Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 1559, 2023 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-36944680

RESUMEN

Schwannomas are common sporadic tumors and hallmarks of familial neurofibromatosis type 2 (NF2) that develop predominantly on cranial and spinal nerves. Virtually all schwannomas result from inactivation of the NF2 tumor suppressor gene with few, if any, cooperating mutations. Despite their genetic uniformity schwannomas exhibit remarkable clinical and therapeutic heterogeneity, which has impeded successful treatment. How heterogeneity develops in NF2-mutant schwannomas is unknown. We have found that loss of the membrane:cytoskeleton-associated NF2 tumor suppressor, merlin, yields unstable intrinsic polarity and enables Nf2-/- Schwann cells to adopt distinct programs of ErbB ligand production and polarized signaling, suggesting a self-generated model of schwannoma heterogeneity. We validated the heterogeneous distribution of biomarkers of these programs in human schwannoma and exploited the synchronous development of lesions in a mouse model to establish a quantitative pipeline for studying how schwannoma heterogeneity evolves. Our studies highlight the importance of intrinsic mechanisms of heterogeneity across human cancers.


Asunto(s)
Neurilemoma , Neurofibromatosis 2 , Animales , Ratones , Humanos , Neurofibromatosis 2/genética , Neurilemoma/genética , Neurilemoma/patología , Neurofibromina 2/genética , Mutación , Células de Schwann/patología , Genes Supresores de Tumor
2.
J Cell Biol ; 220(6)2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-33999116

RESUMEN

EPH/EPHRIN signaling is crucial to the segregation of cell populations during the morphogenesis of many tissues. In this issue, Kindberg et al. (2021. J. Cell Biol.https://doi.org/10.1083/jcb.202005216) show that EPH activation can drive both heterotypic cell repulsion and homotypic aggregation by triggering increased cortical tension.


Asunto(s)
Efrinas , Transducción de Señal , Efrinas/metabolismo , Morfogénesis , Unión Proteica , Transporte de Proteínas
3.
Nat Commun ; 11(1): 3377, 2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32632100

RESUMEN

The mammary gland is a highly vascularized tissue capable of expansion and regression during development and disease. To enable mechanistic insight into the coordinated morphogenic crosstalk between the epithelium and vasculature, we introduce a 3D microfluidic platform that juxtaposes a human mammary duct in proximity to a perfused endothelial vessel. Both compartments recapitulate stable architectural features of native tissue and the ability to undergo distinct forms of branching morphogenesis. Modeling HER2/ERBB2 amplification or activating PIK3CA(H1047R) mutation each produces ductal changes observed in invasive progression, yet with striking morphogenic and behavioral differences. Interestingly, PI3KαH1047R ducts also elicit increased permeability and structural disorganization of the endothelium, and we identify the distinct secretion of IL-6 as the paracrine cause of PI3KαH1047R-associated vascular dysfunction. These results demonstrate the functionality of a model system that facilitates the dissection of 3D morphogenic behaviors and bidirectional signaling between mammary epithelium and endothelium during homeostasis and pathogenesis.


Asunto(s)
Glándulas Mamarias Humanas/metabolismo , Morfogénesis/genética , Mutación , Comunicación Paracrina/genética , Biomimética/métodos , Línea Celular , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Endotelio Vascular/crecimiento & desarrollo , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Femenino , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Humanos , Glándulas Mamarias Humanas/irrigación sanguínea , Glándulas Mamarias Humanas/crecimiento & desarrollo , Fenotipo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo
4.
Dev Cell ; 47(6): 693-695, 2018 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-30562513

RESUMEN

The transcriptional activators YAP and TAZ integrate mechanical and biochemical signals to support growth and regeneration. A recent study in Nature (Chang et al., 2018) showed that SWI/SNF sequesters YAP from its DNA-binding partner TEAD. Mechanical coupling between cytoskeleton and nucleus reverses this antagonism by nuclear F-actin sequestering of SWI/SNF.


Asunto(s)
Actinas , Factores de Transcripción , Citoesqueleto de Actina , Núcleo Celular , Citoesqueleto
5.
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
6.
Development ; 145(9)2018 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-29712669

RESUMEN

The architecture of individual cells and cell collectives enables functional specification, a prominent example being the formation of epithelial tubes that transport fluid or gas in many organs. The intrahepatic bile ducts (IHBDs) form a tubular network within the liver parenchyma that transports bile to the intestine. Aberrant biliary 'neoductulogenesis' is also a feature of several liver pathologies including tumorigenesis. However, the mechanism of biliary tube morphogenesis in development or disease is not known. Elimination of the neurofibromatosis type 2 protein (NF2; also known as merlin or neurofibromin 2) causes hepatomegaly due to massive biliary neoductulogenesis in the mouse liver. We show that this phenotype reflects unlimited biliary morphogenesis rather than proliferative expansion. Our studies suggest that NF2 normally limits biliary morphogenesis by coordinating lumen expansion and cell architecture. This work provides fundamental insight into how biliary fate and tubulogenesis are coordinated during development and will guide analyses of disease-associated and experimentally induced biliary pathologies.


Asunto(s)
Conductos Biliares Intrahepáticos/embriología , Proliferación Celular/fisiología , Neurofibromina 2/metabolismo , Organogénesis/fisiología , Animales , Conductos Biliares Intrahepáticos/patología , Eliminación de Gen , Hepatomegalia/embriología , Hepatomegalia/genética , Hepatomegalia/patología , Ratones , Ratones Noqueados , Neurofibromina 2/genética
7.
Sci Signal ; 11(515)2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29382786

RESUMEN

Cytoskeletal networks are dramatically reorganized upon EGF stimulation to enable complex cell behaviors such as cell-cell communication, migration and invasion, and cell division. In this issue of Science Signaling, Roth et al. and Pike et al. use proteomic methods to identify several effectors of EGF responses. The findings show the interdependent nature of growth factor signaling and the cytoskeleton and identify potential new therapeutic targets for treating cancer and other growth factor-driven diseases.


Asunto(s)
Proliferación Celular , Citoesqueleto/metabolismo , Neoplasias/patología , Proteoma/metabolismo , Animales , Citoesqueleto/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo
8.
Artículo en Inglés | MEDLINE | ID: mdl-28716887

RESUMEN

The behavior of cells within tissues is governed by the activities of adhesion receptors that provide spatial cues and transmit forces through intercellular junctions, and by growth-factor receptors, particularly receptor tyrosine kinases (RTKs), that respond to biochemical signals from the environment. Coordination of these two activities is essential for the patterning and polarized migration of cells during morphogenesis and for homeostasis in mature tissues; loss of this coordination is a hallmark of developing cancer and driver of metastatic progression. Although much is known about the individual functions of adhesion and growth factor receptors, we have a surprisingly superficial understanding of the mechanisms by which their activities are coordinated.


Asunto(s)
Comunicación Celular/fisiología , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal/fisiología , Animales , Movimiento Celular/fisiología , Homeostasis , Humanos , Uniones Intercelulares/fisiología , Proteínas Tirosina Quinasas/genética
9.
J Cell Biol ; 211(2): 391-405, 2015 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-26483553

RESUMEN

The proliferation of normal cells is inhibited at confluence, but the molecular basis of this phenomenon, known as contact-dependent inhibition of proliferation, is unclear. We previously identified the neurofibromatosis type 2 (NF2) tumor suppressor Merlin as a critical mediator of contact-dependent inhibition of proliferation and specifically found that Merlin inhibits the internalization of, and signaling from, the epidermal growth factor receptor (EGFR) in response to cell contact. Merlin is closely related to the membrane-cytoskeleton linking proteins Ezrin, Radixin, and Moesin, and localization of Merlin to the cortical cytoskeleton is required for contact-dependent regulation of EGFR. We show that Merlin and Ezrin are essential components of a mechanism whereby mechanical forces associated with the establishment of cell-cell junctions are transduced across the cell cortex via the cortical actomyosin cytoskeleton to control the lateral mobility and activity of EGFR, providing novel insight into how cells inhibit mitogenic signaling in response to cell contact.


Asunto(s)
Actomiosina/metabolismo , Inhibición de Contacto/fisiología , Proteínas del Citoesqueleto/metabolismo , Receptores ErbB/metabolismo , Neurofibromina 2/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Proliferación Celular/fisiología , Células Cultivadas , Inhibición de Contacto/genética , Proteínas del Citoesqueleto/genética , Uniones Intercelulares/fisiología , Mecanotransducción Celular/fisiología , Proteínas de la Membrana/metabolismo , Ratones , Proteínas de Microfilamentos/metabolismo , Neurofibromina 2/genética , Miosina Tipo IIA no Muscular/metabolismo , Transporte de Proteínas , Interferencia de ARN , ARN Interferente Pequeño , Estrés Mecánico
10.
Cell Rep ; 10(10): 1692-1707, 2015 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-25772357

RESUMEN

Defective Hippo/YAP signaling in the liver results in tissue overgrowth and development of hepatocellular carcinoma (HCC). Here, we uncover mechanisms of YAP-mediated hepatocyte reprogramming and HCC pathogenesis. YAP functions as a rheostat in maintaining metabolic specialization, differentiation, and quiescence within the hepatocyte compartment. Increased or decreased YAP activity reprograms subsets of hepatocytes to different fates associated with deregulation of the HNF4A, CTNNB1, and E2F transcriptional programs that control hepatocyte quiescence and differentiation. Importantly, treatment with small interfering RNA-lipid nanoparticles (siRNA-LNPs) targeting YAP restores hepatocyte differentiation and causes pronounced tumor regression in a genetically engineered mouse HCC model. Furthermore, YAP targets are enriched in an aggressive human HCC subtype characterized by a proliferative signature and absence of CTNNB1 mutations. Thus, our work reveals Hippo signaling as a key regulator of the positional identity of hepatocytes, supports targeting of YAP using siRNA-LNPs as a paradigm of differentiation-based therapy, and identifies an HCC subtype that is potentially responsive to this approach.

12.
J Cell Sci ; 127(Pt 15): 3199-204, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24951115

RESUMEN

The cell cortex is a dynamic and heterogeneous structure that governs cell identity and behavior. The ERM proteins (ezrin, radixin and moesin) are major architects of the cell cortex, and they link plasma membrane phospholipids and proteins to the underlying cortical actin cytoskeleton. Recent studies in several model systems have uncovered surprisingly dynamic and complex molecular activities of the ERM proteins and have provided new mechanistic insight into how they build and maintain cortical domains. Among many well-established and essential functions of ERM proteins, this Cell Science at a Glance article and accompanying poster will focus on the role of ERMs in organizing the cell cortex during cell division and apical morphogenesis. These examples highlight an emerging appreciation that the ERM proteins both locally alter the mechanical properties of the cell cortex, and control the spatial distribution and activity of key membrane complexes, establishing the ERM proteins as a nexus for the physical and functional organization of the cell cortex and making it clear that they are much more than scaffolds. This article is part of a Minifocus on Establishing polarity.


Asunto(s)
Membrana Basal/metabolismo , Polaridad Celular , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/fisiología , Proteínas Activadoras de GTPasa/metabolismo , Integrinas/metabolismo , Neoplasias/fisiopatología , Factores de Transcripción/metabolismo , Animales , Humanos
13.
Sci Transl Med ; 6(237): 237ra68, 2014 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-24848258

RESUMEN

The goal of targeted therapy is to match a selective drug with a genetic lesion that predicts for drug sensitivity. In a diverse panel of cancer cell lines, we found that the cells most sensitive to focal adhesion kinase (FAK) inhibition lack expression of the neurofibromatosis type 2 (NF2) tumor suppressor gene product, Merlin. Merlin expression is often lost in malignant pleural mesothelioma (MPM), an asbestos-induced aggressive cancer with limited treatment options. Our data demonstrate that low Merlin expression predicts for increased sensitivity of MPM cells to a FAK inhibitor, VS-4718, in vitro and in tumor xenograft models. Disruption of MPM cell-cell or cell-extracellular matrix (ECM) contacts with blocking antibodies suggests that weak cell-cell adhesions in Merlin-negative MPM cells underlie their greater dependence on cell-ECM-induced FAK signaling. This provides one explanation of why Merlin-negative cells are vulnerable to FAK inhibitor treatment. Furthermore, we validated aldehyde dehydrogenase as a marker of cancer stem cells (CSCs) in MPM, a cell population thought to mediate tumor relapse after chemotherapy. Whereas pemetrexed and cisplatin, standard-of-care agents for MPM, enrich for CSCs, FAK inhibitor treatment preferentially eliminates these cells. These preclinical results provide the rationale for a clinical trial in MPM patients using a FAK inhibitor as a single agent after first-line chemotherapy. With this design, the FAK inhibitor could potentially induce a more durable clinical response through reduction of CSCs along with a strong antitumor effect. Furthermore, our data suggest that patients with Merlin-negative tumors may especially benefit from FAK inhibitor treatment.


Asunto(s)
Antineoplásicos/farmacología , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Mesotelioma/tratamiento farmacológico , Neurofibromina 2/deficiencia , Inhibidores de Proteínas Quinasas/farmacología , Aldehído Deshidrogenasa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Mesotelioma/enzimología , Mesotelioma/genética , Mesotelioma/patología , Mesotelioma Maligno , Ratones , Terapia Molecular Dirigida , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/enzimología , Neurofibromina 2/genética , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Am J Med Genet A ; 164A(3): 563-78, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24443315

RESUMEN

The neurofibromatoses (NF) are autosomal dominant genetic disorders that encompass the rare diseases NF1, NF2, and schwannomatosis. The NFs affect more people worldwide than Duchenne muscular dystrophy and Huntington's disease combined. NF1 and NF2 are caused by mutations of known tumor suppressor genes (NF1 and NF2, respectively). For schwannomatosis, although mutations in SMARCB1 were identified in a subpopulation of schwannomatosis patients, additional causative gene mutations are still to be discovered. Individuals with NF1 may demonstrate manifestations in multiple organ systems, including tumors of the nervous system, learning disabilities, and physical disfigurement. NF2 ultimately can cause deafness, cranial nerve deficits, and additional severe morbidities caused by tumors of the nervous system. Unmanageable pain is a key finding in patients with schwannomatosis. Although today there is no marketed treatment for NF-related tumors, a significant number of clinical trials have become available. In addition, significant preclinical efforts have led to a more rational selection of potential drug candidates for NF trials. An important element in fueling this progress is the sharing of knowledge. For over 20 years the Children's Tumor Foundation has convened an annual NF Conference, bringing together NF professionals to share novel findings, ideas, and build collaborations. The 2012 NF Conference held in New Orleans hosted over 350 NF researchers and clinicians. This article provides a synthesis of the highlights presented at the conference and as such, is a "state-of-the-field" for NF research in 2012.


Asunto(s)
Neurilemoma/etiología , Neurofibromatosis/etiología , Neurofibromatosis 1/etiología , Neurofibromatosis 2/etiología , Neoplasias Cutáneas/etiología , Humanos , Neurilemoma/genética , Neurilemoma/terapia , Neurofibromatosis/genética , Neurofibromatosis/terapia , Neurofibromatosis 1/genética , Neurofibromatosis 1/terapia , Neurofibromatosis 2/genética , Neurofibromatosis 2/terapia , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/terapia
15.
J Immunol ; 191(8): 4048-58, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24043890

RESUMEN

Ezrin is a member of the ezrin-radixin-moesin family of membrane-actin cytoskeleton cross-linkers that participate in a variety of cellular processes. In B cells, phosphorylation of ezrin at different sites regulates multiple processes, such as lipid raft coalescence, BCR diffusion, microclustering, and endosomal JNK activation. In this study, we generated mice with conditional deletion of ezrin in the B cell lineage to investigate the physiological significance of ezrin's function in Ag receptor-mediated B cell activation and humoral immunity. B cell development, as well as the proportion and numbers of major B cell subsets in peripheral lymphoid organs, was unaffected by the loss of ezrin. Using superresolution imaging methods, we show that, in the absence of ezrin, BCRs respond to Ag binding by accumulating into larger and more stable signaling microclusters. Loss of ezrin led to delayed BCR capping and accelerated lipid raft coalescence. Although proximal signaling proteins showed stronger activation in the absence of ezrin, components of the distal BCR signaling pathways displayed distinct effects. Ezrin deficiency resulted in increased B cell proliferation and differentiation into Ab-secreting cells ex vivo and stronger T cell-independent and -dependent responses to Ag in vivo. Overall, our data demonstrate that ezrin regulates amplification of BCR signals and tunes the strength of B cell activation and humoral immunity.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Linfocitos B/inmunología , Proteínas del Citoesqueleto/metabolismo , Inmunidad Humoral , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Citoesqueleto de Actina/inmunología , Citoesqueleto de Actina/metabolismo , Animales , Subgrupos de Linfocitos B/metabolismo , Linfocitos B/metabolismo , Diferenciación Celular/inmunología , Membrana Celular/inmunología , Membrana Celular/metabolismo , Proliferación Celular , Proteínas del Citoesqueleto/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Transducción de Señal/inmunología
16.
Genes Dev ; 26(24): 2709-23, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23249734

RESUMEN

The ability to generate asymmetry at the cell cortex underlies cell polarization and asymmetric cell division. Here we demonstrate a novel role for the tumor suppressor Merlin and closely related ERM proteins (Ezrin, Radixin, and Moesin) in generating cortical asymmetry in the absence of external cues. Our data reveal that Merlin functions to restrict the cortical distribution of the actin regulator Ezrin, which in turn positions the interphase centrosome in single epithelial cells and three-dimensional organotypic cultures. In the absence of Merlin, ectopic cortical Ezrin yields mispositioned centrosomes, misoriented spindles, and aberrant epithelial architecture. Furthermore, in tumor cells with centrosome amplification, the failure to restrict cortical Ezrin abolishes centrosome clustering, yielding multipolar mitoses. These data uncover fundamental roles for Merlin/ERM proteins in spatiotemporally organizing the cell cortex and suggest that Merlin's role in restricting cortical Ezrin may contribute to tumorigenesis by disrupting cell polarity, spindle orientation, and, potentially, genome stability.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Neurofibromina 2/metabolismo , Animales , Células CACO-2 , Ciclo Celular/fisiología , Línea Celular Tumoral , Polaridad Celular , Centrosoma/metabolismo , Proteínas del Citoesqueleto/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Humanos , Ratones , Neurofibromina 2/genética , Huso Acromático/metabolismo
18.
Curr Opin Cell Biol ; 24(5): 685-94, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22835462

RESUMEN

It has long been appreciated that proliferation of many cells is inhibited by density, a phenomenon that is often attributed to cell-cell contact. The basic properties of this phenomenon were established in the 1960s, along with the observation that such density-dependence was also lost in transformed cells. The mechanistic basis of contact inhibition of proliferation (CIP) has been slower to reveal itself. Here we discuss recent progress in elucidating the roles that cell-cell adhesion molecules play as receptors for CIP and in characterising the intracellular signaling pathways that mediate adhesion-dependent proliferative inhibition.


Asunto(s)
Proliferación Celular , Inhibición de Contacto/fisiología , Animales , Cadherinas/metabolismo , Cateninas/metabolismo , Adhesión Celular , Moléculas de Adhesión Celular/metabolismo , Citoesqueleto/metabolismo , Humanos , Mecanotransducción Celular , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Transducción de Señal
19.
Genes Dev ; 26(14): 1515-9, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22802525

RESUMEN

Mutations in the SPRED1 (Sprouty-related protein with an EVH [Ena/Vasp homology] domain 1) and NF1 (neurofibromatosis 1) genes underlie clinically related human disorders. The NF1-encoded protein neurofibromin is a Ras GTPase-activating protein (GAP) and can directly limit Ras activity. Spred proteins also negatively regulate Ras signaling, but the mechanism by which they do so is not clear. In the July 1, 2012, issue of Genes & Development, Stowe and colleagues (pp. 1421-1426) present evidence that Spred1 recruits neurofibromin to the membrane, where it dampens growth factor-induced Ras activity, providing a satisfying explanation for the overlapping features of two human diseases.


Asunto(s)
Manchas Café con Leche/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Neurofibromatosis 1/metabolismo , Neurofibromina 1/metabolismo , Proteínas Represoras/metabolismo , Animales , Humanos
20.
Nat Rev Cancer ; 12(6): 387-400, 2012 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-22622641

RESUMEN

During development and tissue homeostasis, patterns of cellular organization, proliferation and movement are highly choreographed. Receptor tyrosine kinases (RTKs) have a crucial role in establishing these patterns. Individual cells and tissues exhibit tight spatial control of the RTKs that they express, enabling tissue morphogenesis and function, while preventing unwarranted cell division and migration that can contribute to tumorigenesis. Indeed, RTKs are deregulated in most human cancers and are a major focus of targeted therapeutics. A growing appreciation of the essential role of spatial RTK regulation during development prompts the realization that spatial deregulation of RTKs is likely to contribute broadly to cancer development and may affect the sensitivity and resistance of cancer to pharmacological RTK inhibitors.


Asunto(s)
Morfogénesis/fisiología , Neoplasias/enzimología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Membrana Celular/enzimología , Cetuximab , Diseño de Fármacos , Regulación Enzimológica de la Expresión Génica , Humanos , Neoplasias/etiología , Neoplasias/patología , Proteínas Tirosina Quinasas Receptoras/genética , Transducción de Señal , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...