Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
Sci Adv ; 10(25): eadi0707, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38905351

RESUMEN

Ras is a small GTPase that is central to important functional decisions in diverse cell types. An important aspect of Ras signaling is its ability to exhibit bimodal or switch-like activity. We describe the total reconstitution of a receptor-mediated Ras activation-deactivation reaction catalyzed by SOS and p120-RasGAP on supported lipid membrane microarrays. The results reveal a bimodal Ras activation response, which is not a result of deterministic bistability but is rather driven by the distinct processivity of the Ras activator, SOS. Furthermore, the bimodal response is controlled by the condensation state of the scaffold protein, LAT, to which SOS is recruited. Processivity-driven bimodality leads to stochastic bursts of Ras activation even under strongly deactivating conditions. This behavior contrasts deterministic bistability and may be more resistant to pharmacological inhibition.


Asunto(s)
Transducción de Señal , Proteínas ras , Proteínas ras/metabolismo , Proteínas Son Of Sevenless/metabolismo , Humanos
2.
Biochem Biophys Res Commun ; 625: 87-93, 2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-35952612

RESUMEN

Drk, a Drosophila homologue of human GRB2, interacts with Sevenless (Sev) receptor via its SH2 domain, while the N- and C-terminal SH3 domains (Drk-NSH3 and Drk-CSH3, respectively) are responsible for the interaction with proline-rich motifs (PRMs) of Son of sevenless (Sos) or Daughter of Sevenless (Dos). Drk-NSH3 on its own has a conformational equilibrium between folded and unfolded states, and the folded state is stabilised by the association with a Sos-derived proline-rich peptide with PxxPxR motif. In contrast, Drk-CSH3 is supposed to bind PxxxRxxKP motifs in Dos. Aiming at clarifying the structural and functional differences between the two SH3 domains, we performed NMR studies of Drk-CSH3. The resulting solution structure and the 15N-relaxation data showed that Drk-CSH3 consists of a stable domain. Large chemical shift perturbation was commonly found around the RT loop and the hydrophobic patch, while there were also changes that occur characteristically for Sos- or Dos-derived peptides. Sos-derived two peptides with PxxPxR motif showed stronger affinity to Drk-CSH3, indicating that the Sos PRMs can bind both N- and C-SH3 domains. Dos-derived two peptides could also bind Drk-CSH3, but with much weaker affinity, suggesting a possibility that any cooperative binding of Dos-PRMs may strengthen the Drk-Dos interaction. The NMR studies as well as the docking simulations provide valuable insights into the biological and biophysical functions of two SH3 domains in Drk.


Asunto(s)
Drosophila , Dominios Homologos src , Secuencia de Aminoácidos , Animales , Drosophila/metabolismo , Proteína Adaptadora GRB2/metabolismo , Humanos , Núcleo Familiar , Péptidos/metabolismo , Prolina/metabolismo , Unión Proteica , Proteínas Son Of Sevenless/metabolismo
3.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34740968

RESUMEN

Son of Sevenless (SOS) is a Ras guanine nucleotide exchange factor (GEF) that plays a central role in numerous cellular signaling pathways. Like many other signaling molecules, SOS is autoinhibited in the cytosol and activates only after recruitment to the membrane. The mean activation time of individual SOS molecules has recently been measured to be ∼60 s, which is unexpectedly long and seemingly contradictory with cellular signaling timescales, which have been measured to be as fast as several seconds. Here, we rectify this discrepancy using a first-passage time analysis to reconstruct the effective signaling timescale of multiple SOS molecules from their single-molecule activation kinetics. Along with corresponding experimental measurements, this analysis reveals how the functional response time, comprised of many slowly activating molecules, can become substantially faster than the average molecular kinetics. This consequence stems from the enzymatic processivity of SOS in a highly out-of-equilibrium reaction cycle during receptor triggering. Ultimately, rare, early activation events dominate the macroscopic reaction dynamics.


Asunto(s)
Modelos Químicos , Proteínas Son Of Sevenless/metabolismo , Proteínas ras/metabolismo , Activación Enzimática , Cinética , Imagen Individual de Molécula
4.
ACS Synth Biol ; 10(5): 990-999, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33909409

RESUMEN

Intervention in protein-protein interactions (PPIs) has tremendous effects in the molecular therapy of many diseases. To fulfill the requirements for targeting intracellular proteins, here we develop SOS-localization-based interaction screening (SOLIS), which elaborately mimics signaling via the Ras-mitogen-activated protein kinase pathway. SOLIS employs two chimeric proteins in which a membrane localization motif (CaaX) is fused at the C-terminus of a protein of interest and the catalytic domain of SOS is fused at the C-terminus of another protein of interest. Interaction between the two proteins of interest induces membrane localization of the SOS chimera and cell proliferation. Thus, the SOLIS system enables enrichment of superior binders based on cell proliferation in an intracellular PPI-dependent manner. This was verified by three major modalities against intracellular PPIs (small molecules, peptide aptamers, and intrabodies). The system worked over a broad range of affinities (KD = 0.32-140 nM). In a screening of a site-directed randomized library, novel intrabody clones were selected on the basis of the potency of cell proliferation. Three other PPI detection methods (NanoBiT, SPR, and pull-down assays) were employed to characterize the SOLIS system, and several intrabody clones were judged as false negatives in these assays. SOLIS signals would be less sensitive to the orientation/conformation of the chimeric proteins, and this feature emerges as the advantage of SOLIS as a mammalian cytosolic PPI detection system with few false negatives.


Asunto(s)
Espacio Intracelular/metabolismo , Células Precursoras de Linfocitos B/metabolismo , Mapas de Interacción de Proteínas , Transducción de Señal/genética , Proteínas Son Of Sevenless/metabolismo , Animales , Anticuerpos/metabolismo , Antígenos/metabolismo , Aptámeros de Péptidos/metabolismo , Línea Celular , Membrana Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Interleucina-3/genética , Interleucina-3/metabolismo , Interleucina-3/farmacología , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Terapia Molecular Dirigida/métodos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Transducción Genética , Transfección , Proteínas ras/metabolismo
5.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33723061

RESUMEN

Ras is regulated by a specific guanine nucleotide exchange factor Son of Sevenless (SOS), which facilitates the exchange of inactive, GDP-bound Ras with GTP. The catalytic activity of SOS is also allosterically modulated by an active Ras (Ras-GTP). However, it remains poorly understood how oncogenic Ras mutants interact with SOS and modulate its activity. Here, native ion mobility-mass spectrometry is employed to monitor the assembly of the catalytic domain of SOS (SOScat) with KRas and three cancer-associated mutants (G12C, G13D, and Q61H), leading to the discovery of different molecular assemblies and distinct conformers of SOScat engaging KRas. We also find KRasG13D exhibits high affinity for SOScat and is a potent allosteric modulator of its activity. A structure of the KRasG13D•SOScat complex was determined using cryogenic electron microscopy providing insight into the enhanced affinity of the mutant protein. In addition, we find that KRasG13D-GTP can allosterically increase the nucleotide exchange rate of KRas at the active site more than twofold compared to KRas-GTP. Furthermore, small-molecule Ras•SOS disruptors fail to dissociate KRasG13D•SOScat complexes, underscoring the need for more potent disruptors. Taken together, a better understanding of the interaction between oncogenic Ras mutants and SOS will provide avenues for improved therapeutic interventions.


Asunto(s)
Dominio Catalítico , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Son Of Sevenless/metabolismo , Catálisis , Dominio Catalítico/genética , Espectrometría de Masas , Oncogenes , Unión Proteica , Proteínas Son Of Sevenless/química
6.
Biophys J ; 120(7): 1257-1265, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33080222

RESUMEN

Lipid miscibility phase separation has long been considered to be a central element of cell membrane organization. More recently, protein condensation phase transitions, into three-dimensional droplets or in two-dimensional lattices on membrane surfaces, have emerged as another important organizational principle within cells. Here, we reconstitute the linker for activation of T cells (LAT):growth-factor-receptor-bound protein 2 (Grb2):son of sevenless (SOS) protein condensation on the surface of giant unilamellar vesicles capable of undergoing lipid phase separations. Our results indicate that the assembly of the protein condensate on the membrane surface can drive lipid phase separation. This phase transition occurs isothermally and is governed by tyrosine phosphorylation on LAT. Furthermore, we observe that the induced lipid phase separation drives localization of the SOS substrate, K-Ras, into the LAT:Grb2:SOS protein condensate.


Asunto(s)
Lípidos de la Membrana , Proteínas de la Membrana , Proteína Adaptadora GRB2/metabolismo , Proteínas de la Membrana/metabolismo , Fosforilación , Fosfotirosina , Proteínas Son Of Sevenless/metabolismo
7.
Biochim Biophys Acta Rev Cancer ; 1874(2): 188445, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33035641

RESUMEN

SOS1 and SOS2 are the most universal and widely expressed family of guanine exchange factors (GEFs) capable or activating RAS or RAC1 proteins in metazoan cells. SOS proteins contain a sequence of modular domains that are responsible for different intramolecular and intermolecular interactions modulating mechanisms of self-inhibition, allosteric activation and intracellular homeostasis. Despite their homology, analyses of SOS1/2-KO mice demonstrate functional prevalence of SOS1 over SOS2 in cellular processes including proliferation, migration, inflammation or maintenance of intracellular redox homeostasis, although some functional redundancy cannot be excluded, particularly at the organismal level. Specific SOS1 gain-of-function mutations have been identified in inherited RASopathies and various sporadic human cancers. SOS1 depletion reduces tumorigenesis mediated by RAS or RAC1 in mouse models and is associated with increased intracellular oxidative stress and mitochondrial dysfunction. Since WT RAS is essential for development of RAS-mutant tumors, the SOS GEFs may be considered as relevant biomarkers or therapy targets in RAS-dependent cancers. Inhibitors blocking SOS expression, intrinsic GEF activity, or productive SOS protein-protein interactions with cellular regulators and/or RAS/RAC targets have been recently developed and shown preclinical and clinical effectiveness blocking aberrant RAS signaling in RAS-driven and RTK-driven tumors.


Asunto(s)
Mutación , Neoplasias/genética , Proteínas Son Of Sevenless/genética , Proteínas Son Of Sevenless/metabolismo , Regulación Alostérica , Animales , Homeostasis , Humanos , Ratones , Neoplasias/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteínas ras/metabolismo
8.
J Biol Chem ; 295(39): 13651-13663, 2020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-32753483

RESUMEN

Ras family proteins play an essential role in several cellular functions, including growth, differentiation, and survival. The mechanism of action of Ras mutants in Costello syndrome and cancers has been identified, but the contribution of Ras mutants to Noonan syndrome, a genetic disorder that prevents normal development in various parts of the body, is unknown. Son of Sevenless (SOS) is a Ras guanine nucleotide exchange factor. In response to Ras-activating cell signaling, SOS autoinhibition is released and is followed by accelerative allosteric feedback autoactivation. Here, using mutagenesis-based kinetic and pulldown analyses, we show that Noonan syndrome Ras mutants I24N, T50I, V152G, and D153V deregulate the autoactivation of SOS to populate their active form. This previously unknown process has been linked so far only to the development of Noonan syndrome. In contrast, other Noonan syndrome Ras mutants-V14I, T58I, and G60E-populate their active form by deregulation of the previously documented Ras GTPase activities. We propose a novel mechanism responsible for the deregulation of SOS autoactivation, where I24N, T50I, V152G, and D153V Ras mutants evade SOS autoinhibition. Consequently, they are capable of forming a complex with the SOS allosteric site, thus aberrantly promoting SOS autoactivation, resulting in the population of active Ras mutants in cells. The results of this study elucidate the molecular mechanism of the Ras mutant-mediated development of Noonan syndrome.


Asunto(s)
Síndrome de Noonan/metabolismo , Proteínas Son Of Sevenless/metabolismo , Sitio Alostérico , Células HEK293 , Humanos , Cinética , Modelos Moleculares , Mutación , Síndrome de Noonan/genética , Proteínas Son Of Sevenless/química
9.
Curr Genet ; 66(3): 495-499, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31925496

RESUMEN

In all living organisms, genome replication and cell division must be coordinated to produce viable offspring. In the event of DNA damage, bacterial cells employ the SOS response to simultaneously express damage repair systems and halt cell division. Extensive characterization of SOS-controlled cell division inhibition in Escherichia coli has laid the ground for a long-standing paradigm where the cytosolic SulA protein inhibits polymerization of the central division protein, FtsZ, and thereby prevents recruitment of the division machinery at the future division site. Within the last decade, it has become clear that another, likely more general, paradigm exists, at least within the broad group of Gram-positive bacterial species, namely membrane-localized, SOS-induced cell division inhibition. We recently identified such an inhibitor in Staphylococci, SosA, and established a model for SosA-mediated cell division inhibition in Staphylococcus aureus in response to DNA damage. SosA arrests cell division subsequent to the septal localization of FtsZ and later membrane-bound division proteins, while preventing progression to septum closure, leading to synchronization of cells at this particular stage. A membrane-associated protease, CtpA negatively regulates SosA activity and likely allows growth to resume once conditions are favorable. Here, we provide a brief summary of our findings in the context of what already is known for other membrane cell division inhibitors and we emphasize how poorly characterized these intriguing processes are mechanistically. Furthermore, we put some perspective on the relevance of our findings and future developments within the field.


Asunto(s)
Proteínas Bacterianas/metabolismo , Membrana Celular/metabolismo , Respuesta SOS en Genética , Proteínas Son Of Sevenless/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus/crecimiento & desarrollo , Proteínas Bacterianas/genética , División Celular , Proteínas Son Of Sevenless/genética , Staphylococcus/genética , Staphylococcus/metabolismo
10.
Sci Signal ; 12(591)2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31337738

RESUMEN

T cell receptor (TCR) stimulation activates diverse kinase pathways, which include the mitogen-activated protein kinases (MAPKs) ERK and p38, the phosphoinositide 3-kinases (PI3Ks), and the kinase mTOR. Although TCR stimulation activates the p38 pathway through a "classical" MAPK cascade that is mediated by the adaptor protein LAT, it also stimulates an "alternative" pathway in which p38 is activated by the kinase ZAP70. Here, we used dual-parameter, phosphoflow cytometry and in silico computation to investigate how both classical and alternative p38 pathways contribute to T cell activation. We found that basal ZAP70 activation in resting T cell lines reduced the threshold ("primed") TCR-stimulated activation of the classical p38 pathway. Classical p38 signals were reduced after T cell-specific deletion of the guanine nucleotide exchange factors Sos1 and Sos2, which are essential LAT signalosome components. As a consequence of Sos1/2 deficiency, production of the cytokine IL-2 was impaired, differentiation into regulatory T cells was reduced, and the autoimmune disease EAE was exacerbated in mice. These data suggest that the classical and alternative p38 activation pathways exist to generate immune balance.


Asunto(s)
Linfocitos T/citología , Proteína Tirosina Quinasa ZAP-70/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Diferenciación Celular , Pollos , Encefalomielitis Autoinmune Experimental/metabolismo , Activación Enzimática , Femenino , Citometría de Flujo , Humanos , Interleucina-2/metabolismo , Células Jurkat , Cinética , Leucocitos Mononucleares/metabolismo , Ratones , Ratones Noqueados , Unión Proteica , Receptores de Antígenos de Linfocitos T/metabolismo , Proteína SOS1/metabolismo , Proteínas Son Of Sevenless/metabolismo , Procesos Estocásticos , Células TH1/citología , Células Th2/citología
11.
Science ; 363(6431): 1098-1103, 2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30846600

RESUMEN

The guanine nucleotide exchange factor (GEF) Son of Sevenless (SOS) is a key Ras activator that is autoinhibited in the cytosol and activates upon membrane recruitment. Autoinhibition release involves structural rearrangements of the protein at the membrane and thus introduces a delay between initial recruitment and activation. In this study, we designed a single-molecule assay to resolve the time between initial receptor-mediated membrane recruitment and the initiation of GEF activity of individual SOS molecules on microarrays of Ras-functionalized supported membranes. The rise-and-fall shape of the measured SOS activation time distribution and the long mean time scale to activation (~50 seconds) establish a basis for kinetic proofreading in the receptor-mediated activation of Ras. We further demonstrate that this kinetic proofreading is modulated by the LAT (linker for activation of T cells)-Grb2-SOS phosphotyrosine-driven phase transition at the membrane.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína Adaptadora GRB2/metabolismo , Proteínas de la Membrana/metabolismo , Transición de Fase , Proteínas Son Of Sevenless/metabolismo , Proteínas ras/metabolismo , Humanos , Fosfotirosina/metabolismo , Análisis por Matrices de Proteínas , Imagen Individual de Molécula
12.
J Leukoc Biol ; 106(4): 815-822, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30720883

RESUMEN

Circulating neutrophils are, by necessity, quiescent and relatively unresponsive to acute stimuli. In regions of inflammation, mediators can prime neutrophils to react to acute stimuli with stronger proinflammatory, pathogen-killing responses. In neutrophils G protein-coupled receptor (GPCR)-driven proinflammatory responses, such as reactive oxygen species (ROS) formation and accumulation of the key intracellular messenger phosphatidylinositol (3,4,5)-trisphosphate (PIP3 ), are highly dependent on PI3K-γ, a Ras-GTP, and Gßγ coincidence detector. In unprimed cells, the major GPCR-triggered activator of Ras is the Ras guanine nucleotide exchange factor (GEF), Ras guanine nucleotide releasing protein 4 (RasGRP4). Although priming is known to increase GPCR-PIP3 signaling, the mechanisms underlying this augmentation remain unclear. We used genetically modified mice to address the role of the 2 RasGEFs, RasGRP4 and son of sevenless (SOS)1/2, in neutrophil priming. We found that following GM-CSF/TNFα priming, RasGRP4 had only a minor role in the enhanced responses. In contrast, SOS1/2 acquired a substantial role in ROS formation, PIP3 accumulation, and ERK activation in primed cells. These results suggest that SOS1/2 signaling plays a key role in determining the responsiveness of neutrophils in regions of inflammation.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inflamación/patología , Neutrófilos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína SOS1/metabolismo , Proteínas Son Of Sevenless/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas ras/metabolismo , Animales , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ratones , N-Formilmetionina Leucil-Fenilalanina/farmacología , Neutrófilos/efectos de los fármacos , Fosfatos de Fosfatidilinositol/metabolismo , Fosfolipasa C beta/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido ras/metabolismo
13.
Semin Cancer Biol ; 54: 149-161, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29307570

RESUMEN

Inhibition of Ras signalling has been a goal almost since its central role in cell signalling and its deregulation in disease were discovered. Early attempts at inhibiting its post-translational modification using peptidomimetics were successful in cell culture but failed spectacularly in clinical trials, making industry wary of targeting this critical oncoprotein. Small molecule inhibition of the protein-protein interactions involving Ras has also been difficult due to the nature of the interaction interface. Recent improvements in design, synthesis and selection of stabilised peptides, peptidomimetics and macrocycles have suggested that these biologics may represent a new hope in Ras inhibition. Here we review the various ways in which Ras has been targeted with these molecules. We also describe work on related small G proteins of the Ras superfamily, since many of the principles may be applicable to Ras, and these also provide inhibition of pathways downstream of Ras.


Asunto(s)
Descubrimiento de Drogas , Proteínas de Unión al GTP Monoméricas/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Humanos , Proteínas de Unión al GTP Monoméricas/química , Familia de Multigenes , Péptidos/química , Péptidos/metabolismo , Unión Proteica/efectos de los fármacos , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Proteínas Son Of Sevenless/química , Proteínas Son Of Sevenless/metabolismo , Relación Estructura-Actividad , Proteínas ras/química , Proteínas ras/genética , Proteínas ras/metabolismo
14.
Artículo en Inglés | MEDLINE | ID: mdl-29610148

RESUMEN

The guanine-nucleotide exchange factor (GEF) Son-of-Sevenless (SOS) plays a critical role in metazoan signaling by converting Ras•GDP (guanosine diphosphate) to Ras•GTP (guanosine triphosphate) in response to tyrosine kinase activation. Structural studies have shown that SOS differs from other Ras-specific GEFs in that SOS is itself activated by Ras•GTP binding to an allosteric site, distal to the site of nucleotide exchange. The activation of SOS involves membrane recruitment and conformational changes, triggered by lipid binding, that open the allosteric binding site for Ras•GTP. This is in contrast to other Ras-specific GEFs, which are activated by second messengers that more directly affect the active site. Allosteric Ras•GTP binding stabilizes SOS at the membrane, where it can turn over other Ras molecules processively, leading to an ultrasensitive response that is distinct from that of other Ras-specific GEFs.


Asunto(s)
Membrana Celular/metabolismo , Guanosina Difosfato/metabolismo , Proteínas ras/metabolismo , ras-GRF1/metabolismo , Sitio Alostérico , Animales , Humanos , Proteínas Son Of Sevenless/metabolismo
15.
Sci Signal ; 11(546)2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30181243

RESUMEN

About a third of tumors have activating mutations in HRAS, NRAS, or KRAS, genes encoding guanosine triphosphatases (GTPases) of the RAS family. In these tumors, wild-type RAS cooperates with mutant RAS to promote downstream effector activation and cell proliferation and transformation, suggesting that upstream activators of wild-type RAS are important modulators of mutant RAS-driven oncogenesis. The guanine nucleotide exchange factor (GEF) SOS1 mediates KRAS-driven proliferation, but little is understood about the role of SOS2. We found that RAS family members have a hierarchical requirement for the expression and activity of SOS2 to drive cellular transformation. In mouse embryonic fibroblasts (MEFs), SOS2 critically mediated mutant KRAS-driven, but not HRAS-driven, transformation. Sos2 deletion reduced epidermal growth factor (EGF)-dependent activation of wild-type HRAS and phosphorylation of the kinase AKT in cells expressing mutant RAS isoforms. Assays using pharmacological inhibitors revealed a hierarchical requirement for signaling by phosphoinositide 3-kinase (PI3K) in promoting RAS-driven cellular transformation that mirrored the requirement for SOS2. KRAS-driven transformation required the GEF activity of SOS2 and was restored in Sos2-/- MEFs by expression of constitutively activated PI3K. Finally, CRISPR/Cas9-mediated deletion of SOS2 reduced EGF-stimulated AKT phosphorylation and synergized with MEK inhibition to revert the transformed phenotype of human KRAS mutant pancreatic and lung tumor cells. These results indicate that SOS2-dependent PI3K signaling mediates mutant KRAS-driven transformation, revealing therapeutic targets in KRAS-driven cancers. Our data also reveal the importance of three-dimensional culture systems in investigating the mediators of mutant KRAS.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Fibroblastos/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Son Of Sevenless/metabolismo , Animales , Transformación Celular Neoplásica/genética , Células Cultivadas , Embrión de Mamíferos/citología , Fibroblastos/citología , Humanos , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Transducción de Señal/genética , Proteínas Son Of Sevenless/genética
16.
Mol Cell Biol ; 38(16)2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29844066

RESUMEN

Using Sos1 knockout (Sos1-KO), Sos2-KO, and Sos1/2 double-knockout (Sos1/2-DKO) mice, we assessed the functional role of Sos1 and Sos2 in skin homeostasis under physiological and/or pathological conditions. Sos1 depletion resulted in significant alterations of skin homeostasis, including reduced keratinocyte proliferation, altered hair follicle and blood vessel integrity in dermis, and reduced adipose tissue in hypodermis. These defects worsened significantly when both Sos1 and Sos2 were absent. Simultaneous Sos1/2 disruption led to severe impairment of the ability to repair skin wounds, as well as to almost complete ablation of the neutrophil-mediated inflammatory response in the injury site. Furthermore, Sos1 disruption delayed the onset of tumor initiation, decreased tumor growth, and prevented malignant progression of papillomas in a DMBA (7,12-dimethylbenz[α]anthracene)/TPA (12-O-tetradecanoylphorbol-13-acetate)-induced skin carcinogenesis model. Finally, Sos1 depletion in preexisting chemically induced papillomas resulted also in decreased tumor growth, probably linked to significantly reduced underlying keratinocyte proliferation. Our data unveil novel, distinctive mechanistic roles of Sos 1 and Sos2 in physiological control of skin homeostasis and wound repair, as well as in pathological development of chemically induced skin tumors. These observations underscore the essential role of Sos proteins in cellular proliferation and migration and support the consideration of these RasGEFs as potential biomarkers/therapy targets in Ras-driven epidermal tumors.


Asunto(s)
Proteína SOS1/metabolismo , Neoplasias Cutáneas/etiología , Piel/metabolismo , Proteínas Son Of Sevenless/metabolismo , Animales , Carcinogénesis , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Homeostasis , Ratones , Ratones Noqueados , Neovascularización Fisiológica , Papiloma/metabolismo , Papiloma/patología , Proteína SOS1/deficiencia , Proteína SOS1/genética , Piel/irrigación sanguínea , Piel/citología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Proteínas Son Of Sevenless/deficiencia , Proteínas Son Of Sevenless/genética , Cicatrización de Heridas
17.
PLoS One ; 13(4): e0195513, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29630631

RESUMEN

Cell signaling is the process by which extracellular information is transmitted into the cell to perform useful biological functions. The ERK (extracellular-signal-regulated kinase) signaling controls several cellular processes such as cell growth, proliferation, differentiation and apoptosis. The ERK signaling pathway considered in this work starts with an extracellular stimulus and ends with activated (double phosphorylated) ERK which gets translocated into the nucleus. We model and analyze this complex pathway by decomposing it into three functional subsystems. The first subsystem spans the initial part of the pathway from the extracellular growth factor to the formation of the SOS complex, ShC-Grb2-SOS. The second subsystem includes the activation of Ras which is mediated by the SOS complex. This is followed by the MAPK subsystem (or the Raf-MEK-ERK pathway) which produces the double phosphorylated ERK upon being activated by Ras. Although separate models exist in the literature at the subsystems level, a comprehensive model for the complete system including the important regulatory feedback loops is missing. Our dynamic model combines the existing subsystem models and studies their steady-state and dynamic interactions under feedback. We establish conditions under which bistability and oscillations exist for this important pathway. In particular, we show how the negative and positive feedback loops affect the dynamic characteristics that determine the cellular outcome.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Modelos Biológicos , Animales , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Retroalimentación Fisiológica , Humanos , Cinética , Fosforilación , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Son Of Sevenless/metabolismo , Biología de Sistemas , Proteínas ras/metabolismo
18.
Cell Physiol Biochem ; 44(5): 2057-2072, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29241165

RESUMEN

BACKGROUND/AIMS: Self-renewal is one of the most important features of embryonic stem (ES) cells. SC1 is a small molecule modulator that effectively maintains the self-renewal of mouse ES cells in the absence of leukemia inhibitory factor (LIF), serum and feeder cells. However, the mechanism by which SC1 maintains the undifferentiated state of mouse ES cells remains unclear. METHODS: In this study, microarray and small RNA deep-sequencing experiments were performed on mouse ES cells treated with or without SC1 to identify the key genes and microRNAs that contributed to self-renewal. RESULTS: SC1 regulates the expressions of pluripotency and differentiation factors, and antagonizes the retinoic acid (RA)-induced differentiation in the presence or absence of LIF. SC1 inhibits the MEK/ERK pathway through Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis and pathway reporting experiments. Small RNA deep-sequencing revealed that SC1 significantly modulates the expression of multiple microRNAs with crucial functions in ES cells. The expression of miR124-3p is upregulated in SC1-treated ES cells, which significantly inhibits the MEK/ERK pathway by targeting Grb2, Sos2 and Egr1. CONCLUSION: SC1 enhances the self-renewal capacity of mouse ES cells by modulating the expression of key regulatory genes and pluripotency-associated microRNAs. SC1 significantly upregulates miR124-3p expression to further inhibit the MEK/ ERK pathway by targeting Grb2, Sos2 and Egr1.


Asunto(s)
Autorrenovación de las Células/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , MicroARNs/metabolismo , Células Madre Embrionarias de Ratones/citología , Pirazoles/farmacología , Pirimidinas/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Proteína 1 de la Respuesta de Crecimiento Precoz/antagonistas & inhibidores , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteína Adaptadora GRB2/antagonistas & inhibidores , Proteína Adaptadora GRB2/metabolismo , Factor Inhibidor de Leucemia/química , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , MicroARNs/química , MicroARNs/genética , Células Madre Embrionarias de Ratones/metabolismo , Análisis de Secuencia de ARN , Proteínas Son Of Sevenless/antagonistas & inhibidores , Proteínas Son Of Sevenless/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Tretinoina/farmacología , Regulación hacia Arriba/efectos de los fármacos
19.
Int J Mol Sci ; 18(9)2017 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-28895882

RESUMEN

Salt stress is one of the major abiotic stresses that severely impact plant growth and development. In this study, we investigated the physiological and transcriptomic responses of Chinese cabbage "Qingmaye" to salt stress, a main variety in North China. Our results showed that the growth and photosynthesis of Chinese cabbage were significantly inhibited by salt treatment. However, as a glycophyte, Chinese cabbage could cope with high salinity; it could complete an entire life cycle at 100 mM NaCl. The high salt tolerance of Chinese cabbage was achieved by accumulating osmoprotectants and by maintaining higher activity of antioxidant enzymes. Transcriptomic responses were analyzed using the digital gene expression profiling (DGE) technique after 12 h of treatment by 200 mM NaCl. A total of 1235 differentially expressed genes (DEGs) including 740 up- and 495 down-regulated genes were identified. Functional annotation analyses showed that the DEGs were related to signal transduction, osmolyte synthesis, transcription factors, and antioxidant proteins. Taken together, this study contributes to our understanding of the mechanism of salt tolerance in Chinese cabbage and provides valuable information for further improvement of salt tolerance in Chinese cabbage breeding programs.


Asunto(s)
Brassica rapa/fisiología , Tolerancia a la Sal/fisiología , Estrés Fisiológico/fisiología , Transcriptoma , Antioxidantes/metabolismo , Brassica rapa/genética , Brassica rapa/crecimiento & desarrollo , China , Clorofila/análisis , Perfilación de la Expresión Génica , Regulación de la Expresión Génica de las Plantas , Genoma de Planta , Estadios del Ciclo de Vida , Fotosíntesis/genética , Salinidad , Tolerancia a la Sal/genética , Transducción de Señal , Cloruro de Sodio , Proteínas Son Of Sevenless/genética , Proteínas Son Of Sevenless/metabolismo , Estrés Fisiológico/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
Sci Rep ; 7(1): 3687, 2017 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-28623374

RESUMEN

Simple reversible competitive inhibition of nucleotide binding of GTP to Ras family GTPases has long been recognized as an unlikely approach to manipulating the activity of such proteins for experimental or therapeutic purposes. This is due to the high affinity of GTP to GTPases coupled with high cellular GTP concentrations, but also to problems of specificity for the highly conserved binding sites in GTPases. A recent approach suggested that these problems might be overcome by using GDP derivatives that can undergo a covalent reaction with disease specific mutants, in particular addressing inhibition of KRasG12C using GDP equipped with an electrophilic group at the ß-phosphate. We show here that a major drawback to this approach is a loss of reversible affinity of such ß-modified derivatives for Ras of at least 104 compared to GTP and GDP. With the help of a thorough kinetic characterization, we show that this leads to covalent reaction times that are too slow to make the compounds attractive for intracellular use, but that generation of a hypothetical reactive GDP derivative that retains the high reversible affinity of GDP/GTP to Ras might be a viable alternative.


Asunto(s)
Guanosina Trifosfato/metabolismo , Nucleótidos/metabolismo , Nucleótidos/farmacología , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Acetamidas/química , Acetamidas/metabolismo , Acetamidas/farmacología , Guanosina Difosfato/análogos & derivados , Guanosina Difosfato/química , Guanosina Difosfato/metabolismo , Guanosina Difosfato/farmacología , Guanosina Trifosfato/química , Cinética , Modelos Biológicos , Estructura Molecular , Nucleótidos/química , Unión Proteica , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Recombinantes , Proteínas Son Of Sevenless/química , Proteínas Son Of Sevenless/metabolismo , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...