Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Immunity ; 45(1): 94-105, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27438768

RESUMEN

Although the intracellular trafficking system is integral to most physiologic activities, its role in mediating immune responses to infection has remained elusive. Here, we report that infected bladder epithelial cells (BECs) mobilized the exocyst complex, a powerful exporter of subcellular vesicles, to rapidly expel intracellular bacteria back for clearance. Toll-like receptor (TLR) 4 signals emanating from bacteria-containing vesicles (BCVs) were found to trigger K33-linked polyubiquitination of TRAF3 at Lys168, which was then detected by RalGDS, a guanine nucleotide exchange factor (GEF) that precipitated the assembly of the exocyst complex. Although this distinct modification of TRAF3 served to connect innate immune signaling to the cellular trafficking apparatus, it crucially ensured temporal and spatial accuracy in determining which among the many subcellular vesicles was recognized and selected for expulsion in response to innate immune signaling.


Asunto(s)
Escherichia coli/inmunología , Inmunidad Innata , Factor 3 Asociado a Receptor de TNF/metabolismo , Vesículas Transportadoras/metabolismo , Vejiga Urinaria/patología , Infecciones Urinarias/inmunología , Urotelio/inmunología , Animales , Células Cultivadas , Escherichia coli/genética , Exocitosis , Femenino , Humanos , Espacio Intracelular , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño/genética , Transducción de Señal , Factor 3 Asociado a Receptor de TNF/genética , Receptor Toll-Like 4/genética , Ubiquitinación , Vejiga Urinaria/microbiología , Urotelio/microbiología , Factor de Intercambio de Guanina Nucleótido ral/genética , Factor de Intercambio de Guanina Nucleótido ral/metabolismo
2.
Exp Eye Res ; 185: 107688, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31185219

RESUMEN

Photoreceptor degeneration is a significant cause of visual impairment in the United States and globally. Cell replacement therapy shows great promise in restoring vision by transplanting stem-like cells into the sub-retinal space as substitutes for damaged photoreceptors. However, vision repair via transplantation has been limited, in large part, by low numbers of replacement cells able to migrate into damaged retinal tissue and integrate with native photoreceptors. Projects have used external chemical fields and applied electric fields to induce the chemotaxis and electrotaxis of replacement cells, respectively, with limited success. However, the application of combined electro-chemotactic fields in directing cells within biomaterials and host tissue has been surprisingly understudied. The current work examined the ability of combined electro-chemotactic fields to direct the migration of transplantable retinal progenitor cells (RPCs) in controlled microenvironments. Experiments used our established galvano-microfluidic system (Gal-MµS) to generate tunable chemotactic concentration fields with and without superimposed electric fields. Result illustrate that combination fields increased the distance migrated by RPCs by over three times that seen in either field, individually, and with greater directionality towards increasing gradients. Interestingly, immunofluorescence assays showed no significant differences in the distribution of the total and/or activated cognate receptor of interest, indicating that changes in ligand binding alone were not responsible for the measured increases in migration. Bioinformatics analysis was then performed to identity potential, synergistic mechanistic pathways involved in the electro-chemotaxis measured. Results indicate that increased RPC migration in electro-chemotactic fields may arise from down-regulation of cell adhesion proteins in tandem with up-regulation of cytoskeletal regulation proteins. These comprehensive results point towards a novel migration-targeted treatment that may dramatically improve transplantation outcomes as well as elucidate unreported synergy across biological mechanisms in response to electro-chemotactic fields.


Asunto(s)
Movimiento Celular/fisiología , Quimiotaxis/fisiología , Campos Electromagnéticos , Retina/citología , Trasplante de Células Madre , Células Madre/citología , Animales , Cadherinas/metabolismo , Células Cultivadas , ADN-Topoisomerasas de Tipo II/genética , Expresión Génica , Inmunohistoquímica , Dispositivos Laboratorio en un Chip , Ratones , Ratones Endogámicos C57BL , Proteínas de Unión a Poli-ADP-Ribosa/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores CXCR4/genética , Células Madre/fisiología , beta Catenina/metabolismo , Factor de Intercambio de Guanina Nucleótido ral/genética
3.
Proc Natl Acad Sci U S A ; 112(21): 6625-30, 2015 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-25941399

RESUMEN

K-RAS4B (Kirsten rat sarcoma viral oncogene homolog 4B) is a prenylated, membrane-associated GTPase protein that is a critical switch for the propagation of growth factor signaling pathways to diverse effector proteins, including rapidly accelerated fibrosarcoma (RAF) kinases and RAS-related protein guanine nucleotide dissociation stimulator (RALGDS) proteins. Gain-of-function KRAS mutations occur frequently in human cancers and predict poor clinical outcome, whereas germ-line mutations are associated with developmental syndromes. However, it is not known how these mutations affect K-RAS association with biological membranes or whether this impacts signal transduction. Here, we used solution NMR studies of K-RAS4B tethered to nanodiscs to investigate lipid bilayer-anchored K-RAS4B and its interactions with effector protein RAS-binding domains (RBDs). Unexpectedly, we found that the effector-binding region of activated K-RAS4B is occluded by interaction with the membrane in one of the NMR-observable, and thus highly populated, conformational states. Binding of the RAF isoform ARAF and RALGDS RBDs induced marked reorientation of K-RAS4B from the occluded state to RBD-specific effector-bound states. Importantly, we found that two Noonan syndrome-associated mutations, K5N and D153V, which do not affect the GTPase cycle, relieve the occluded orientation by directly altering the electrostatics of two membrane interaction surfaces. Similarly, the most frequent KRAS oncogenic mutation G12D also drives K-RAS4B toward an exposed configuration. Further, the D153V and G12D mutations increase the rate of association of ARAF-RBD with lipid bilayer-tethered K-RAS4B. We revealed a mechanism of K-RAS4B autoinhibition by membrane sequestration of its effector-binding site, which can be disrupted by disease-associated mutations. Stabilizing the autoinhibitory interactions between K-RAS4B and the membrane could be an attractive target for anticancer drug discovery.


Asunto(s)
Genes ras , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Secuencia de Aminoácidos , Sitios de Unión/genética , Humanos , Membrana Dobles de Lípidos , Modelos Moleculares , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Proto-Oncogénicas p21(ras)/química , Homología de Secuencia de Aminoácido , Transducción de Señal , Electricidad Estática , Factor de Intercambio de Guanina Nucleótido ral/química , Factor de Intercambio de Guanina Nucleótido ral/genética , Factor de Intercambio de Guanina Nucleótido ral/metabolismo
4.
J Cell Sci ; 128(16): 2957-64, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26240175

RESUMEN

The exocyst is an octameric protein complex that is implicated in the tethering of secretory vesicles to the plasma membrane prior to SNARE-mediated fusion. Spatial and temporal control of exocytosis through the exocyst has a crucial role in a number of physiological processes, such as morphogenesis, cell cycle progression, primary ciliogenesis, cell migration and tumor invasion. In this Cell Science at a Glance poster article, we summarize recent works on the molecular organization, function and regulation of the exocyst complex, as they provide rationales to the involvement of this complex in such a diverse array of cellular processes.


Asunto(s)
Membrana Celular/metabolismo , Exocitosis/genética , Proteínas de la Fusión de la Membrana/genética , Fusión de Membrana/genética , Animales , Ciclo Celular/genética , Membrana Celular/genética , Humanos , Proteínas de la Fusión de la Membrana/metabolismo , Morfogénesis/genética , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo , Factor de Intercambio de Guanina Nucleótido ral/genética , Factor de Intercambio de Guanina Nucleótido ral/metabolismo
5.
J Mol Cell Cardiol ; 59: 128-38, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23473774

RESUMEN

Recent work has demonstrated that autophagy, a phylogenetically conserved, lysosome-mediated pathway of protein degradation, is a key participant in pathological cardiac remodeling. One common feature of cell growth and autophagy is membrane biogenesis and processing. The exocyst, an octomeric protein complex involved in vesicle trafficking, is implicated in numerous cellular processes, yet its role in cardiomyocyte plasticity is unknown. Here, we set out to explore the role of small G protein-dependent control of exocyst function and membrane trafficking in stress-induced cardiomyocyte remodeling and autophagy. First, we tested in cultured neonatal rat cardiomyocytes (NRCMs) two isoforms of Ral (RalA, RalB) whose actions are mediated by the exocyst. In these experiments, mTOR inhibition in response to starvation or Torin1 was preserved despite RalA or RalB knockdown; however, activation of autophagy was suppressed only in NRCMs depleted of RalB, implicating RalB as being required for mTOR-dependent cardiomyocyte autophagy. To define further the role of RalB in cardiomyocyte autophagy, we analyzed hearts from mice lacking RalGDS (Ralgds(-/-)), a guanine exchange factor (GEF) for the Ral family of small GTPases. RalGDS-null hearts were similar to wild-type (WT) littermates in terms of ventricular structure, contractile performance, and gene expression. However, Ralgds(-/-) hearts manifested a blunted growth response (p<0.05) to TAC-mediated pressure-overload stress. Ventricular chamber size and contractile performance were preserved in response to TAC in Ralgds(-/-) mice, and load-induced cardiomyocyte autophagy was suppressed. Interestingly, TAC-induced activation of the fetal gene program was similar in both genotypes despite the relative lack of hypertrophic growth in mutant hearts. Together, these data implicate RalGDS-mediated induction of autophagy and exocyst function as a critical feature of load-induced cardiac hypertrophy.


Asunto(s)
Autofagia/fisiología , Cardiomegalia/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Factor de Intercambio de Guanina Nucleótido ral/metabolismo , Animales , Autofagia/genética , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Factor de Intercambio de Guanina Nucleótido ral/genética
6.
Cancer Cell ; 7(3): 219-26, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15766660

RESUMEN

To investigate the role of signaling by the small GTPase Ral, we have generated mice deficient for RalGDS, a guanine nucleotide exchange factor that activates Ral. We show that RalGDS is dispensable for mouse development but plays a substantial role in Ras-induced oncogenesis. Lack of RalGDS results in reduced tumor incidence, size, and progression to malignancy in multistage skin carcinogenesis, and reduced transformation by Ras in tissue culture. RalGDS does not appear to participate in the regulation of cell proliferation, but instead controls survival of transformed cells. Experiments performed in cells isolated from skin tumors suggest that RalGDS mediates cell survival through the activation of the JNK/SAPK pathway. These studies identify RalGDS as a key component in Ras-dependent carcinogenesis in vivo.


Asunto(s)
Transformación Celular Neoplásica , Transducción de Señal/fisiología , Neoplasias Cutáneas , Proteínas de Unión al GTP ral/metabolismo , Factor de Intercambio de Guanina Nucleótido ral/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Noqueados , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Proteínas de Unión al GTP ral/genética , Factor de Intercambio de Guanina Nucleótido ral/genética , Proteínas ras/metabolismo
7.
Cancer Cell ; 7(6): 533-45, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15950903

RESUMEN

RalGEFs were recently shown to be critical for Ras-mediated transformed and tumorigenic growth of human cells. We now show that the oncogenic activity of these proteins is propagated by activation of one RalGEF substrate, RalA, but blunted by another closely related substrate, RalB, and that the oncogenic signaling requires binding of the RalBP1 and exocyst subunit effector proteins. Knockdown of RalA expression impeded, if not abolished, the ability of human cancer cells to form tumors. RalA was also commonly activated in a panel of cell lines from pancreatic cancers, a disease characterized by activation of Ras. Activation of RalA signaling thus appears to be a critical step in Ras-induced transformation and tumorigenesis of human cells.


Asunto(s)
Transformación Celular Neoplásica/patología , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Proteínas de Unión al GTP ral/fisiología , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Proteínas Portadoras/metabolismo , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Expresión Génica/genética , Guanosina Trifosfato/metabolismo , Humanos , Ratones , Ratones SCID , Trasplante de Neoplasias/patología , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Unión Proteica/fisiología , Transporte de Proteínas/fisiología , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , ARN Interferente Pequeño/genética , Transfección , Proteínas de Transporte Vesicular , Proteínas de Unión al GTP ral/genética , Proteínas de Unión al GTP ral/metabolismo , Factor de Intercambio de Guanina Nucleótido ral/genética , Factor de Intercambio de Guanina Nucleótido ral/metabolismo , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
8.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 34(4): 313-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22954110

RESUMEN

OBJECTIVE: To construct certain chimeric E3s expression plasmids targetting oncoprotein Ras by harnessing the theory of protein knockdown. METHODS: We chose the binding domain of Raf-1, PI3K, RalGDS, and the function domain of F-Box as well as the U-Box to construct the plasmids. Then used the double enzyme, PCR, and sequence to test the validity and integrity of the cloned nucleotide fragments. The expression efficiency of the plasmids in eukaryotic cells was detected by Western blot analysis. RESULTS: Five of 6 plasmids in this study expressed the corresponding fusion proteins in HEK293T cells, and (RBD+CRD)(Raf-1)- U-Box-pcDNA3.1 can knocked down the protein level of Ras in PANC-1 cells. CONCLUSIONS: We successfully constructed the chimeric E3 expression plasmids, which provides a solid basis for further research on protein knockdown.


Asunto(s)
Vectores Genéticos , Proteínas Recombinantes de Fusión/genética , Ubiquitina-Proteína Ligasas/genética , Proteínas ras/genética , Clonación Molecular , Células HEK293 , Humanos , Fosfatidilinositol 3-Quinasas/genética , Plásmidos , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Factor de Intercambio de Guanina Nucleótido ral/genética
9.
Neuron ; 56(4): 604-20, 2007 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-18031680

RESUMEN

To discover new genes involved in axon navigation, we conducted a forward genetic screen for recessive alleles affecting motor neuron pathfinding in GFP reporter mice mutagenized with ENU. In Magellan mutant embryos, motor axons were error prone and wandered inefficiently at choice points within embryos, but paradoxically responded to guidance cues with normal sensitivity in vitro. We mapped the Magellan mutation to the Phr1 gene encoding a large multidomain E3 ubiquitin ligase. Phr1 is associated with the microtubule cytoskeleton within neurons and selectively localizes to axons but is excluded from growth cones. Motor and sensory neurons from Magellan mutants display abnormal morphologies due to a breakdown in the polarized distribution of components that segregate between axons and growth cones. The Magellan phenotype can be reversed by stabilizing microtubules with taxol or inhibiting p38MAPK activity. Thus, efficacious pathfinding requires Phr1 activity for coordinating the cytoskeletal organization that distinguishes axons from growth cones.


Asunto(s)
Conos de Crecimiento/metabolismo , Microtúbulos/metabolismo , Neuronas Motoras/metabolismo , Médula Espinal/anomalías , Médula Espinal/metabolismo , Factor de Intercambio de Guanina Nucleótido ral/metabolismo , Animales , Diferenciación Celular/genética , Células Cultivadas , Vías Eferentes/anomalías , Vías Eferentes/citología , Vías Eferentes/metabolismo , Inhibidores Enzimáticos/farmacología , Regulación del Desarrollo de la Expresión Génica/genética , Pruebas Genéticas , Conos de Crecimiento/ultraestructura , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Microtúbulos/ultraestructura , Neuronas Motoras/citología , Mutación/genética , Paclitaxel/farmacología , Médula Espinal/citología , Moduladores de Tubulina/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Factor de Intercambio de Guanina Nucleótido ral/genética
10.
Blood ; 112(1): 56-63, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18417737

RESUMEN

The small GTP-binding protein Ral has been implicated in regulated exocytosis via its interaction with the mammalian exocyst complex. We have previously demonstrated that Ral is involved in exocytosis of Weibel-Palade bodies (WPBs). Little is known about intracellular signaling pathways that promote activation of Ral in response to ligand binding of G protein-coupled receptors. Here we show that RNAi-mediated knockdown of RalGDS, an exchange factor for Ral, results in inhibition of thrombin- and epinephrine-induced exocytosis of WPBs, while overexpression of RalGDS promotes exocytosis of WPBs. A RalGDS variant lacking its exchange domain behaves in a dominant negative manner by blocking release of WPBs. We also provide evidence that RalGDS binds calmodulin (CaM) via an amino-terminal CaM-binding domain. RalGDS association to CaM is required for Ral activation because a cell-permeable peptide comprising this RalGDS CaM-binding domain inhibits Ral activation and WPB exocytosis. Together our findings suggest that RalGDS plays a vital role in the regulation of Ral-dependent WPB exocytosis after stimulation with Ca(2+)- or cAMP-raising agonists.


Asunto(s)
Exocitosis/fisiología , Cuerpos de Weibel-Palade/fisiología , Factor de Intercambio de Guanina Nucleótido ral/fisiología , Secuencia de Aminoácidos , Sitios de Unión/genética , Calmodulina/metabolismo , Línea Celular , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Células Endoteliales/ultraestructura , Epinefrina/farmacología , Exocitosis/efectos de los fármacos , Variación Genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Trombina/farmacología , Transfección , Proteínas de Unión al GTP ral/metabolismo , Factor de Intercambio de Guanina Nucleótido ral/antagonistas & inhibidores , Factor de Intercambio de Guanina Nucleótido ral/química , Factor de Intercambio de Guanina Nucleótido ral/genética
11.
Cell Biol Int ; 34(5): 513-22, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20067447

RESUMEN

BRAK/CXCL14 (breast- and kidney-expressed chemokine/CXC chemokine ligand 14) is a chemokine that is expressed in many normal cells and tissues but is absent from or expressed at very low levels in transformed cells and cancerous tissues, including HNSCC (head and neck squamous cell carcinoma). We reported previously that the forced expression of BRAK/CXCL14 in HNSCC (HSC-3 BRAK) cells decreased the rate of tumour formation and size of tumour xenografts compared with mock-vector-introduced (HSC-3 Mock) cells in athymic nude mice, even though the growth rates of these cells were the same under in vitro culture conditions, suggesting that high-level expression of the gene is important for the suppression of tumour establishment in vivo. For the first step to study the mechanisms of BRAK-dependent tumour suppression, we compared characteristics between HSC-3 BRAK and HSC-3 Mock cells under in vitro culture conditions. The cell migration rate was lower in HSC-3 BRAK cells than in HSC-3 Mock cells. Also, HSC-3 BRAK cells showed more rapid adhesion than HSC-3 Mock cells when cultured on type I collagen-coated dishes but not on fibronectin or laminin 1-coated ones. This adhesion was mediated by alpha2beta1 integrin. Immunofluorescent analysis of the cells cultured on type I collagen showed that HSC-3 BRAK cells formed much more elongated focal adhesions co-localized with paxillin and actin stress fibres than did HSC-3 Mock cells. Treatment of parental HSC-3 cells with recombinant BRAK stimulated the activation of Rap1, which is a ras family small GTPase, and formation of elongated focal adhesions, indicating that the difference in cell character observed between HSC-3 Mock and HSC-3 BRAK was not due to selection of clones of different character but due to expression of BRAK in the cells. The characteristic morphology of focal adhesions in HSC-3 BRAK cells was perturbed by the introduction of an expression vector of the Rap-binding domain of the Ral guanine nucleotide dissociation stimulator, a target of Rap1, into HSC-3 BRAK cells, suggesting that Rap1 regulated the formation of the morphology of the focal adhesions. These data indicate that the expression of BRAK stimulated the formation of elongated focal adhesions of the HSC-3 cells in an autocrine or paracrine fashion, in which stimulation may be responsible for the reduced migration of the cells.


Asunto(s)
Adhesión Celular/fisiología , Movimiento Celular/fisiología , Quimiocinas CXC/metabolismo , Colágeno Tipo I/metabolismo , Adhesiones Focales , Neoplasias de la Lengua , Animales , Proteínas Reguladoras de la Apoptosis , Comunicación Celular/fisiología , Línea Celular Tumoral , Quimiocinas CXC/genética , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Adhesiones Focales/metabolismo , Adhesiones Focales/ultraestructura , Proteínas de Unión al GTP , Humanos , Integrina alfa2beta1/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Desnudos , Paxillin/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Neoplasias de la Lengua/metabolismo , Neoplasias de la Lengua/patología , Proteína de Unión al GTP cdc42/metabolismo , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Factor de Intercambio de Guanina Nucleótido ral/genética , Factor de Intercambio de Guanina Nucleótido ral/metabolismo , Proteínas de Unión al GTP rap1/metabolismo
12.
Int Immunopharmacol ; 83: 106454, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32259700

RESUMEN

Lung adenocarcinoma (LUAD) is a frequently diagnosed histologic subtype with increasing morbidity and mortality. RalGDS-Like 4 (RGL4) has not been reported to be associated with cancer risk, prognosis, immunotherapy or any other treatments. We perform a bioinformatics analysis on data downloaded from the Cancer Genome Atlas (TCGA)-LUAD, and we find that low expression of RGL4 is accompanied by worse outcomes and prognosis in LUAD patients. As a promising predictor, the potential influence and mechanisms of RGL4 on overall survival are worth exploring. Moreover, RGL4 expression is significantly associated with a variety of tumor-infiltrating immune cells (TIICs), particularly memory B cells, CD8+T cells and neutrophils. Subsequently, we evaluated the most notable KEGG pathways, including glycolysis gluconeogenesis, the P53 signaling pathway, RNA degradation, and the B cell receptor signaling pathway, among others. Our findings provide evidence that the decreased expression of RGL4 is significantly associated with poor prognosis and immune cell infiltration in patients with LUAD and highlight the use of RGL4 as a novel predictive biomarker for the prognosis of LUAD and other cancers. RGL4 may also be used in combination with immune checkpoints to identify the benefits of immunotherapy. Subjects: Bioinformatics, Genomics, Oncology, Thoracic surgery.


Asunto(s)
Adenocarcinoma/metabolismo , Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Neoplasias Pulmonares/metabolismo , Pulmón/inmunología , Factor de Intercambio de Guanina Nucleótido ral/metabolismo , Adenocarcinoma/diagnóstico , Adenocarcinoma/mortalidad , Movimiento Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/mortalidad , Masculino , Persona de Mediana Edad , Pronóstico , Transducción de Señal , Análisis de Supervivencia , Proteína p53 Supresora de Tumor/metabolismo , Factor de Intercambio de Guanina Nucleótido ral/genética
13.
Mol Cell Biol ; 26(10): 3966-75, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16648489

RESUMEN

Hyperactivation of Ras is one of the most common abnormalities in acute myeloid leukemia. In experimental models, Ras inhibits myeloid differentiation, which is characteristic of leukemia; however, the mechanism through which it disrupts hematopoiesis is poorly understood. In multipotent FDCP-mix cells, Ras inhibits terminal neutrophil differentiation, thereby indefinitely extending their proliferative potential. Ras also strongly promotes the sensitivity of these cells to granulocyte-macrophage colony-stimulating factor (GM-CSF). Using this model, we have dissected the signaling elements downstream of Ras to determine their relative contribution to the dysregulation of hematopoiesis. Cells expressing Ras mutants selectively activating Raf (Ras*T35S) or phosphatidylinositol 3-kinase (Ras*Y40C) did not significantly affect differentiation or proliferative capacity, whereas Ras*E37G (which selectively activates RalGEFs) perpetuated proliferation and blocked neutrophil development in a manner similar to that of Ras. Correspondingly, expression of constitutively active versions of these effectors confirmed the overriding importance of Ral guanine nucleotide exchange factors. Cells expressing Ras demonstrated hyperactivation of Ral, which itself was able to exactly mimic the phenotype of Ras, including hypersensitivity to GM-CSF. Conversely, dominant negative Ral promoted spontaneous neutrophil development. Ral, in turn, appears to influence differentiation through multiple effectors. These data show, for the first time, the importance of Ral in regulating differentiation and self-renewal in hematopoietic cells.


Asunto(s)
Diferenciación Celular , Genes ras , Leucemia Mieloide/fisiopatología , Factor de Intercambio de Guanina Nucleótido ral/fisiología , Proteínas ras/fisiología , Animales , Western Blotting , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Proliferación Celular , Células Clonales , Relación Dosis-Respuesta a Droga , Activación Enzimática , Regulación Leucémica de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Leucemia Mieloide/genética , Ratones , Mutación , Neutrófilos/citología , Neutrófilos/metabolismo , Retroviridae/genética , Factor de Intercambio de Guanina Nucleótido ral/genética
14.
Cell Signal ; 19(7): 1575-82, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17382517

RESUMEN

A cDNA encoding a RalGDS-related protein, Rgl3, was isolated by yeast two-hybrid screening using a small G-protein, Rap1, as a bait. Rgl3 mRNA is commonly detectable in several visceral organs (e.g. kidney, heart, liver, and lung) in the mouse and human. The Rgl3 protein mainly localizes in the cytoplasm when expressed in fibroblasts. Yeast two-hybrid assay indicated that Rgl3 could interact with Rap1, Rap2, H-Ras, N-Ras, and R-Ras but failed to interact efficiently with Ral and Rho. Interestingly, Rgl3 was found to affect cell morphology in two assay systems in culture. First, Rgl3 suppressed cell-spreading induced by Rap1, R-Ras, or C3G-CAAX (a membrane-targeted Rap/R-Ras activator) in HEK-293 cells. Second, Rgl3 enhanced the focus-formation induced by oncogenic H-Ras and N-Ras mutants in NIH3T3 cells. Moreover, we identified profilin II as a potential binding partner for Rgl3 by yeast two-hybrid screening. This interaction requires the characteristic proline cluster in the Rgl3 amino-terminal domain. Profilin II and Rgl3 co-operated in enhancing the N-Ras-induced focus-formation. These findings raise the possibility that Rgl3 mediates interaction between Ras/Rap-family proteins and profilin II, an important activator of actin polymerization.


Asunto(s)
Profilinas/metabolismo , Factor de Intercambio de Guanina Nucleótido ral/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular Transformada , Movimiento Celular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Fracciones Subcelulares/metabolismo , Factor de Intercambio de Guanina Nucleótido ral/química , Factor de Intercambio de Guanina Nucleótido ral/genética , Proteínas ras/metabolismo
15.
Mol Cell Biol ; 23(3): 1112-24, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12529414

RESUMEN

Ras GTPases are central to many physiological and pathological signaling pathways and act via a combination of effectors. In mammals, at least three Ral exchange factors (RalGEFs) contain a Ras association domain and constitute a discrete subgroup of Ras effectors. Despite their ability to bind activated Rap as well as activated Ras, they seem to act downstream of Ras but not downstream of Rap. We have revisited the Ras/Rap-Ral connections in Drosophila melanogaster by using iterative two-hybrid screens with these three GTPases as primary baits and a subsequent genetic approach. We show that (i) the Ral-centered protein network appears to be extremely conserved in human and flies, (ii) in this network, RGL is a functional Drosophila orthologue of RalGEFs, and (iii) the RGL-Ral pathway functionally interacts with both the Ras and Rap pathways. Our data do not support the paradigmatic model where Ral is in the effector pathway of Ras. They reveal a signaling circuitry where Ral is functionally downstream of the Rap GTPase, at odds with the pathways described for mammalian cell lines. Thus, in vivo data show variations in the connectivity of pathways described for cell lines which might display only a subset of the biological possibilities.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Factor de Intercambio de Guanina Nucleótido ral/genética , Factor de Intercambio de Guanina Nucleótido ral/metabolismo , Proteínas de Unión al GTP rap/genética , Proteínas de Unión al GTP rap/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo , Alelos , Animales , Animales Modificados Genéticamente , Drosophila melanogaster/crecimiento & desarrollo , Ojo/crecimiento & desarrollo , Ojo/ultraestructura , Regulación del Desarrollo de la Expresión Génica , Genes de Insecto , Humanos , Microscopía Electrónica de Rastreo , Especificidad de la Especie , Técnicas del Sistema de Dos Híbridos
16.
Mol Cell Biol ; 21(8): 2650-8, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11283245

RESUMEN

Ras proteins have the capacity to bind to and activate at least three families of downstream target proteins: Raf kinases, phosphatidylinositol 3 (PI 3)-kinase, and Ral-specific guanine nucleotide exchange factors (Ral-GEFs). We have previously shown that the Ras/Ral-GEF and Ras/Raf pathways oppose each other upon nerve growth factor stimulation, with the former promoting proliferation and the latter promoting cell cycle arrest. Moreover, the pathways are not activated equally. While the Ras/Raf/Erk signaling pathway is induced for hours, the Ras/Ral-GEF/Ral signaling pathway is induced for only minutes. Here we show that this preferential down-regulation of Ral signaling is mediated, at least in part, by protein kinase C (PKC). In particular, we show that PKC activation by phorbol ester treatment of cells blocks growth factor-induced Ral activation while it enhances Erk activation. Moreover, suppression of growth factor-induced PKC activation enhances and prolongs Ral activation. PKC does not influence the basal activity of the Ral-GEF designated Ral-GDS but suppresses its activation by Ras. Interestingly, Ras binding to the C-terminal Ras binding domain of Ral-GDS is not affected by PKC activity. Instead, suppression of Ral-GDS activation occurs through the region N terminal to the catalytic domain, which becomes phosphorylated in response to phorbol ester treatment of cells. These findings identify a role for PKC in determining the specificity of Ras signaling by its ability to differentially modulate Ras effector protein activation.


Asunto(s)
Proteína Quinasa C/metabolismo , Proteínas ras/metabolismo , Animales , Células COS , Activación Enzimática/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Factor de Crecimiento Nervioso/farmacología , Células PC12 , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas c-raf/metabolismo , Ratas , Eliminación de Secuencia , Transducción de Señal , Acetato de Tetradecanoilforbol/farmacología , Factor de Intercambio de Guanina Nucleótido ral/química , Factor de Intercambio de Guanina Nucleótido ral/genética , Factor de Intercambio de Guanina Nucleótido ral/metabolismo
17.
Mol Cell Biol ; 23(17): 6037-48, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12917328

RESUMEN

Development and regeneration of muscle tissue is a highly organized, multistep process that requires cell proliferation, migration, differentiation, and maturation. Previous data implicate fibroblast growth factors (FGFs) as critical regulators of these processes, although their precise role in vivo is still not clear. We have explored the consequences of the loss of multiple FGFs (FGF2 and FGF6 in particular) for muscle regeneration in mdx mice, which serve as a model for chronic muscle damage. We show that the combined loss of FGF2 and FGF6 leads to severe dystrophic changes in the musculature. We found that FGF6 mutant myoblasts had decreased migration ability in vivo, whereas wild-type myoblasts migrated normally in a FGF6 mutant environment after transplantation of genetically labeled myoblasts from FGF6 mutants in wild-type mice and vice versa. In addition, retrovirus-mediated expression of dominant-negative versions of Ras and Ral led to a reduced migration of transplanted myoblasts in vivo. We propose that FGFs are critical components of the muscle regeneration machinery that enhance skeletal muscle regeneration, probably by stimulation of muscle stem cell migration.


Asunto(s)
Movimiento Celular/genética , Factor 2 de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/genética , Distrofias Musculares/patología , Mioblastos Esqueléticos/patología , Proteínas Proto-Oncogénicas/genética , Animales , Trasplante de Células , Células Cultivadas , Distrofina/genética , Factor 2 de Crecimiento de Fibroblastos/deficiencia , Factor 6 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones Mutantes , Músculo Esquelético/patología , Músculo Esquelético/fisiología , Distrofias Musculares/genética , Mioblastos Esqueléticos/fisiología , Mioblastos Esqueléticos/trasplante , Proteínas Proto-Oncogénicas/deficiencia , Regeneración/genética , Retroviridae/genética , Transducción de Señal , Factor de Intercambio de Guanina Nucleótido ral/genética , Factor de Intercambio de Guanina Nucleótido ral/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
18.
Cancer Res ; 64(17): 6041-9, 2004 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15342385

RESUMEN

To study the oncogenic potential of Rgr in vivo, we have generated several transgenic Rgr mouse lines, which express the oncogene under the control of different promoters. These studies revealed that Rgr expression leads to the generation of various pathological alterations, including fibrosarcomas, when its transgenic expression is restricted to nonlymphoid tissues. Moreover, the overall incidence and latency of fibrosarcomas were substantially increased and shortened, respectively, in a p15INK4b-defective background. More importantly, we also have demonstrated that Rgr expression in thymocytes of transgenic mice induces severe alterations in the development of the thymocytes, which eventually lead to a high incidence of thymic lymphomas. This study demonstrates that oncogenic Rgr can induce expression of p15INK4b and, more importantly, that both Rgr and p15INK4b cooperate in the malignant phenotype in vivo. These findings provide new insights into the tumorigenic role of Rgr as a potent oncogene and show that p15INK4b can act as a tumor suppressor gene.


Asunto(s)
Transformación Celular Neoplásica/genética , Linfoma de Células T/genética , Factor de Intercambio de Guanina Nucleótido ral/genética , Animales , Antígenos CD4/genética , Proteínas de Ciclo Celular/genética , Inhibidor p15 de las Quinasas Dependientes de la Ciclina , Progresión de la Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células 3T3 NIH , Transfección , Proteínas Supresoras de Tumor/genética
19.
Cancer Res ; 63(14): 4188-95, 2003 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12874025

RESUMEN

Previous studies have identified a novel oncogene, rgr, which has homology to the guanine nucleotide exchange factor (GEF) Ral guanine dissociation stimulator (RALGDS). To determine the mechanism of activation of rgr, the wild-type form was isolated. rgr is expressed physiologically at very low levels, due, at least in part, to a long 5'-untranslated region that contains eight AUGs, which inhibit translation of the main open reading frame. When these regulatory sequences are removed, the wild-type gene is expressed at high levels. An investigation of how this GEF could transform cells showed that RGR interacts with RAS, supporting its involvement as a RAS-GEF. Because RAL is localized mainly to the Golgi, the expression of the RGR protein was identified in RK13 cells, a cell line that expresses endogenous rgr. RGR localizes to endomembranes. To determine its location upon transformation, a green fluorescent protein-RGR fusion protein was used to track the movement of RGR. Increasing amounts of expression result in enhanced localization of RGR to the plasma membrane. These results indicate that rgr is activated when its tight translational controls are eliminated and increased expression allows its relocation to the plasma membrane, where efficient activation of RAS occurs.


Asunto(s)
Oncogenes/genética , Factor de Intercambio de Guanina Nucleótido ral/genética , Factor de Intercambio de Guanina Nucleótido ral/metabolismo , Células 3T3 , Regiones no Traducidas 5'/genética , Animales , Secuencia de Bases , Transformación Celular Neoplásica/genética , ADN Complementario/genética , ADN Complementario/aislamiento & purificación , Regulación de la Expresión Génica , Humanos , Ratones , Datos de Secuencia Molecular , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Biosíntesis de Proteínas , ARN Mensajero/genética , Conejos , Fracciones Subcelulares/metabolismo , Factor de Intercambio de Guanina Nucleótido ral/biosíntesis , Proteínas ras/fisiología
20.
PLoS One ; 11(12): e0167145, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27936046

RESUMEN

RAS effectors specifically interact with the GTP-bound form of RAS in response to extracellular signals and link them to downstream signaling pathways. The molecular nature of effector interaction by RAS is well-studied but yet still incompletely understood in a comprehensive and systematic way. Here, structure-function relationships in the interaction between different RAS proteins and various effectors were investigated in detail by combining our in vitro data with in silico data. Equilibrium dissociation constants were determined for the binding of HRAS, KRAS, NRAS, RRAS1 and RRAS2 to both the RAS binding (RB) domain of CRAF and PI3Kα, and the RAS association (RA) domain of RASSF5, RALGDS and PLCε, respectively, using fluorescence polarization. An interaction matrix, constructed on the basis of available crystal structures, allowed identification of hotspots as critical determinants for RAS-effector interaction. New insights provided by this study are the dissection of the identified hotspots in five distinct regions (R1 to R5) in spite of high sequence variability not only between, but also within, RB/RA domain-containing effectors proteins. Finally, we propose that intermolecular ß-sheet interaction in R1 is a central recognition region while R3 may determine specific contacts of RAS versus RRAS isoforms with effectors.


Asunto(s)
Proteínas Portadoras/metabolismo , GTP Fosfohidrolasas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Aminoácidos , Proteínas Reguladoras de la Apoptosis , Sitios de Unión/genética , Unión Competitiva , Proteínas Portadoras/química , Proteínas Portadoras/genética , Fosfatidilinositol 3-Quinasa Clase I , GTP Fosfohidrolasas/química , GTP Fosfohidrolasas/genética , Humanos , Cinética , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Modelos Moleculares , Proteínas de Unión al GTP Monoméricas/química , Proteínas de Unión al GTP Monoméricas/genética , Fosfatidilinositol 3-Quinasas/química , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica , Dominios Proteicos , Estructura Secundaria de Proteína , Proteínas Proto-Oncogénicas p21(ras)/química , Proteínas Proto-Oncogénicas p21(ras)/genética , Homología de Secuencia de Aminoácido , Transducción de Señal , Factor de Intercambio de Guanina Nucleótido ral/química , Factor de Intercambio de Guanina Nucleótido ral/genética , Factor de Intercambio de Guanina Nucleótido ral/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda