether discrete adhesion structures actually exist. Using high-resolution confocal microscopy we imaged the endogenous adhesome proteins in three different cancer cell types embedded in non-pepsinized collagen type I, polymerized at a slow rate, to allow the formation of a network that resembles the organization of EMC observed in vivo. Vinculin aggregates were detected in the cellular protrusions, frequently colocalizing with collagen fibers, implying they correspond to adhesion structures in 3D. As the distance from the substrate bottom increases, adhesion aggregates become smaller and almost undetectable in some cell lines. Using intravital imaging we show here, for the first time, the existence of adhesome proteins aggregates in vivo. These aggregates share similarities with the ones found in 3D collagen matrices. It still remains to be determined if adhesions assembled in 3D and in vivo share functional similarities to the well-described adhesions in 2D. This will provide a major step forward in understanding cell migration in more physiological environments.


Asunto(s)
Adhesión Celular , Colágeno Tipo I/química , Neoplasias/ultraestructura , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Adhesiones Focales/metabolismo , Adhesiones Focales/ultraestructura , Células HCT116 , Humanos , Conformación Molecular , Vinculina/metabolismo
10.
PLoS One ; 7(5): e37490, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22629406

RESUMEN

The membrane cytoskeleton linker ezrin participates in several functions downstream of the receptor Met in response to Hepatocyte Growth Factor (HGF) stimulation. Here we report a novel interaction of ezrin with a HECT E3 ubiquitin ligase, WWP1/Aip5/Tiul1, a potential oncogene that undergoes genomic amplification and overexpression in human breast and prostate cancers. We show that ezrin binds to the WW domains of WWP1 via the consensus motif PPVY(477) present in ezrin's C-terminus. This association results in the ubiquitylation of ezrin, a process that requires an intact PPVY(477) motif. Interestingly ezrin ubiquitylation does not target the protein for degradation by the proteasome. We find that ezrin ubiquitylation by WWP1 in epithelial cells leads to the upregulation of Met level in absence of HGF stimulation and increases the response of Met to HGF stimulation as measured by the ability of the cells to heal a wound. Interestingly this effect requires ubiquitylated ezrin since it can be rescued, after depletion of endogenous ezrin, by wild type ezrin but not by a mutant of ezrin that cannot be ubiquitylated. Taken together our data reveal a new role for ezrin in Met receptor stability and activity through its association with the E3 ubiquitin ligase WWP1. Given the role of Met in cell proliferation and tumorigenesis, our results may provide a mechanistic basis for understanding the role of ezrin in tumor progression.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/fisiología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Proteínas del Citoesqueleto/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Células Tumorales Cultivadas , Ubiquitina-Proteína Ligasas/genética
11.
Sci Transl Med ; 4(130): 130fs7, 2012 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-22517881

RESUMEN

A single mouse Lgr5-positive colon stem cell can be expanded into a three-dimensional organoid that, after transplant, contributes to the repair of injured colon epithelia in a mouse model of colitis.


Asunto(s)
Colon/citología , Células Madre/citología , Animales , Colitis/terapia , Ratones , Organoides/citología , Trasplante de Células Madre , Células Madre/fisiología
12.
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
13.
Mol Biol Cell ; 23(6): 1080-94, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22262457

RESUMEN

The mechanisms that regulate actin filament polymerization resulting in the morphogenesis of the brush border microvilli in epithelial cells remain unknown. Eps8, the prototype of a family of proteins capable of capping and bundling actin filaments, has been shown to bundle the microvillar actin filaments. We report that Eps8L1a, a member of the Eps8 family and a novel ezrin-interacting partner, controls microvillus length through its capping activity. Depletion of Eps8L1a leads to the formation of long microvilli, whereas its overexpression has the opposite effect. We demonstrate that ezrin differentially modulates the actin-capping and -bundling activities of Eps8 and Eps8L1a during microvillus assembly. Coexpression of ezrin with Eps8 promotes the formation of membrane ruffles and tufts of microvilli, whereas expression of ezrin and Eps8L1a induces the clustering of actin-containing structures at the cell surface. These distinct morphological changes are neither observed when a mutant of ezrin defective in its binding to Eps8/Eps8L1a is coexpressed with Eps8 or Eps8L1a nor observed when ezrin is expressed with mutants of Eps8 or Eps8L1a defective in the actin-bundling or -capping activities, respectively. Our data show a synergistic effect of ezrin and Eps8 proteins in the assembly and organization of actin microvillar filaments.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Túbulos Renales Proximales/citología , Microvellosidades/metabolismo , Actinas/metabolismo , Animales , Células Epiteliales/metabolismo , Células LLC-PK1 , Dominios y Motivos de Interacción de Proteínas , Sus scrofa , Porcinos
14.
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
15.
Eur J Cell Biol ; 90(2-3): 93-9, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-20609495

RESUMEN

The basement membrane (BM) is a dense, tightly cross-linked matrix that acts as physiological barrier to maintain tissue homeostasis. Studies on Caenorhabditis elegans, leucocytes diapedesis and cancer cell invasion have shown that BM transmigration is a conserved three-stage process. Firstly, invadopodia-like protrusions form at the ventral surface of invasive cells; later, one protrusion elongates that lastly drives the infiltration of cells into the underlying compartment. This review illustrates the mechanism used by invasive cancer cells to cross the BM barrier by focusing on the role of key cytoskeleton components. We also describe currently available in vitro assays to study each step of the BM transmigration program.


Asunto(s)
Membrana Basal/fisiología , Movimiento Celular/fisiología , Citoesqueleto/fisiología , Animales , Membrana Basal/metabolismo , Citoesqueleto/metabolismo , Humanos , Microtúbulos/metabolismo
16.
Mol Biol Cell ; 22(3): 375-85, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21148287

RESUMEN

In the degradative pathway, the progression of cargos through endosomal compartments involves a series of fusion and maturation events. The HOPS (homotypic fusion and protein sorting) complex is part of the machinery that promotes the progression from early to late endosomes and lysosomes by regulating the exchange of small GTPases. We report that an interaction between subunits of the HOPS complex and the ERM (ezrin, radixin, moesin) proteins is required for the delivery of EGF receptor (EGFR) to lysosomes. Inhibiting either ERM proteins or the HOPS complex leads to the accumulation of the EGFR into early endosomes, delaying its degradation. This impairment in EGFR trafficking observed in cells depleted of ERM proteins is due to a delay in the recruitment of Rab7 on endosomes. As a consequence, the maturation of endosomes is perturbed as reflected by an accumulation of hybrid compartments positive for both early and late endosomal markers. Thus, ERM proteins represent novel regulators of the HOPS complex in the early to late endosomal maturation.


Asunto(s)
Proteínas del Citoesqueleto/fisiología , Endosomas/metabolismo , Receptores ErbB/metabolismo , Proteínas de la Membrana/fisiología , Proteínas de Microfilamentos/fisiología , Proteínas del Citoesqueleto/análisis , Proteínas del Citoesqueleto/genética , Receptores ErbB/análisis , Células HeLa , Humanos , Lisosomas/metabolismo , Proteínas de la Membrana/análisis , Proteínas de la Membrana/genética , Proteínas de Microfilamentos/análisis , Proteínas de Microfilamentos/genética , Transporte de Proteínas , Proteínas de Transporte Vesicular/análisis , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Unión al GTP rab/análisis , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión a GTP rab7
17.
J Cell Biol ; 189(3): 541-56, 2010 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-20421424

RESUMEN

Invasive cancer cells are believed to breach the basement membrane (BM) using specialized protrusions called invadopodia. We found that the crossing of a native BM is a three-stage process: invadopodia indeed form and perforate the BM, elongate into mature invadopodia, and then guide the cell toward the stromal compartment. We studied the remodeling of cytoskeleton networks during invadopodia formation and elongation using ultrastructural analysis, spatial distribution of molecular markers, and RNA interference silencing of protein expression. We show that formation of invadopodia requires only the actin cytoskeleton and filopodia- and lamellipodia-associated proteins. In contrast, elongation of invadopodia is mostly dependent on filopodial actin machinery. Moreover, intact microtubules and vimentin intermediate filament networks are required for further growth. We propose that invadopodia form by assembly of dendritic/diagonal and bundled actin networks and then mature by elongation of actin bundles, followed by the entry of microtubules and vimentin filaments. These findings provide a link between the epithelial to mesenchymal transition and BM transmigration.


Asunto(s)
Actinas/metabolismo , Extensiones de la Superficie Celular/ultraestructura , Microtúbulos/metabolismo , Vimentina/metabolismo , Animales , Membrana Basal/metabolismo , Movimiento Celular , Células Cultivadas , Humanos , Interferencia de ARN , Transfección
18.
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
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA