Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Biol Chem ; 295(39): 13474-13487, 2020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-32690605

RESUMEN

Yes-associated protein (YAP) signaling has emerged as a crucial pathway in several normal and pathological processes. Although the main upstream effectors that regulate its activity have been extensively studied, the role of the endosomal system has been far less characterized. Here, we identified the late endosomal/lysosomal adaptor MAPK and mTOR activator (LAMTOR) complex as an important regulator of YAP signaling in a preosteoblast cell line. We found that p18/LAMTOR1-mediated peripheral positioning of late endosomes allows delivery of SRC proto-oncogene, nonreceptor tyrosine kinase (SRC) to the plasma membrane and promotes activation of an SRC-dependent signaling cascade that controls YAP nuclear shuttling. Moreover, ß1 integrin engagement and mechano-sensitive cues, such as external stiffness and related cell contractility, controlled LAMTOR targeting to the cell periphery and thereby late endosome recycling and had a major impact on YAP signaling. Our findings identify the late endosome recycling pathway as a key mechanism that controls YAP activity and explains YAP mechano-sensitivity.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Endosomas/metabolismo , Integrina beta1/metabolismo , Factores de Transcripción/metabolismo , Familia-src Quinasas/metabolismo , Animales , Proteínas de Ciclo Celular/deficiencia , Línea Celular , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Proto-Oncogenes Mas , Transducción de Señal , Factores de Transcripción/deficiencia , Familia-src Quinasas/deficiencia
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.
Neuro Oncol ; 20(7): 917-929, 2018 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-29409008

RESUMEN

Background: Clinical overlap between neurofibromatosis type 2 (NF2), schwannomatosis, and meningiomatosis can make clinical diagnosis difficult. Hence, molecular investigation of germline and tumor tissues may improve the diagnosis. Methods: We present the targeted next-generation sequencing (NGS) of NF2, SMARCB1, LZTR1, SMARCE1, and SUFU tumor suppressor genes, using an amplicon-based approach. We analyzed blood DNA from a cohort of 196 patients, including patients with NF2 (N = 79), schwannomatosis (N = 40), meningiomatosis (N = 12), and no clearly established diagnosis (N = 65). Matched tumor DNA was analyzed when available. Forty-seven NF2-/SMARCB1-negative schwannomatosis patients and 27 NF2-negative meningiomatosis patients were also evaluated. Results: A NF2 variant was found in 41/79 (52%) NF2 patients. SMARCB1 or LZTR1 variants were identified in 5/40 (12.5%) and 13/40 (∼32%) patients in the schwannomatosis cohort. Potentially pathogenic variants were found in 12/65 (18.5%) patients with no clearly established diagnosis. A LZTR1 variant was identified in 16/47 (34%) NF2/SMARCB1-negative schwannomatosis patients. A SMARCE1 variant was found in 3/39 (∼8%) meningiomatosis patients. No SUFU variant was found in the cohort. NGS was an effective and sensitive method to detect mutant alleles in blood or tumor DNA of mosaic NF2 patients. Interestingly, we identified a 4-hit mechanism resulting in the complete NF2 loss-of-function combined with SMARCB1 and LZTR1 haploinsufficiency in two-thirds of tumors from NF2 patients. Conclusions: Simultaneous investigation of NF2, SMARCB1, LZTR1, and SMARCE1 is a key element in the differential diagnosis of NF2, schwannomatosis, and meningiomatosis. The targeted NGS strategy is suitable for the identification of NF2 mosaicism in blood and for the investigation of tumors from these patients.


Asunto(s)
Genes Supresores de Tumor , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Neoplasias Meníngeas/diagnóstico , Meningioma/diagnóstico , Mutación , Neurilemoma/diagnóstico , Neurofibromatosis/diagnóstico , Neurofibromatosis 2/diagnóstico , Neoplasias Cutáneas/diagnóstico , Biomarcadores de Tumor , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/genética , Diagnóstico Diferencial , Estudios de Seguimiento , Humanos , Neoplasias Meníngeas/genética , Meningioma/genética , Neurilemoma/genética , Neurofibromatosis/genética , Neurofibromatosis 2/genética , Neurofibromina 2/genética , Pronóstico , Estudios Prospectivos , Proteínas Represoras/genética , Estudios Retrospectivos , Proteína SMARCB1/genética , Neoplasias Cutáneas/genética , Factores de Transcripción/genética
4.
J Biol Chem ; 292(47): 19179-19197, 2017 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-28972170

RESUMEN

Cell adhesion to the extracellular matrix or to surrounding cells plays a key role in cell proliferation and differentiation and is critical for proper tissue homeostasis. An important pathway in adhesion-dependent cell proliferation is the Hippo signaling cascade, which is coregulated by the transcription factors Yes-associated protein 1 (YAP1) and transcriptional coactivator with PDZ-binding motif (TAZ). However, how cells integrate extracellular information at the molecular level to regulate YAP1's nuclear localization is still puzzling. Herein, we investigated the role of ß1 integrins in regulating this process. We found that ß1 integrin-dependent cell adhesion is critical for supporting cell proliferation in mesenchymal cells both in vivo and in vitro ß1 integrin-dependent cell adhesion relied on the relocation of YAP1 to the nucleus after the down-regulation of its phosphorylated state mediated by large tumor suppressor gene 1 and 2 (LATS1/2). We also found that this phenotype relies on ß1 integrin-dependent local activation of the small GTPase RAC1 at the plasma membrane to control the activity of P21 (RAC1)-activated kinase (PAK) of group 1. We further report that the regulatory protein merlin (neurofibromin 2, NF2) interacts with both YAP1 and LATS1/2 via its C-terminal moiety and FERM domain, respectively. PAK1-mediated merlin phosphorylation on Ser-518 reduced merlin's interactions with both LATS1/2 and YAP1, resulting in YAP1 dephosphorylation and nuclear shuttling. Our results highlight RAC/PAK1 as major players in YAP1 regulation triggered by cell adhesion.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Genes de la Neurofibromatosis 2/fisiología , Integrina beta1/fisiología , Neurofibromina 2/metabolismo , Fosfoproteínas/metabolismo , Quinasas p21 Activadas/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Adhesión Celular , Proteínas de Ciclo Celular , Proliferación Celular , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Ratones , Ratones Noqueados , Neurofibromina 2/genética , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas Señalizadoras YAP , Quinasas p21 Activadas/genética , Proteína de Unión al GTP rac1/genética
5.
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
6.
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
7.
Soins ; (759): 45-7, 2011 Oct.
Artículo en Francés | MEDLINE | ID: mdl-22145439

RESUMEN

The French national agency for the assessment and quality of social and medical-social institutes and services (ANESM) recently published recommendations aiming to promote the quality of life of elderly people in nursing homes. These recommendations take into account the specific characteristics of the quality of life of this sector of the population.


Asunto(s)
Anciano Frágil/psicología , Calidad de Vida/psicología , Anciano , Anciano de 80 o más Años , Francia , Evaluación Geriátrica , Promoción de la Salud , Hogares para Ancianos , Humanos , Casas de Salud , Ajuste Social , Medio Social , Identificación Social , Factores Socioeconómicos
11.
J Cell Sci ; 122(Pt 22): 4141-9, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19910496

RESUMEN

Merlin is the product of the Nf2 tumor-suppressor gene, and inactivation of Nf2 leads to the development of neural tumors such as schwannomas and meningiomas in humans and mice. Merlin is a member of the ERM (ezrin, radixin and moesin) family of proteins that function as organizers of the actin cytoskeleton. Merlin structure is thought to be similar to that of the ERM proteins, and is held in a closed clamp conformation via intramolecular interactions of its N-terminal FERM (four-point-one, ERM) domain with an alpha-helical C-terminal domain. Like ERMs, merlin can remodel actin-rich cortical structures, yet merlin uniquely inhibits the proliferation of many different cell types. Here, we report that the F2 subdomain of the FERM domain and a domain close to the C-terminus that is defined by residues 532-579 are essential for merlin-mediated inhibition of primary Schwann cell proliferation. Furthermore, we demonstrate that the F1 subdomain of the merlin FERM domain is required for actin colocalization, proper regulation of merlin C-terminal phosphorylation and for remodeling the cytoskeleton, yet is not required for the inhibition of Schwann cell proliferation. Thus, tumor suppression by merlin is independent of its role as an organizer of the actin cytoskeleton in Schwann cells.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Proliferación Celular , Proteínas del Citoesqueleto/metabolismo , Neurofibromina 2/metabolismo , Células de Schwann/fisiología , Animales , Células Cultivadas , Proteínas del Citoesqueleto/genética , Regulación hacia Abajo , Ingeniería Genética , Ratones , Ratones Transgénicos , Neoplasias del Sistema Nervioso/genética , Neoplasias del Sistema Nervioso/patología , Neurofibromina 2/genética , Fosforilación , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Células de Schwann/patología
12.
Hum Mol Genet ; 16(14): 1742-51, 2007 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-17566081

RESUMEN

The lack of neurofibromatosis 2 tumor suppressor protein merlin leads to the formation of nervous system tumors, specifically schwannomas and meningiomas. Merlin is considered to act as a tumor suppressor at the cell membrane, where it links transmembrane receptors to the actin cytoskeleton. Several tumor suppressors interact with another component of the cytoskeleton, the microtubules, in a regulated manner and control their dynamics. In this work, we identify merlin as a novel microtubule-organizing protein. We identify two tubulin-binding sites in merlin, one residing at the N-terminal FERM-domain and another at the C-terminal domain. Merlin's intramolecular association and phosphorylation of serine 518 regulate the interaction between merlin and tubulin. Analysis of cultured glioma cells indicates colocalization between merlin and microtubules especially during cell division. In primary mouse Schwann cells only minor colocalization at the cell periphery of interphase cells is seen. However, these cells drastically change their microtubule organization upon loss of merlin indicating a functional association of the proteins. Both in vitro assays and in vivo studies in Schwann cells indicate that merlin promotes tubulin polymerization. The results show that merlin plays a key role in the regulation of the Schwann cell microtubule cytoskeleton and suggest a mechanism by which loss of merlin leads to cytoskeletal defects observed in human schwannomas.


Asunto(s)
Citoesqueleto/metabolismo , Microtúbulos/metabolismo , Neurofibromina 2/genética , Neurofibromina 2/fisiología , Células de Schwann/metabolismo , Animales , Bovinos , Línea Celular Tumoral , Escherichia coli/metabolismo , Glioma/metabolismo , Humanos , Insectos , Ratones , Modelos Biológicos , Neurofibromina 2/metabolismo , Isoformas de Proteínas , Tubulina (Proteína)/metabolismo
13.
J Cell Biol ; 177(5): 893-903, 2007 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-17548515

RESUMEN

The neurofibromatosis type 2 (NF2) tumor suppressor, Merlin, is a membrane/cytoskeleton-associated protein that mediates contact-dependent inhibition of proliferation. Here we show that upon cell-cell contact Merlin coordinates the processes of adherens junction stabilization and negative regulation of epidermal growth factor receptor (EGFR) signaling by restraining the EGFR into a membrane compartment from which it can neither signal nor be internalized. In confluent Nf2(-/-) cells, EGFR activation persists, driving continued proliferation that is halted by specific EGFR inhibitors. These studies define a new mechanism of tumor suppression, provide mechanistic insight into the poorly understood phenomenon of contact-dependent inhibition of proliferation, and suggest a therapeutic strategy for NF2-mutant tumors.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Neurofibromina 2/fisiología , Transducción de Señal , Animales , Adhesión Celular , Línea Celular , Proliferación Celular , Receptores ErbB/metabolismo , Ratones , Modelos Biológicos , Neurofibromina 2/metabolismo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Estructura Terciaria de Proteína , Intercambiadores de Sodio-Hidrógeno/química , Intercambiadores de Sodio-Hidrógeno/metabolismo
14.
Genes Dev ; 17(9): 1090-100, 2003 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-12695331

RESUMEN

Mutation of the Neurofibromatosis 2 (NF2) tumor suppressor gene leads to cancer development in humans and mice. Recent studies suggest that Nf2 loss also contributes to tumor metastasis. The Nf2-encoded protein, merlin, is related to the ERM (ezrin, radixin, and moesin) family of membrane:cytoskeleton-associated proteins. However, the cellular mechanism whereby merlin controls cell proliferation from this location is not known. Here we show that the major cellular consequence of Nf2 deficiency in primary cells is an inability to undergo contact-dependent growth arrest and to form stable cadherin-containing cell:cell junctions. Merlin colocalizes and interacts with adherens junction (AJ) components in confluent wild-type cells, suggesting that the lack of AJs and contact-dependent growth arrest in Nf2(-/-) cells directly results from the absence of merlin at sites of cell:cell contact. Our studies indicate that merlin functions as a tumor and metastasis suppressor by controlling cadherin-mediated cell:cell contact.


Asunto(s)
Uniones Adherentes/fisiología , Metástasis de la Neoplasia , Neoplasias/genética , Neurofibromina 2/deficiencia , Animales , Ratones , Neoplasias/etiología , Neurofibromina 2/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...