Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 292(46): 19076-19086, 2017 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-28931606

RESUMEN

Prion transmission between species is governed in part by primary sequence similarity between the infectious prion aggregate, PrPSc, and the cellular prion protein of the host, PrPC A puzzling feature of prion formation is that certain PrPC sequences, such as that of bank vole, can be converted by a remarkably broad array of different mammalian prions, whereas others, such as rabbit, show robust resistance to cross-species prion conversion. To examine the structural determinants that confer susceptibility or resistance to prion conversion, we systematically tested over 40 PrPC variants of susceptible and resistant PrPC sequences in a prion conversion assay. Five key residue positions markedly impacted prion conversion, four of which were in steric zipper segments where side chains from amino acids tightly interdigitate in a dry interface. Strikingly, all five residue substitutions modulating prion conversion involved the gain or loss of an asparagine or glutamine residue. For two of the four positions, Asn and Gln residues were not interchangeable, revealing a strict requirement for either an Asn or Gln residue. Bank voles have a high number of Asn and Gln residues and a high Asn:Gln ratio. These findings suggest that a high number of Asn and Gln residues at specific positions may stabilize ß-sheets and lower the energy barrier for cross-species prion transmission, potentially because of hydrogen bond networks from side chain amides forming extended Asn/Gln ladders. These data also suggest that multiple PrPC segments containing Asn/Gln residues may act in concert along a replicative interface to promote prion conversion.


Asunto(s)
Asparagina/química , Glutamina/química , Proteínas PrPC/química , Enfermedades por Prión/transmisión , Sustitución de Aminoácidos , Amiloide/química , Amiloide/genética , Amiloide/metabolismo , Animales , Arvicolinae , Asparagina/genética , Asparagina/metabolismo , Glutamina/genética , Glutamina/metabolismo , Humanos , Ratones Endogámicos C57BL , Modelos Moleculares , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Enfermedades por Prión/genética , Enfermedades por Prión/metabolismo , Enfermedades por Prión/veterinaria , Conformación Proteica en Lámina beta , Estabilidad Proteica , Conejos
2.
Cell Rep ; 17(11): 2979-2993, 2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27974211

RESUMEN

The complexity of liver tumorigenesis is underscored by the recently observed anti-oncogenic effects of oncoproteins, although the mechanisms are unclear. Shp2/Ptpn11 is a proto-oncogene in hematopoietic cells and antagonizes the effect of tumor suppressor Pten in leukemogenesis. In contrast, we show here cooperative functions of Shp2 and Pten in suppressing hepatocarcinogenesis. Ablating both Shp2 and Pten in hepatocytes induced early-onset non-alcoholic steatohepatitis (NASH) and promoted genesis of liver tumor-initiating cells likely due to augmented cJun expression/activation and elevated ROS and inflammation in the hepatic microenvironment. Inhibiting cJun partially suppressed NASH-driven liver tumorigenesis without improving NASH. SHP2 and PTEN deficiencies were detected in liver cancer patients with poor prognosis. These data depict a mechanism of hepato-oncogenesis and suggest a potential therapeutic strategy.


Asunto(s)
Carcinogénesis/genética , Enfermedad del Hígado Graso no Alcohólico/genética , Fosfohidrolasa PTEN/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Ratones Noqueados , Células Madre Neoplásicas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Proto-Oncogenes Mas , Transducción de Señal
3.
Cell Metab ; 20(2): 320-32, 2014 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-24981838

RESUMEN

Bile acid (BA) biosynthesis is tightly controlled by intrahepatic negative feedback signaling elicited by BA binding to farnesoid X receptor (FXR) and also by enterohepatic communication involving ileal BA reabsorption and FGF15/19 secretion. However, how these pathways are coordinated is poorly understood. We show here that nonreceptor tyrosine phosphatase Shp2 is a critical player that couples and regulates the intrahepatic and enterohepatic signals for repression of BA synthesis. Ablating Shp2 in hepatocytes suppressed signal relay from FGFR4, receptor for FGF15/19, and attenuated BA activation of FXR signaling, resulting in elevation of systemic BA levels and chronic hepatobiliary disorders in mice. Acting immediately downstream of FGFR4, Shp2 associates with FRS2α and promotes the receptor activation and signal relay to several pathways. These results elucidate a molecular mechanism for the control of BA homeostasis by Shp2 through the orchestration of multiple signals in hepatocytes.


Asunto(s)
Ácidos y Sales Biliares/biosíntesis , Hígado/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Transducción de Señal , Animales , Conductos Biliares/lesiones , Línea Celular , Colesterol 7-alfa-Hidroxilasa/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Hígado/patología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteína Tirosina Fosfatasa no Receptora Tipo 11/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Regulación hacia Arriba
4.
ACS Med Chem Lett ; 5(1): 61-64, 2014 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-24490002

RESUMEN

Influenza is a global human health threat, and there is an immediate need for new antiviral therapies to circumvent the limitations of vaccination and current small molecule therapies. During viral transcription, influenza incorporates the 5'-end of the host cell's mRNA in a process that requires the influenza endonuclease. Based on recently published endonuclease crystalized structures, a three-dimensional pharmacophore was developed and used to virtually screen 450,000 compounds for influenza endonuclease inhibitors. Of 264 compounds tested in a FRET-based endonuclease-inhibition assay, 16 inhibitors (IC50 <50 µM) that span 5 molecular classes novel to this endonuclease were found (6.1% hit rate). To determine cytotoxicity and antiviral activity, subsequent cellular assays were performed. Two compounds suppress viral replication with negligible cell toxicity.

5.
Proc Natl Acad Sci U S A ; 110(1): E79-88, 2013 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-23236157

RESUMEN

The molecular mechanism underlying adipogenesis and the physiological functions of adipose tissue are not fully understood. We describe here a unique mouse model of severe lipodystrophy. Ablation of Ptpn11/Shp2 in adipocytes, mediated by aP2-Cre, led to premature death, lack of white fat, low blood pressure, compensatory erythrocytosis, and hepatic steatosis in Shp2(fat-/-) mice. Fat transplantation partially rescued the lifespan and blood pressure in Shp2(fat-/-) mice, and administration of leptin also restored partially the blood pressure of mutant animals with endogenous leptin deficiency. Consistently, homozygous deletion of Shp2 inhibited adipocyte differentiation from embryonic stem (ES) cells. Biochemical analyses suggest a Shp2-TAO2-p38-p300-PPARγ pathway in adipogenesis, in which Shp2 suppresses p38 activation, leading to stabilization of p300 and enhanced PPARγ expression. Inhibition of p38 restored adipocyte differentiation from Shp2(-/-) ES cells, and p38 signaling is also suppressed in obese patients and obese animals. These results illustrate an essential role of adipose tissue in mammalian survival and physiology and also suggest a common signaling mechanism involved in adipogenesis and obesity development.


Asunto(s)
Adipogénesis/fisiología , Modelos Animales de Enfermedad , Lipodistrofia/fisiopatología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Tejido Adiposo/trasplante , Animales , Presión Sanguínea/efectos de los fármacos , Cartilla de ADN/genética , Proteína p300 Asociada a E1A/metabolismo , Eliminación de Gen , Leptina/administración & dosificación , Leptina/deficiencia , Leptina/farmacología , Ratones , Ratones Noqueados , PPAR gamma/metabolismo , Proteínas Quinasas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
Mol Cell Biol ; 32(10): 1867-78, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22431513

RESUMEN

In mammals, leptin regulates food intake and energy balance mainly through the activation of LepRb in the hypothalamus, and estrogen has a leptin-like effect in the hypothalamic control of metabolism. However, it remains to be elucidated how estrogen signaling is intertwined with the leptin pathway. We show here that Shp2, a nonreceptor tyrosine phosphatase, acts to integrate leptin and estrogen signals. The expression of a dominant-active mutant (Shp2(D61A)) in forebrain neurons conferred female, but not male, transgenic mice resistance to high-fat diet (HFD)-induced obesity and liver steatosis, accompanied by improved insulin sensitivity and glucose homeostasis. Fed with either HFD or regular chow food, Shp2(D61A) female mice showed dramatically enhanced leptin sensitivity. Microinjection of Shp2(D61A)-expressing adeno-associated virus into mediobasal hypothalamus elicited a similar antiobese effect in female mice. Biochemical analyses showed a physical association of Shp2 with estrogen receptor alpha, which is necessary for the synergistic and persistent activation of Erk by leptin and estrogen. Together, these results elucidate a mechanism for the direct cross talk of leptin and estrogen signaling and offer one explanation for the propensity of postmenopausal women to develop obesity.


Asunto(s)
Peso Corporal/genética , Metabolismo Energético/genética , Estrógenos/metabolismo , Leptina/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Animales , Dieta Alta en Grasa , Receptor alfa de Estrógeno/metabolismo , Femenino , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Transgénicos , Mutación , Neuronas/metabolismo , Obesidad/genética , Obesidad/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Receptores de Leptina/metabolismo , Factores Sexuales , Transducción de Señal
7.
Blood ; 117(20): 5350-61, 2011 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-21450902

RESUMEN

The stem cell factor (SCF)/Kit system has served as a classic model in deciphering molecular signaling events in the hematopoietic compartment, and Kit expression is a most critical marker for hematopoietic stem cells (HSCs) and progenitors. However, it remains to be elucidated how Kit expression is regulated in HSCs. Herein we report that a cytoplasmic tyrosine phosphatase Shp2, acting downstream of Kit and other RTKs, promotes Kit gene expression, constituting a Kit-Shp2-Kit signaling axis. Inducible ablation of PTPN11/Shp2 resulted in severe cytopenia in BM, spleen, and peripheral blood in mice. Shp2 removal suppressed the functional pool of HSCs/progenitors, and Shp2-deficient HSCs failed to reconstitute lethally irradiated recipients because of defects in homing, self-renewal, and survival. We show that Shp2 regulates coordinately multiple signals involving up-regulation of Kit expression via Gata2. Therefore, this study reveals a critical role of Shp2 in maintenance of a functional HSC/progenitor pool in adult mammals, at least in part through a kinase-phosphatase-kinase cascade.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Anemia Aplásica , Animales , Apoptosis , Secuencia de Bases , Enfermedades de la Médula Ósea , Trastornos de Fallo de la Médula Ósea , Trasplante de Médula Ósea , Proliferación Celular , Regulación hacia Abajo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Retroalimentación Fisiológica , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA2/metabolismo , Hematopoyesis , Hemoglobinuria Paroxística/etiología , Leucopenia/etiología , Ratones , Ratones Noqueados , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 11/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Proto-Oncogénicas c-kit/genética , ARN Interferente Pequeño/genética , Transducción de Señal
8.
Stem Cells ; 27(8): 1772-81, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19544437

RESUMEN

Abundant cell death is observed when human embryonic stem cells (hESCs) undergo neuralization, a critical first step for future cell-based therapies addressing neurodegeneration. Using hESC neuralization as an in vitro model of human development, we demonstrated that the developing neuroepithelium acquires increased susceptibility to spontaneous cell death. We found that poly(ADP-ribose) polymerase-1 (PARP1)/apoptosis-inducing factor (AIF)-mediated cell death (parthanatos) is a dominant mechanism responsible for cell loss during hESC neuralization. The demise of neural progenitor cells, at least in part, is due to decreased endogenous antioxidant defenses and enhanced reactive oxygen species leakage from mitochondria fuelled by nonphysiological culture conditions. Under such conditions, PARP1 overactivation triggered cell death through the mitochondrial-nuclear translocation of AIF. Blocking PARP1 activity with small hairpin RNA interference or nicotinamide dramatically enhanced hESC neuralization, providing optimal survival of the developing neuroepithelium. Because nicotinamide is a physiological metabolite, our results raise the possibility that neural stem/progenitor cell survival in vivo requires a metabolic niche. We argue that small natural metabolites provide a powerful physiological tool to optimize hESC differentiation compatible with the requirements of regenerative medicine.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Placa Neural/citología , Niacinamida/farmacología , Animales , Factor Inductor de la Apoptosis/genética , Factor Inductor de la Apoptosis/metabolismo , Técnicas de Cultivo de Célula , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Células Cultivadas , Células Madre Embrionarias/metabolismo , Activación Enzimática , Humanos , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Placa Neural/efectos de los fármacos , Placa Neural/metabolismo , Neuronas/metabolismo , Niacinamida/genética , Niacinamida/metabolismo , Estrés Oxidativo/fisiología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , ARN Interferente Pequeño/genética
9.
J Cell Biol ; 177(4): 683-94, 2007 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-17517963

RESUMEN

Development, angiogenesis, wound healing, and metastasis all involve the movement of cells in response to changes in the extracellular environment. To determine whether caveolin-1 plays a role in cell migration, we have used fibroblasts from knockout mice. Caveolin-1-deficient cells lose normal cell polarity, exhibit impaired wound healing, and have decreased Rho and increased Rac and Cdc42 GTPase activities. Directional persistency of migration is lost, and the cells show an impaired response to external directional stimuli. Both Src inactivation and p190RhoGAP knockdown restore the wild-type phenotype to caveolin-1-deficient cells, suggesting that caveolin-1 stimulates normal Rho GTP loading through inactivation of the Src-p190RhoGAP pathway. These findings highlight the importance of caveolin-1 in the establishment of cell polarity during directional migration through coordination of the signaling of Src kinase and Rho GTPases.


Asunto(s)
Caveolina 1/fisiología , Movimiento Celular/fisiología , Polaridad Celular/fisiología , Proteínas de Unión al GTP rho/fisiología , Familia-src Quinasas/fisiología , Animales , Caveolina 1/deficiencia , Caveolina 1/genética , Línea Celular , Células Cultivadas , Fibroblastos/citología , Fibroblastos/enzimología , Fibroblastos/fisiología , Humanos , Ratones , Ratones Noqueados , Células 3T3 NIH , Transducción de Señal/fisiología
10.
Nat Cell Biol ; 7(9): 901-8, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16113676

RESUMEN

Growth of normal cells is anchorage dependent because signalling through multiple pathways including Erk, phosphatidylinositol-3-OH kinase (PI(3)K) and Rac requires integrin-mediated cell adhesion. Components of these pathways localize to low-density, cholesterol-rich domains in the plasma membrane named 'lipid rafts' or 'cholesterol-enriched membrane microdomains' (CEMM). We previously reported that integrin-mediated adhesion regulates CEMM transport such that cell detachment from the extracellular matrix triggers CEMM internalization and clearance from the plasma membrane. We now report that this internalization is mediated by dynamin-2 and caveolin-1. Internalization requires phosphorylation of caveolin-1 on Tyr 14. A shift in localization of phospho-caveolin-1 from focal adhesions to caveolae induces CEMM internalization upon cell detachment, which mediates inhibition of Erk, PI(3)K and Rac. These data define a novel molecular mechanism for growth and tumour suppression by caveolin-1.


Asunto(s)
Caveolinas/metabolismo , Endocitosis/fisiología , Integrinas/metabolismo , Microdominios de Membrana/metabolismo , Animales , Caveolas/metabolismo , Caveolina 1 , Adhesión Celular/fisiología , Proliferación Celular , Dinamina II/metabolismo , Matriz Extracelular/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Adhesiones Focales/metabolismo , Microdominios de Membrana/ultraestructura , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Células 3T3 NIH , Invasividad Neoplásica/fisiopatología , Neoplasias/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas de Unión al GTP rac/metabolismo
11.
Science ; 303(5659): 839-42, 2004 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-14764880

RESUMEN

Translocation of the small GTP-binding protein Rac1 to the cell plasma membrane is essential for activating downstream effectors and requires integrin-mediated adhesion of cells to extracellular matrix. We report that active Rac1 binds preferentially to low-density, cholesterol-rich membranes, and specificity is determined at least in part by membrane lipids. Cell detachment triggered internalization of plasma membrane cholesterol and lipid raft markers. Preventing internalization maintained Rac1 membrane targeting and effector activation in nonadherent cells. Regulation of lipid rafts by integrin signals may regulate the location of membrane domains such as lipid rafts and thereby control domain-specific signaling events in anchorage-dependent cells.


Asunto(s)
Membrana Celular/metabolismo , Integrinas/metabolismo , Microdominios de Membrana/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Sitios de Unión , Adhesión Celular , Línea Celular , Células Cultivadas , Toxina del Cólera/metabolismo , Colesterol/metabolismo , Gangliósido G(M1)/metabolismo , Glicosilfosfatidilinositoles/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Integrina beta1/metabolismo , Liposomas/metabolismo , Ratones , Células 3T3 NIH , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Transfección , Proteína de Unión al GTP rac1/genética
12.
Circ Res ; 90(6): 697-702, 2002 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-11934838

RESUMEN

PAK1 is a protein kinase downstream of the small GTPases Rac and Cdc42 that previous work has implicated in endothelial cell migration via modulation of cell contraction. The first proline-rich region of PAK that binds to an SH3 domain from the adapter protein NCK was responsible for these dominant-negative effects. To test the role of PAK in angiogenesis, we prepared a peptide in which the proline-rich region was fused to the polybasic sequence from the HIV Tat protein to facilitate entry into cells. We show that the short peptide selectively binds NCK, whereas a mutant peptide does not. Treatment of cells with the PAK peptide but not the control peptide disrupts localization of PAK. This peptide specifically inhibited endothelial cell migration and contractility similarly to full-length dominant-negative PAK. In an in vitro tube-forming assay, the PAK peptide specifically blocked formation of multicellular networks. In an in vivo chick chorioallantoic membrane assay, the PAK peptide specifically blocked angiogenesis. These results, therefore, suggest a role for PAK in angiogenesis.


Asunto(s)
Movimiento Celular/fisiología , Endotelio Vascular/fisiología , Neovascularización Fisiológica/fisiología , Proteínas Oncogénicas/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Adaptadoras Transductoras de Señales , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Productos del Gen tat/genética , Productos del Gen tat/fisiología , Humanos , Mutación , Neovascularización Fisiológica/efectos de los fármacos , Péptidos/genética , Péptidos/farmacología , Unión Proteica , Proteínas Recombinantes de Fusión/farmacología , Quinasas p21 Activadas
13.
Nat Cell Biol ; 4(3): 232-9, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11862216

RESUMEN

The proper function of Rho GTPases requires precise spatial and temporal regulation of effector interactions. Integrin-mediated cell adhesion modulates the interaction of GTP-Rac with its effectors by controlling GTP-Rac membrane targeting. Here, we show that the translocation of GTP-Rac to membranes is independent of effector interactions, but instead requires the polybasic sequence near the carboxyl terminus. Cdc42 also requires integrin-mediated adhesion for translocation to membranes. A recently developed fluorescence resonance energy transfer (FRET)-based assay yields the surprising result that, despite its uniform distribution, the interaction of activated V12-Rac with a soluble, cytoplasmic effector domain is enhanced at specific regions near cell edges and is induced locally by integrin stimulation. This enhancement requires Rac membrane targeting. We show that Rho-GDI, which associates with cytoplasmic GTP-Rac, blocks effector binding. Release of Rho-GDI after membrane translocation allows Rac to bind to effectors. Thus, Rho-GDI confers spatially restricted regulation of Rac-effector interactions.


Asunto(s)
Inhibidores de Disociación de Guanina Nucleótido/metabolismo , Integrinas/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Células 3T3 , Animales , Membrana Celular/metabolismo , Humanos , Ratones , Modelos Biológicos , Unión Proteica , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/genética , Inhibidores de la Disociación del Nucleótido Guanina rho-Específico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA