Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Genes Dev ; 32(17-18): 1201-1214, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30143526

RESUMEN

The architectural and biochemical features of the plasma membrane are governed by its intimate association with the underlying cortical cytoskeleton. The neurofibromatosis type 2 (NF2) tumor suppressor merlin and closely related membrane:cytoskeleton-linking protein ezrin organize the membrane:cytoskeleton interface, a critical cellular compartment that both regulates and is regulated by growth factor receptors. An example of this poorly understood interrelationship is macropinocytosis, an ancient process of nutrient uptake and membrane remodeling that can both be triggered by growth factors and manage receptor availability. We show that merlin deficiency primes the membrane:cytoskeleton interface for epidermal growth factor (EGF)-induced macropinocytosis via a mechanism involving increased cortical ezrin, altered actomyosin, and stabilized cholesterol-rich membranes. These changes profoundly alter EGF receptor (EGFR) trafficking in merlin-deficient cells, favoring increased membrane levels of its heterodimerization partner, ErbB2; clathrin-independent internalization; and recycling. Our work suggests that, unlike Ras transformed cells, merlin-deficient cells do not depend on macropinocytic protein scavenging and instead exploit macropinocytosis for receptor recycling. Finally, we provide evidence that the macropinocytic proficiency of NF2-deficient cells can be used for therapeutic uptake. This work provides new insight into fundamental mechanisms of macropinocytic uptake and processing and suggests new ways to interfere with or exploit macropinocytosis in NF2 mutant and other tumors.


Asunto(s)
Membrana Celular/metabolismo , Factor de Crecimiento Epidérmico/fisiología , Receptores ErbB/metabolismo , Neurofibromina 2/fisiología , Pinocitosis , Actomiosina/metabolismo , Animales , Células Cultivadas , Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Humanos , Ratones , Neurofibromina 2/genética , Biosíntesis de Proteínas
2.
Dev Biol ; 477: 133-144, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34044021

RESUMEN

The Drosophila testis is a model organism stem cell niche in which two stem cell populations coordinate together to produce sperm; thus, these stem cells must be balanced in the niche. Merlin, a tumor-suppressor and human disease gene required for contact inhibition of proliferation, is known to limit the proliferation of the somatic cyst stem cells in the testis niche. Expanded encodes a protein that is structurally similar to Merlin in Drosophila, and is semi-redundant with Merlin in multiple tissues. We found that expanded depletion caused similar cyst lineage cell over-proliferation as observed with Merlin, and double mutants showed more severe phenotypes than either gene individually. Thus, these genes have partially redundant functions in the cyst lineage cells of this niche. We also expressed non-phosphorylatable constitutively "tumor suppressing" alleles of Merlin in cyst lineage cells, and surprisingly, we observed a similar cyst lineage over-proliferation phenotype. Merlin is known to impact multiple different signaling pathways to exert its effect on proliferation. We found that the Merlin loss of function phenotype was associated with an increase in MAPK/ERK signaling, consistent with Merlin's established role in transmembrane receptor inhibition. Constitutive Merlin displayed a reduction in both MAPK/ERK signaling and PI3K/Tor signaling. PI3K/Tor signaling is required for cyst cell differentiation, and inhibition of this pathway by Merlin activation phenocopied the Tor cyst lineage loss of function phenotype. Thus, Merlin impacts and integrates the activity of multiple signaling pathways in the testis niche. The ability of Merlin to dynamically change its activity via phosphorylation in response to local contact cues provides an intriguing mechanism whereby the signaling pathways that control these stem cells might be dynamically regulated in response to the division of a neighboring germ cell.


Asunto(s)
Células Madre Adultas/fisiología , Proliferación Celular/fisiología , Proteínas de Drosophila/fisiología , Drosophila/citología , Proteínas de la Membrana/fisiología , Neurofibromina 2/fisiología , Transducción de Señal , Testículo/citología , Animales , Linaje de la Célula , Drosophila/embriología , Proteínas de Drosophila/metabolismo , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Masculino , Modelos Biológicos , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Péptidos de Invertebrados/metabolismo , Testículo/embriología
3.
Hum Mol Genet ; 29(20): 3373-3387, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33075808

RESUMEN

Uveal coloboma represents one of the most common congenital ocular malformations accounting for up to 10% of childhood blindness (~1 in 5000 live birth). Coloboma originates from defective fusion of the optic fissure (OF), a transient gap that forms during eye morphogenesis by asymmetric, ventral invagination. Genetic heterogeneity combined with the activity of developmentally regulated genes suggests multiple mechanisms regulating OF closure. The tumor suppressor and FERM domain protein Neurofibromin 2 (NF2) controls diverse processes in cancer, development and regeneration, via Hippo pathway and cytoskeleton regulation. In humans, NF2 mutations can cause ocular abnormalities, including coloboma, however, its actual role in OF closure is unknown. Using conditional inactivation in the embryonic mouse eye, our data indicate that loss of Nf2 function results in a novel underlying cause for coloboma. In particular, mutant eyes show substantially increased retinal pigmented epithelium (RPE) proliferation in the fissure region with concomitant acquisition of RPE cell fate. Cells lining the OF margin can maintain RPE fate ectopically and fail to transition from neuroepithelial to cuboidal shape. In the dorsal RPE of the optic cup, Nf2 inactivation leads to a robust increase in cell number, with local disorganization of the cytoskeleton components F-actin and pMLC2. We propose that RPE hyperproliferation is the primary cause for the observed defects causing insufficient alignment of the OF margins in Nf2 mutants and failure to fuse properly, resulting in persistent coloboma. Our findings indicate that limiting proliferation particularly in the RPE layer is a critical mechanism during OF closure.


Asunto(s)
Proliferación Celular , Coloboma/patología , Ojo/patología , Regulación del Desarrollo de la Expresión Génica , Neurofibromina 2/fisiología , Organogénesis , Epitelio Pigmentado de la Retina/patología , Animales , Coloboma/etiología , Coloboma/metabolismo , Ojo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Epitelio Pigmentado de la Retina/metabolismo
4.
Cell Commun Signal ; 18(1): 63, 2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32299434

RESUMEN

BACKGROUND: In this review, we describe how the cytoskeletal protein Merlin, encoded by the Neurofibromin 2 (NF2) gene, orchestrates developmental signaling to ensure normal ontogeny, and we discuss how Merlin deficiency leads to aberrant activation of developmental pathways that enable tumor development and malignant progression. MAIN BODY: Parallels between embryonic development and cancer have underscored the activation of developmental signaling pathways. Hippo, WNT/ß-catenin, TGF-ß, receptor tyrosine kinase (RTK), Notch, and Hedgehog pathways are key players in normal developmental biology. Unrestrained activity or loss of activity of these pathways causes adverse effects in developing tissues manifesting as developmental syndromes. Interestingly, these detrimental events also impact differentiated and functional tissues. By promoting cell proliferation, migration, and stem-cell like phenotypes, deregulated activity of these pathways promotes carcinogenesis and cancer progression. The NF2 gene product, Merlin, is a tumor suppressor classically known for its ability to induce contact-dependent growth inhibition. Merlin plays a role in different stages of an organism development, ranging from embryonic to mature states. While homozygous deletion of Nf2 in murine embryos causes embryonic lethality, Merlin loss in adult tissue is implicated in Neurofibromatosis type 2 disorder and cancer. These manifestations, cumulatively, are reminiscent of dysregulated developmental signaling. CONCLUSION: Understanding the molecular and cellular repercussions of Merlin loss provides fundamental insights into the etiology of developmental disorders and cancer and has the potential, in the long term, to identify new therapeutic strategies. Video Abstract.


Asunto(s)
Desarrollo Embrionario , Neoplasias/metabolismo , Neurofibromina 2 , Animales , Humanos , Neurofibromina 2/deficiencia , Neurofibromina 2/fisiología , Transducción de Señal
5.
Dev Biol ; 442(2): 301-314, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30118662

RESUMEN

Disruption of endometrial gland formation or function can cause female infertility. Formation of endometrial glands via tubulogenesis of luminal epithelial cells requires the establishment and maintenance of cell polarity and cell adhesion. The FERM domain-containing protein Merlin coordinates epithelial cell polarity and cell adhesion and is critical for epithelial tissue function in the skin and kidney. We now demonstrate a requirement for Merlin in endometrial gland development. Conditional deletion of Merlin in the endometrium results in female infertility caused by the absence of gland formation. Interestingly, we observed glandular epithelial markers within discrete groups of cells in the Merlin-deficient luminal epithelium. Wnt signaling, a pathway necessary for endometrial gland development is maintained in Merlin-deficient endometrium, suggesting the glandular fate program is active. Instead, we observe increased levels of apical actin and markers indicative of high membrane tension on the basal surface of the Merlin-deficient luminal epithelium. These findings suggest that the structural integrity of the luminal epithelium during gland formation is required for appropriate endometrial tubulogenesis and tissue function. Moreover, our work implicates Merlin-dependent regulation of mechanical tension in the proper formation of endometrial gland architecture and function.


Asunto(s)
Endometrio/crecimiento & desarrollo , Neurofibromina 2/fisiología , Animales , Adhesión Celular/fisiología , Polaridad Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Endometrio/citología , Endometrio/metabolismo , Endometrio/patología , Células Epiteliales/metabolismo , Femenino , Hibridación in Situ , Infertilidad Femenina , Ratones , Ratones Endogámicos C57BL , Morfogénesis/fisiología , Neurofibromina 2/deficiencia , Neurofibromina 2/genética , Neurofibromina 2/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología
6.
Hepatology ; 62(4): 1227-36, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26108984

RESUMEN

UNLABELLED: The intercellular adhesion molecule 1 (ICAM-1) is induced in mouse liver after bile duct ligation (BDL) and plays a key role in neutrophil-mediated liver injury in BDL mice. ICAM-1 has been shown to interact with cytoskeletal ezrin-radixin-moesin (ERM) proteins that also interact with the PDZ protein, Na(+) /H(+) exchanger regulatory factor 1 (NHERF-1/EBP50). In NHERF-1(-/-) mice, ERM proteins are significantly reduced in brush-border membranes from kidney and small intestine. ERM knockdown reduces ICAM-1 expression in response to tumor necrosis factor alpha. Here we show that NHERF-1 assembles ERM proteins, ICAM-1 and F-actin into a macromolecule complex that is increased in mouse liver after BDL. Compared to wild-type (WT) mice, both sham-operated and BDL NHERF-1(-/-) mice have lower levels of activated ERM and ICAM-1 protein in the liver accompanied by significantly reduced hepatic neutrophil accumulation, serum alanine aminotransferase, and attenuated liver injury after BDL. However, total bile acid concentrations in serum and liver of sham and BDL NHERF-1(-/-) mice were not significantly different from WT controls, although hepatic tetrahydroxylated bile acids and Cyp3a11 messenger RNA levels were higher in NHERF-1(-/-) BDL mice. CONCLUSION: NHERF-1 participates in the inflammatory response that is associated with BDL-induced liver injury. Deletion of NHERF-1 in mice leads to disruption of the formation of ICAM-1/ERM/NHERF-1 complex and reduction of hepatic ERM proteins and ICAM-1, molecules that are up-regulated and are essential for neutrophil-mediated liver injury in cholestasis. Further study of the role of NHERF-1 in the inflammatory response in cholestasis and other forms of liver injury should lead to discovery of new therapeutic targets in hepatic inflammatory diseases.


Asunto(s)
Colestasis Intrahepática/etiología , Molécula 1 de Adhesión Intercelular/fisiología , Hepatopatías/etiología , Fosfoproteínas/fisiología , Intercambiadores de Sodio-Hidrógeno/fisiología , Animales , Hepatitis/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neurofibromina 2/fisiología , Fosfoproteínas/genética , Intercambiadores de Sodio-Hidrógeno/genética
7.
Brain ; 137(Pt 2): 420-32, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24309211

RESUMEN

Axonal surface proteins encompass a group of heterogeneous molecules, which exert a variety of different functions in the highly interdependent relationship between axons and Schwann cells. We recently revealed that the tumour suppressor protein merlin, mutated in the hereditary tumour syndrome neurofibromatosis type 2, impacts significantly on axon structure maintenance in the peripheral nervous system. We now report on a role of neuronal merlin in the regulation of the axonal surface protein neuregulin 1 important for modulating Schwann cell differentiation and myelination. Specifically, neuregulin 1 type III expression is reduced in sciatic nerve tissue of neuron-specific knockout animals as well as in biopsies from seven patients with neurofibromatosis type 2. In vitro experiments performed on both the P19 neuronal cell line and primary dorsal root ganglion cells demonstrate the influence of merlin on neuregulin 1 type III expression. Moreover, expression of ERBB2, a Schwann cell receptor for neuregulin 1 ligands is increased in nerve tissue of both neuron-specific merlin knockout animals and patients with neurofibromatosis type 2, demonstrating for the first time that axonal merlin indirectly regulates Schwann cell behaviour. Collectively, we have identified that neuronally expressed merlin can influence Schwann cell activity in a cell-extrinsic manner.


Asunto(s)
Neurregulina-1/fisiología , Neurofibromina 2/fisiología , Neuronas/fisiología , Receptor ErbB-2/biosíntesis , Células de Schwann/metabolismo , Transducción de Señal/fisiología , Adulto , Anciano , Animales , Línea Celular , Células Cultivadas , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Neurofibromatosis 2/metabolismo , Neurofibromatosis 2/patología , Neuronas/patología , Células de Schwann/patología
8.
Biochim Biophys Acta ; 1826(2): 400-6, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22750751

RESUMEN

Neurofibromatosis type 2 (NF2), characterized by tumors of the nervous system, is a result of functional loss of the NF2 gene. The NF2 gene encodes Merlin (moesin-ezrin-radixin-like protein), an ERM (Ezrin, Radixin, Moesin) protein family member. Merlin functions as a tumor suppressor through impacting mechanisms related to proliferation, apoptosis, survival, motility, adhesion, and invasion. Several studies have summarized the tumor intrinsic mutations in Merlin. Given the fact that tumor cells are not in isolation, but rather in an intricate, mutually sustaining synergy with their surrounding stroma, the dialog between the tumor cells and the stroma can potentially impact the molecular homeostasis and promote evolution of the malignant phenotype. This review summarizes the epigenetic modifications, transcript stability, and post-translational modifications that impact Merlin. We have reviewed the role of extrinsic factors originating from the tumor milieu that influence the availability of Merlin inside the cell. Information regarding Merlin regulation could lead to novel therapeutics by stabilizing Merlin protein in tumors that have reduced Merlin protein expression without displaying any NF2 genetic alterations.


Asunto(s)
Neurofibromina 2/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Epigénesis Genética , Humanos , Neurofibromina 2/química , Proteínas Proto-Oncogénicas c-akt/fisiología , Estabilidad del ARN
9.
Proc Natl Acad Sci U S A ; 107(16): 7311-6, 2010 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-20368450

RESUMEN

The neurofibromatosis type 2 (NF2) tumor-suppressor protein Merlin is a member of the ERM family of proteins that links the cytoskeleton to the plasma membrane. In humans, mutations in the NF2 gene cause neurofibromatosis type-2 (NF2), a cancer syndrome characterized by the development of tumors of the nervous system. Previous reports have suggested that the subcellular distribution of Merlin is critical to its function, and that several NF2 mutants that lack tumor-suppressor activity present improper localization. Here we used a Drosophila cell culture model to study the distribution and mechanism of intracellular transport of Merlin and its mutants. We found that Drosophila Merlin formed cytoplasmic particles that move bidirectionally along microtubules. A single NF2-causing amino acid substitution in the FERM domain dramatically inhibited Merlin particle movement. Surprisingly, the presence of this immotile Merlin mutant also inhibited trafficking of the WT protein. Analysis of the movement of WT protein using RNAi and pull-downs showed that Merlin particles are associated with and moved by microtubule motors (kinesin-1 and cytoplasmic dynein), and that binding of motors and movement is regulated by Merlin phosphorylation. Inhibition of Merlin transport by expression of the dominant-negative mutant or depletion of kinesin-1 results in increased nuclear accumulation of the transcriptional coactivator Yorkie. These results demonstrate the requirement of microtubule-dependent transport for Merlin function.


Asunto(s)
Drosophila melanogaster/metabolismo , Dineínas/metabolismo , Microtúbulos/metabolismo , Neurofibromina 2/metabolismo , Neurofibromina 2/fisiología , Animales , Transporte Biológico , Concanavalina A/metabolismo , Citoplasma/metabolismo , Proteínas de Drosophila/metabolismo , Endocitosis , Proteínas Fluorescentes Verdes/metabolismo , Cinesinas/metabolismo , Modelos Biológicos , Proteínas Nucleares/metabolismo , Fosforilación , Estructura Terciaria de Proteína , Transactivadores/metabolismo , Proteínas Señalizadoras YAP
10.
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
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.
Pathol Int ; 61(6): 331-44, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21615608

RESUMEN

Malignant mesothelioma (MM) is an aggressive tumor arising primarily from pleural or peritoneal cavities, which is caused by asbestos exposure after long latency. One of the most frequently mutated genes detected in MM cells is the neurofibromatosis type 2 (NF2) tumor suppressor gene which is located at chromosome 22q12. The NF2 gene encodes Merlin, an ERM (Ezrin/Radixin/Moesin) protein. The underphosphorylated form of Merlin is active and acts as a tumor suppressor by regulating several distinct cellular signaling pathways. One of the downstream pathways regulated by Merlin is the Hippo signaling pathway, which is conserved from Drosophila to mammalian cells and plays important roles in organ size control and cancer development. Recent studies have identified alterations of the components in the Hippo signaling cascade in MM cells, including overexpression of Yes-associated protein (YAP) and inactivation of large tumor suppressor homolog 2 (LATS2). Dysregulation of the Merlin-Hippo signaling cascade is one of the frequent and key events of MM cell development and/or progression. Thus, a strategy to normalize this signaling cascade may be the rationale for developing a new target therapy against MM.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/fisiología , Mesotelioma/genética , Neurofibromina 2/fisiología , Neoplasias Pleurales/genética , Transducción de Señal/fisiología , Animales , Línea Celular Tumoral , Cromosomas Humanos Par 22 , Modelos Animales de Enfermedad , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Genes Supresores de Tumor , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mesotelioma/patología , Mutación , Neurofibromina 2/química , Neurofibromina 2/genética , Fosforilación , Neoplasias Pleurales/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Supresoras de Tumor/genética
13.
Biochim Biophys Acta ; 1788(4): 755-60, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18952051

RESUMEN

Recent studies have revealed unexpected links between cell polarity and proliferation, suggesting that the polarized organization of cells is necessary to regulate growth. Drosophila melanogaster is a genetically simple model that is especially suited for the study of polarity and growth control, as polarized tissues undergo a well-defined pattern of proliferation and differentiation during the development. In addition, genetic studies have identified a number of tumor suppressor genes, which later studies have shown to be associated with junctions, or in the regulation of junctional proteins. We will explore in this review the links between growth and apical junction proteins in the regulation of growth control in Drosophila.


Asunto(s)
Uniones Adherentes/fisiología , Drosophila melanogaster/crecimiento & desarrollo , Animales , Moléculas de Adhesión Celular/fisiología , Proteínas de Drosophila/fisiología , Receptores ErbB/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas de la Membrana/fisiología , Neurofibromina 2/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Transducción de Señal/fisiología
14.
J Cell Biol ; 171(2): 361-71, 2005 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-16247032

RESUMEN

Introduction of activated p21-activated kinase (PAK) is sufficient to release primary endothelial cells from contact inhibition of growth. Confluent cells display deficient activation of PAK and translocation of Rac to the plasma membrane at matrix adhesions. Targeting Rac to the plasma membrane rescues these cells from contact inhibition. PAK's ability to release human umbilical vein endothelial cells from contact inhibition is blocked by an unphosphorylatable form of its target Merlin, suggesting that PAK promotes mitogenesis by phosphorylating, and thus inactivating, Merlin. Merlin mutants, which are presumed to exert a dominant-negative effect, enable recruitment of Rac to matrix adhesions and promote mitogenesis in confluent cells. Small interference RNA-mediated knockdown of Merlin exerts the same effects. Dominant-negative Rac blocks PAK-mediated release from contact inhibition, implying that PAK functions upstream of Rac in this signaling pathway. These results provide a framework for understanding the tumor suppressor function of Merlin and indicate that Merlin mediates contact inhibition of growth by suppressing recruitment of Rac to matrix adhesions.


Asunto(s)
Membrana Celular/metabolismo , Inhibición de Contacto/fisiología , Neurofibromina 2/fisiología , Proteínas de Unión al GTP rac/metabolismo , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Membrana Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Humanos , Neurofibromina 2/antagonistas & inhibidores , Fosforilación , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , Quinasas p21 Activadas , Proteínas de Unión al GTP rac/efectos de los fármacos
15.
Sci Rep ; 10(1): 4211, 2020 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-32144278

RESUMEN

Neurofibromatosis type 2 (NF2) is an inherited disorder characterized by bilateral vestibular schwannomas (VS) that arise from neoplastic Schwann cells (SCs). NF2-associated VSs are often accompanied by meningioma (MN), and the majority of NF2 patients show loss of the NF2 tumor suppressor. mTORC1 and mTORC2-specific serum/glucocorticoid-regulated kinase 1 (SGK1) are constitutively activated in MN with loss of NF2. In a recent high-throughput kinome screen in NF2-null human arachnoidal and meningioma cells, we showed activation of EPH RTKs, c-KIT, and SFK members independent of mTORC1/2 activation. Subsequently, we demonstrated in vitro and in vivo efficacy of combination therapy with the dual mTORC1/2 inhibitor AZD2014 and the multi-kinase inhibitor dasatinib. For these reasons, we investigated activated mTORC1/2 and EPH receptor-mediated signaling in sporadic and NF2-associated VS. Using primary human VS cells and a mouse allograft model of schwannoma, we evaluated the dual mTORC1/2 inhibitor AZD2014 and the tyrosine kinase inhibitor dasatinib as monotherapies and in combination. Escalating dose-response experiments on primary VS cells grown from 15 human tumors show that combination therapy with AZD2014 and dasatinib is more effective at reducing metabolic activity than either drug alone and exhibits a therapeutic effect at a physiologically reasonable concentration (~0.1 µM). In vivo, while AZD2014 and dasatinib each inhibit tumor growth alone, the effect of combination therapy exceeds that of either drug. Co-targeting the mTOR and EPH receptor pathways with these or similar compounds may constitute a novel therapeutic strategy for VS, a condition for which there is no FDA-approved pharmacotherapy.


Asunto(s)
Benzamidas/farmacología , Dasatinib/farmacología , Modelos Animales de Enfermedad , Morfolinas/farmacología , Neurofibromina 2/fisiología , Neuroma Acústico/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Quimioterapia Combinada , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neurofibromina 2/genética , Neurofibromina 2/metabolismo , Neuroma Acústico/metabolismo , Neuroma Acústico/patología , Receptor EphA1/metabolismo
16.
J Neurosci ; 28(42): 10472-81, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18923024

RESUMEN

Schwannomin/merlin is the product of a tumor suppressor gene mutated in neurofibromatosis type 2 (NF2). Although the consequences of NF2 mutations on Schwann cell proliferation are well established, the physiological role of schwannomin in differentiated cells is not known. To unravel this role, we studied peripheral nerves in mice overexpressing in Schwann cells schwannomin with a deletion occurring in NF2 patients (P0-SCH-Delta39-121) or a C-terminal deletion. The myelin sheath and nodes of Ranvier were essentially preserved in both lines. In contrast, the ultrastructural and molecular organization of contacts between Schwann cells and axons in paranodal and juxtaparanodal regions were altered, with irregular juxtaposition of normal and abnormal areas of contact. Similar but more severe alterations were observed in mice with conditional deletion of the Nf2 gene in Schwann cells. The number of Schmidt-Lanterman incisures, which are cytoplasmic channels interrupting the compact myelin and characterized by distinct autotypic contacts, was increased in the three mutant lines. P0-SCH-Delta39-121 and conditionally deleted mice displayed exuberant wrapping of nonmyelinated fibers and short internodes, an abnormality possibly related to altered control of Schwann cell proliferation. In support of this hypothesis, Schwann cell number was increased along fibers before myelination in P0-SCH-Delta39-121 mice but not in those with C-terminal deletion. Schwann cell numbers were also more numerous in mice with conditional deletion. Thus, schwannomin plays an important role in the control of Schwann cell number and is necessary for the correct organization and regulation of axoglial heterotypic and glio-glial autotypic contacts.


Asunto(s)
Comunicación Celular/fisiología , Neurofibromina 2/fisiología , Células de Schwann/metabolismo , Células de Schwann/ultraestructura , Proteínas Supresoras de Tumor/fisiología , Animales , Proliferación Celular , Eliminación de Gen , Humanos , Ratones , Ratones Transgénicos , Neurofibromina 2/biosíntesis , Neurofibromina 2/deficiencia , Neurofibromina 2/genética , Nervios Periféricos/metabolismo , Nervios Periféricos/ultraestructura , Proteínas Supresoras de Tumor/genética
17.
Curr Biol ; 16(7): 702-9, 2006 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-16581517

RESUMEN

The precise coordination of signals that control proliferation is a key feature of growth regulation in developing tissues . While much has been learned about the basic components of signal transduction pathways, less is known about how receptor localization, compartmentalization, and trafficking affect signaling in developing tissues. Here we examine the mechanism by which the Drosophila Neurofibromatosis 2 (NF2) tumor suppressor ortholog Merlin (Mer) and the related tumor suppressor expanded (ex) regulate proliferation and differentiation in imaginal epithelia. Merlin and Expanded are members of the FERM (Four-point one, Ezrin, Radixin, Moesin) domain superfamily, which consists of membrane-associated cytoplasmic proteins that interact with transmembrane proteins and may function as adapters that link to protein complexes and/or the cytoskeleton . We demonstrate that Merlin and Expanded function to regulate the steady-state levels of signaling and adhesion receptors and that loss of these proteins can cause hyperactivation of associated signaling pathways. In addition, pulse-chase labeling of Notch in living tissues indicates that receptor levels are upregulated at the plasma membrane in Mer; ex double mutant cells due to a defect in receptor clearance from the cell surface. We propose that these proteins control proliferation by regulating the abundance, localization, and turnover of cell-surface receptors and that misregulation of these processes may be a key component of tumorigenesis.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila/metabolismo , Endocitosis/fisiología , Proteínas de la Membrana/fisiología , Neurofibromina 2/fisiología , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Animales , Membrana Celular/metabolismo , Proliferación Celular , Drosophila/anatomía & histología , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Glicoproteínas de Membrana , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación , Neurofibromina 2/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria , Transporte de Proteínas/fisiología , Receptores de Superficie Celular/genética , Receptores Notch/metabolismo , Regulación hacia Arriba , Alas de Animales/anatomía & histología , Alas de Animales/metabolismo
18.
Brain ; 131(Pt 3): 606-15, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17940085

RESUMEN

Alterations in the NF2 gene coding for merlin cause all tumours that occur in patients suffering from neurofibromatosis type 2, all spontaneous schwannomas and the majority of meningiomas. Thus merlin's tumours are quite frequent and also numerous when inherited as part of meurofibromatosis type 2. Tumours caused by mutations in the NF2 gene are benign and thus do not respond to classical chemotherapy. Surgery and radiosurgery are only local therapies and the patients frequently require multiple treatments. This highlights the medical need to understand how merlin loss results in tumourigenesis and the need to find new systemic therapies. The benign, and therefore genetically stable and homogenous character of the tumours allows establishment of meaningful tumour models. This brings about the rather unique opportunity to both analyse the consequences of the gene defect and identify new therapeutic targets. In this review, I will first describe the phenotypes associated with 'merlin' mutations and consider differential diagnosis, in particular Schwannomatosis, for which a gene defect has been described recently. Existing therapeutic options, surgery and radiosurgery, including new data on the latter will be reviewed. Finally, I will discuss how loss of merlin leads to tumourigenesis in order to understand the rationale for emerging new therapeutic targets.


Asunto(s)
Mutación , Neurofibromatosis 2/genética , Neurofibromina 2/genética , Adulto , Diagnóstico Diferencial , Ependimoma/diagnóstico , Ependimoma/genética , Ependimoma/terapia , Humanos , Meningioma/diagnóstico , Meningioma/genética , Meningioma/terapia , Persona de Mediana Edad , Neurilemoma/diagnóstico , Neurilemoma/genética , Neurilemoma/terapia , Neurofibromatosis 2/diagnóstico , Neurofibromatosis 2/terapia , Neurofibromina 2/fisiología
19.
Oncogene ; 38(36): 6370-6381, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31312020

RESUMEN

Inactivation of the tumor suppressor NF2/merlin underlies neurofibromatosis type 2 (NF2) and some sporadic tumors. Previous studies have established that merlin mediates contact inhibition of proliferation; however, the exact mechanisms remain obscure and multiple pathways have been implicated. We have previously reported that merlin inhibits Ras and Rac activity during contact inhibition, but how merlin regulates Ras activity has remained elusive. Here we demonstrate that merlin can directly interact with both Ras and p120RasGAP (also named RasGAP). While merlin does not increase the catalytic activity of RasGAP, the interactions with Ras and RasGAP may fine-tune Ras signaling. In vivo, loss of RasGAP in Schwann cells, unlike the loss of merlin, failed to promote tumorigenic growth in an orthotopic model. Therefore, modulation of Ras signaling through RasGAP likely contributes to, but is not sufficient to account for, merlin's tumor suppressor activity. Our study provides new insight into the mechanisms of merlin-dependent Ras regulation and may have additional implications for merlin-dependent regulation of other small GTPases.


Asunto(s)
Neurofibromina 2/fisiología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Animales , Células Cultivadas , Proteínas Activadoras de GTPasa/metabolismo , Genes Supresores de Tumor , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neurofibromatosis 2/genética , Neurofibromatosis 2/metabolismo , Neurofibromina 2/metabolismo , Unión Proteica , Transducción de Señal/genética
20.
Carcinogenesis ; 29(11): 2139-46, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18725387

RESUMEN

We previously reported the results of bacterial artificial chromosome array comprehensive genomic hybridization of malignant pleural mesotheliomas (MPMs), including two cases with high-level amplification in the 11q22 locus. In this study, we found that the YAP1 gene encoding a transcriptional coactivator was localized in this amplified region and overexpressed in both cases, suggesting it as a candidate oncogene in this region. We analyzed the involvement of YAP1 in MPM proliferation, as well as its functional and physical interaction with Merlin encoded by the neurofibromatosis type 2 (NF2) tumor suppressor gene, which is frequently mutated in MPMs. YAP1-RNA interference suppressed growth of a mesothelioma cell line NCI-H290 with NF2 homozygous deletion, probably through cell-cycle arrest and apoptosis induction, whereas YAP1 transfection promoted the growth of MeT-5A, an immortalized mesothelial cell line. We also found that the introduction of NF2 into NCI-H290 induced phosphorylation at serine 127 of YAP1, which was accompanied by reduction of nuclear localization of YAP1, whereas nuclear localization of a YAP1 S 127A mutant was not affected. Furthermore, results of immunoprecipitation and in vitro pull-down assays indicated a physical interaction between Merlin and YAP1. These results suggest that YAP1 is involved in mesothelial cell growth and that the transcriptional coactivator activity of YAP1 is functionally inhibited by Merlin through the induction of phosphorylation and cytoplasmic retention of YAP1. This is the first report of negative regulatory signaling from Merlin to YAP1 in mammalian cells. Future studies of transcriptional targets of YAP1 in MPMs may shed light on the molecular mechanisms of MPM development and lead to new therapeutic strategies.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Mesotelioma/patología , Neurofibromina 2/fisiología , Fosfoproteínas/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencia de Bases , Proliferación Celular , Mapeo Cromosómico , Cromosomas Artificiales Bacterianos , Cromosomas Humanos Par 11 , Cartilla de ADN , Citometría de Flujo , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Mesotelioma/genética , Mesotelioma/fisiopatología , Conformación de Ácido Nucleico , Fosfoproteínas/genética , Fosforilación , Reacción en Cadena de la Polimerasa , Interferencia de ARN , Factores de Transcripción , Ubiquitina-Proteína Ligasas , Proteínas Señalizadoras YAP
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda