Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 524
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35101974

RESUMEN

Cells control the properties of the cytoplasm to ensure proper functioning of biochemical processes. Recent studies showed that cytoplasmic density varies in both physiological and pathological states of cells undergoing growth, division, differentiation, apoptosis, senescence, and metabolic starvation. Little is known about how cellular processes cope with these cytoplasmic variations. Here, we study how a cell cycle oscillator comprising cyclin-dependent kinase (Cdk1) responds to changes in cytoplasmic density by systematically diluting or concentrating cycling Xenopus egg extracts in cell-like microfluidic droplets. We found that the cell cycle maintains robust oscillations over a wide range of deviations from the endogenous density: as low as 0.2× to more than 1.22× relative cytoplasmic density (RCD). A further dilution or concentration from these values arrested the system in a low or high steady state of Cdk1 activity, respectively. Interestingly, diluting an arrested cytoplasm of 1.22× RCD recovers oscillations at lower than 1× RCD. Thus, the cell cycle switches reversibly between oscillatory and stable steady states at distinct thresholds depending on the direction of tuning, forming a hysteresis loop. We propose a mathematical model which recapitulates these observations and predicts that the Cdk1/Wee1/Cdc25 positive feedback loops do not contribute to the observed robustness, supported by experiments. Our system can be applied to study how cytoplasmic density affects other cellular processes.


Asunto(s)
Relojes Biológicos , Citoplasma/metabolismo , Modelos Biológicos , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Citoplasma/genética , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis , ras-GRF1/genética , ras-GRF1/metabolismo
2.
Behav Genet ; 52(4-5): 281-297, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35988119

RESUMEN

We have previously shown that high runner (HR) mice (from a line genetically selected for increased wheel-running behavior) have distinct, genetically based, neurobiological phenotypes as compared with non-selected control (C) mice. However, developmental programming effects during early life, including maternal care and parent-of-origin-dependent expression of imprinted genes, can also contribute to variation in physical activity. Here, we used cross-fostering to address two questions. First, do HR mice have altered DNA methylation profiles of imprinted genes in the brain compared to C mice? Second, does maternal upbringing further modify the DNA methylation status of these imprinted genes? To address these questions, we cross-fostered all offspring at birth to create four experimental groups: C pups to other C dams, HR pups to other HR dams, C pups to HR dams, and HR pups to C dams. Bisulfite sequencing of 16 imprinted genes in the cortex and hippocampus revealed that the HR line had altered DNA methylation patterns of the paternally imprinted genes, Rasgrf1 and Zdbf2, as compared with the C line. Both fostering between the HR and C lines and sex modified the DNA methylation profiles for the paternally expressed genes Mest, Peg3, Igf2, Snrpn, and Impact. Ig-DMR, a gene with multiple paternal and maternal imprinted clusters, was also affected by maternal upbringing and sex. Our results suggest that differential methylation patterns of imprinted genes in the brain could contribute to evolutionary increases in wheel-running behavior and are also dependent on maternal upbringing and sex.


Asunto(s)
Metilación de ADN , Impresión Genómica , Animales , Metilación de ADN/genética , Impresión Genómica/genética , Hipocampo , Ratones , ras-GRF1/genética
3.
Biosci Biotechnol Biochem ; 86(9): 1318-1326, 2022 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-35749464

RESUMEN

Proline is a predominant amino acid in grape must, but it is poorly utilized by the yeast Saccharomyces cerevisiae in wine-making processes. This sometimes leads to a nitrogen deficiency during fermentation and proline accumulation in wine. In this study, we clarified that a glucose response is involved in an inhibitory mechanism of proline utilization in yeast. Our genetic screen showed that strains with a loss-of-function mutation on the CDC25 gene can utilize proline even under fermentation conditions. Cdc25 is a regulator of the glucose response consisting of the Ras/cAMP-dependent protein kinase A (PKA) pathway. Moreover, we found that activation of the Ras/PKA pathway is necessary for the inhibitory mechanism of proline utilization. The present data revealed that crosstalk exists between the carbon and proline metabolisms. Our study could hold promise for the development of wine yeast strains that can efficiently assimilate proline during the fermentation processes.


Asunto(s)
Prolina , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Vino , ras-GRF1 , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fermentación , Glucosa/metabolismo , Mutación con Pérdida de Función , Prolina/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Transducción de Señal , Vino/microbiología , ras-GRF1/genética
4.
Neurobiol Dis ; 157: 105423, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34144125

RESUMEN

BACKGROUND: Dravet syndrome is a rare, severe pediatric epileptic encephalopathy associated with intellectual and motor disabilities. Proteomic profiling in a mouse model of Dravet syndrome can provide information about the molecular consequences of the genetic deficiency and about pathophysiological mechanisms developing during the disease course. METHODS: A knock-in mouse model of Dravet syndrome with Scn1a haploinsufficiency was used for whole proteome, seizure, and behavioral analysis. Hippocampal tissue was dissected from two- (prior to epilepsy manifestation) and four- (following epilepsy manifestation) week-old male mice and analyzed using LC-MS/MS with label-free quantification. Proteomic data sets were subjected to bioinformatic analysis including pathway enrichment analysis. The differential expression of selected proteins was confirmed by immunohistochemical staining. RESULTS: The findings confirmed an increased susceptibility to hyperthermia-associated seizures, the development of spontaneous seizures, and behavioral alterations in the novel Scn1a-A1873V mouse model of Dravet syndrome. As expected, proteomic analysis demonstrated more pronounced alterations following epilepsy manifestation. In particular, proteins involved in neurotransmitter dynamics, receptor and ion channel function, synaptic plasticity, astrogliosis, neoangiogenesis, and nitric oxide signaling showed a pronounced regulation in Dravet mice. Pathway enrichment analysis identified several significantly regulated pathways at the later time point, with pathways linked to synaptic transmission and glutamatergic signaling dominating the list. CONCLUSION: In conclusion, the whole proteome analysis in a mouse model of Dravet syndrome demonstrated complex molecular alterations in the hippocampus. Some of these alterations may have an impact on excitability or may serve a compensatory function, which, however, needs to be further confirmed by future investigations. The proteomic data indicate that, due to the molecular consequences of the genetic deficiency, the pathophysiological mechanisms may become more complex during the course of the disease. As a result, the management of Dravet syndrome may need to consider further molecular and cellular alterations. Ensuing functional follow-up studies, this data set may provide valuable guidance for the future development of novel therapeutic approaches.


Asunto(s)
Epilepsias Mioclónicas/metabolismo , Hipocampo/metabolismo , Proteómica , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Animales , Conducta Animal , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Ligasas de Carbono-Nitrógeno/metabolismo , Cromatografía Liquida , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Prueba de Laberinto Elevado , Epilepsias Mioclónicas/genética , Epilepsias Mioclónicas/fisiopatología , Femenino , Técnicas de Sustitución del Gen , Gliosis , Haploinsuficiencia , Hipertermia/fisiopatología , Inmunohistoquímica , Masculino , Ratones , Canal de Sodio Activado por Voltaje NAV1.1/genética , Neovascularización Fisiológica , Plasticidad Neuronal , Óxido Nítrico , Prueba de Campo Abierto , Prueba de Desempeño de Rotación con Aceleración Constante , Transducción de Señal , Conducta Social , Transmisión Sináptica , Espectrometría de Masas en Tándem , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , ras-GRF1/metabolismo
5.
Int J Mol Sci ; 22(23)2021 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-34884741

RESUMEN

ATP, one of the signaling molecules most commonly secreted in the nervous system and capable of stimulating multiple pathways, binds to the ionotropic purinergic receptors, in particular, the P2X7 receptor (P2X7R) and stimulates neuronal cell death. Given this effect of purinergic receptors on the viability of dopaminergic neurons model cells and that Ras GTPases control Erk1/2-regulated mitogen-activated cell proliferation and survival, we have investigated the role of the small GTPases of the Ras superfamily, together with their regulatory and effector molecules as the potential molecular intermediates in the P2X7R-regulated cell death of SN4741 dopaminergic neurons model cells. Here, we demonstrate that the neuronal response to purinergic stimulation involves the Calmodulin/RasGRF1 activation of the small GTPase Ras and Erk1/2. We also demonstrate that tyrosine phosphatase PTPRß and other tyrosine phosphatases regulate the small GTPase activation pathway and neuronal viability. Our work expands the knowledge on the intracellular responses of dopaminergic cells by identifying new participating molecules and signaling pathways. In this sense, the study of the molecular circuitry of these neurons is key to understanding the functional effects of ATP, as well as considering the importance of these cells in Parkinson's Disease.


Asunto(s)
Neuronas Dopaminérgicas/enzimología , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Animales , Señalización del Calcio , Línea Celular , Supervivencia Celular , Activación Enzimática , Ratones , Enfermedad de Parkinson/enzimología , Proteínas ras/metabolismo , ras-GRF1/metabolismo
6.
J Neurosci ; 39(32): 6325-6338, 2019 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-31182637

RESUMEN

Ras/Raf/MEK/ERK (Ras-ERK) signaling has been implicated in the effects of drugs of abuse. Inhibitors of MEK1/2, the kinases upstream of ERK1/2, have been critical in defining the role of the Ras-ERK cascade in drug-dependent alterations in behavioral plasticity, but the Ras family of small GTPases has not been extensively examined in drug-related behaviors. We examined the role of Ras Guanine Nucleotide Releasing Factor 1 (RasGRF1) and 2 (RasGRF2), upstream regulators of the Ras-ERK signaling cascade, on cocaine self-administration (SA) in male mice. We first established a role for Ras-ERK signaling in cocaine SA, demonstrating that pERK1/2 is upregulated following SA in C57BL/6N mice in striatum. We then compared RasGRF1 and RasGRF2 KO mouse lines, demonstrating that cocaine SA in RasGRF2 KO mice was increased relative to WT controls, whereas RasGRF1 KO and WT mice did not differ. This effect in RasGRF2 mice is likely mediated by the Ras-ERK signaling pathway, as pERK1/2 upregulation following cocaine SA was absent in RasGRF2 KO mice. Interestingly, the lentiviral knockdown of RasGRF2 in the NAc had the opposite effect to that in RasGRF2 KO mice, reducing cocaine SA. We subsequently demonstrated that the MEK inhibitor PD325901 administered peripherally prior to cocaine SA increased cocaine intake, replicating the increase seen in RasGRF2 KO mice, whereas PD325901 administered into the NAc decreased cocaine intake, similar to the effect seen following lentiviral knockdown of RasGRF2. These data indicate a role for RasGRF2 in cocaine SA in mice that is ERK-dependent, and suggest a differential effect of global versus site-specific RasGRF2 inhibition.SIGNIFICANCE STATEMENT Exposure to drugs of abuse activates a variety of intracellular pathways, and following repeated exposure, persistent changes in these pathways contribute to drug dependence. Downstream components of the Ras-ERK signaling cascade are involved in the acute and chronic effects of drugs of abuse, but their upstream mediators have not been extensively characterized. Here we show, using a combination of molecular, pharmacological, and lentiviral techniques, that the guanine nucleotide exchange factor RasGRF2 mediates cocaine self-administration via an ERK-dependent mechanism, whereas RasGRF1 has no effect on responding for cocaine. These data indicate dissociative effects of mediators of Ras activity on cocaine reward and expand the understanding of the contribution of Ras-ERK signaling to drug-taking behavior.


Asunto(s)
Trastornos Relacionados con Cocaína/fisiopatología , Cocaína/farmacología , Cuerpo Estriado/fisiopatología , Sistema de Señalización de MAP Quinasas/fisiología , Recompensa , Factores de Intercambio de Guanina Nucleótido ras/fisiología , Acetilación , Animales , Benzamidas/farmacología , Cocaína/administración & dosificación , Condicionamiento Operante , Cuerpo Estriado/efectos de los fármacos , Difenilamina/análogos & derivados , Difenilamina/farmacología , Técnicas de Silenciamiento del Gen , Vectores Genéticos/genética , Histonas/metabolismo , Lentivirus/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/fisiopatología , Especificidad de Órganos , Fosforilación , Procesamiento Proteico-Postraduccional , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Autoadministración , Factores de Intercambio de Guanina Nucleótido ras/deficiencia , Factores de Intercambio de Guanina Nucleótido ras/genética , ras-GRF1/deficiencia , ras-GRF1/genética , ras-GRF1/fisiología
7.
Int J Cancer ; 146(12): 3369-3378, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32159858

RESUMEN

Prostate cancer is a heterogeneous disease and optimum gene targeting treatment is often impermissible. We aim to determine the intratumoral genomic heterogeneity of prostate cancer and explore candidate genes for targeted therapy. Exome sequencing was performed on 37 samples from 16 patients with prostate cancer. Somatic variant analysis, copy number variant (CNV) analysis, clonal evolution analysis and variant spectrum analysis were used to study the intratumoral genomic heterogeneity and genetic characteristics of metastatic prostate cancer. Our study confirmed the high intratumoral genetic heterogeneity of prostate cancer in many aspects, including number of shared variants, tumor mutation burden (TMB), variant genes, CNV burden, weighted genome instability index (wGII), CNV profiles, clonal evolutionary process, variant spectrum and mutational signatures. Moreover, we identified several common genetic characteristics of prostate cancer. Alterations of DNA damage repair genes, RTK/RAS pathway associated gene RASGRF1 and autophagy gene EPG5 may be involved in tumorigenesis in prostate cancer. CNV burden and DNA damage repair (DDR) genes may be associated with metastasis of prostate cancer.


Asunto(s)
Biomarcadores de Tumor/genética , Heterogeneidad Genética , Neoplasias de la Próstata/genética , Anciano , Anciano de 80 o más Años , Autofagia/genética , Proteínas Relacionadas con la Autofagia/genética , Evolución Clonal , Variaciones en el Número de Copia de ADN , Reparación del ADN/genética , Humanos , Masculino , Persona de Mediana Edad , Mutación , Polimorfismo de Nucleótido Simple , Próstata/patología , Próstata/cirugía , Prostatectomía , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/cirugía , Proteínas de Transporte Vesicular/genética , Secuenciación del Exoma , ras-GRF1/genética
8.
J Cell Sci ; 131(17)2018 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-30072442

RESUMEN

The mitotic spindle is a very dynamic structure that is built de novo and destroyed at each round of cell division. In order to perform its fundamental function during chromosome segregation, mitotic spindle dynamics must be tightly coordinated with other cell cycle events. These changes are driven by several protein kinases, phosphatases and microtubule-associated proteins. In budding yeast, the kinase Swe1 and the phosphatase Mih1 act in concert in controlling the phosphorylation state of Cdc28, the catalytic subunit of Cdk1, the major regulator of the cell cycle. In this study we show that Swe1 and Mih1 are also involved in the control of mitotic spindle dynamics. Our data indicate that Swe1 and the Polo-like kinase Cdc5 control the balance between phosphorylated and unphosphorylated forms of Mih1, which is, in turn, important for mitotic spindle elongation. Moreover, we show that the microtubule-associated protein Bik1 is a phosphoprotein, and that Swe1 and Mih1 are both involved in controlling phosphorylation of Bik1. These results uncover new players and provide insights into the complex regulation of mitotic spindle dynamics.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Mitosis , Proteínas Tirosina Quinasas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Huso Acromático/metabolismo , ras-GRF1/metabolismo , Proteína Quinasa CDC28 de Saccharomyces cerevisiae/genética , Proteína Quinasa CDC28 de Saccharomyces cerevisiae/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/genética , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Transducción de Señal , Huso Acromático/genética , ras-GRF1/genética
9.
Proc Natl Acad Sci U S A ; 114(10): 2693-2698, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28213494

RESUMEN

Regulation of integrins is critical for lymphocyte adhesion to endothelium and migration throughout the body. Inside-out signaling to integrins is mediated by the small GTPase Ras-proximate-1 (Rap1). Using an RNA-mediated interference screen, we identified phospholipase Cε 1 (PLCε1) as a crucial regulator of stromal cell-derived factor 1 alpha (SDF-1α)-induced Rap1 activation. We have shown that SDF-1α-induced activation of Rap1 is transient in comparison with the sustained level following cross-linking of the antigen receptor. We identified that PLCε1 was necessary for SDF-1α-induced adhesion using shear stress, cell morphology alterations, and crawling on intercellular adhesion molecule 1 (ICAM-1)-expressing cells. Structure-function experiments to separate the dual-enzymatic function of PLCε1 uncover necessary contributions of the CDC25, Pleckstrin homology, and Ras-associating domains, but not phospholipase activity, to this pathway. In the mouse model of delayed type hypersensitivity, we have shown an essential role for PLCε1 in T-cell migration to inflamed skin, but not for cytokine secretion and proliferation in regional lymph nodes. Our results reveal a signaling pathway where SDF-1α induces T-cell adhesion through activation of PLCε1, suggesting that PLCε1 is a specific potential target in treating conditions involving migration of T cells to inflamed organs.


Asunto(s)
Quimiocina CXCL12/genética , Inflamación/genética , Fosfoinositido Fosfolipasa C/genética , Proteínas de Unión a Telómeros/genética , Animales , Adhesión Celular/genética , Adhesión Celular/inmunología , Movimiento Celular/genética , Movimiento Celular/inmunología , Quimiocina CXCL12/inmunología , Humanos , Inflamación/inmunología , Inflamación/patología , Molécula 1 de Adhesión Intercelular/inmunología , Linfocitos/inmunología , Linfocitos/patología , Ratones , Fosfoinositido Fosfolipasa C/inmunología , Receptores de Antígenos/genética , Receptores de Antígenos/inmunología , Complejo Shelterina , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/patología , Proteínas de Unión a Telómeros/inmunología , ras-GRF1/inmunología
10.
J Cell Physiol ; 234(12): 22985-22995, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31245854

RESUMEN

N-methyl-D-aspartate receptors (NMDARs) that contain the NR2A and NR2B subunits play a critical role in neuronal plasticity and dendritogenesis. Gain-and-loss-of function studies indicate that NR2B, but not NR2A, promotes dendritic branching. Accumulating evidence indicates that stimulation of NMDARs activates NADPH oxidase (NOX2), thereby generating superoxide. However, the molecular underpinnings of this process are not understood. RasGRF1, a guanine nucleotide exchange factor, is key for several forms of neuronal plasticity and interacts directly with the tail of NR2B. We investigated whether the NR2B-NMDAR/RasGRF1 pathway regulates the activity of NOX2 and whether superoxide production is required for dendritogenesis. We measured superoxide production in developing primary cultures of hippocampal neurons from 3 to 25 days in vitro (DIV) with the probe dihydroethidium (dHE). We found the highest dHE levels at early and intermediate developmental stages (3-15 DIV), when the NR2B-NMDAR expression is abundant. During these early/intermediate developmental stages, but not in mature neurons (>15 DIV), NMDAR activity is required for superoxide production. We also found that disrupting the NR2B-RasGRF1 interaction led to reduced dHE fluorescence intensity and moreover inhibited dendritic branching in hippocampal neurons. Together, our data indicate that superoxide production is induced by the NR2B-NMDARs/RasGRF1/NOX2 pathway and promotes dendritogenesis.


Asunto(s)
NADPH Oxidasa 2/genética , Neurogénesis/genética , Receptores de N-Metil-D-Aspartato/genética , ras-GRF1/genética , Animales , Dendritas/metabolismo , Células Dendríticas/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Plasticidad Neuronal/genética , Neuronas/metabolismo , Ratas , Transducción de Señal/genética , Superóxidos/metabolismo
11.
Immunogenetics ; 71(1): 35-47, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30269158

RESUMEN

A decrease in the incidence of bovine mastitis, the costliest disease in the dairy industry, can be facilitated through genetic marker-assisted selective breeding programs. Identification of genomic variants associated with mastitis resistance is an ongoing endeavor for which genome-wide association studies (GWAS) using high-density arrays provide a valuable tool. We identified single nucleotide polymorphisms (SNPs) in Holstein dairy cattle associated with mastitis resistance in a GWAS by using a high-density SNP array. Mastitis-resistant (15) and mastitis-susceptible (28) phenotypic extremes were identified from 224 lactating dairy cows on commercial dairy farm located in Utah based on multiple criteria of mastitis resistance over an 8-month period. Twenty-seven quantitative trait loci (QTLs) for mastitis resistance were identified based on 117 SNPs suggestive of genome-wide significance for mastitis resistance (p ≤ 1 × 10-4), including 10 novel QTLs. Seventeen QTLs overlapped previously reported QTLs of traits relevant to mastitis, including four QTLs for teat length. One QTL includes the RAS guanyl-releasing protein 1 gene (RASGRP1), a candidate gene for mastitis resistance. This GWAS identifies 117 candidate SNPs and 27 QTLs for mastitis resistance using a selective genotyping approach, including 10 novel QTLs. Based on overlap with previously identified QTLs, teat length appears to be an important trait in mastitis resistance. RASGRP1, overlapped by one QTL, is a candidate gene for mastitis resistance.


Asunto(s)
Estudio de Asociación del Genoma Completo , Técnicas de Genotipaje , Mastitis Bovina/genética , ras-GRF1/genética , Animales , Bovinos , Femenino , Fenotipo , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo
12.
Immunol Cell Biol ; 97(10): 877-887, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31361349

RESUMEN

Previously, we reported induced expression of the p190 Rho guanine nucleotide exchange factor (p190RhoGEF, ARHGEF28) following CD40 stimulation of B cells isolated from mouse spleen. We also reported that p190RhoGEF and a downstream effector molecule RhoA are required for B-cell differentiation, especially for the induction of the plasma cell (PC) differentiation. This study investigates the role of p190RhoGEF in B-cell biology in vivo, using p190RhoGEF transgenic (TG) mice that overexpress a wild-type full gene in B cells. Immunization of these mice with T-cell-dependent antigen showed that populations of germinal center B cells and PCs were significantly increased in TG mice. Furthermore, similar results were shown in recombination activating 1 (Rag1) knockout mice that were reconstituted with B cells isolated from TG mice in combination with T cells isolated from littermate control mice. Analyses of isotype class switching and transcription factors involved in a germinal center reaction and PC differentiation also supported the findings from the cellular responses. These results suggest that p190RhoGEF may play a role in the stage of PC differentiation during T-cell-dependent humoral immune responses.


Asunto(s)
Linfocitos B/inmunología , Centro Germinal/inmunología , Inmunidad Humoral , Activación de Linfocitos/inmunología , Linfocitos T/inmunología , ras-GRF1/metabolismo , Traslado Adoptivo , Animales , Diferenciación Celular , Proteínas de Homeodominio/metabolismo , Cambio de Clase de Inmunoglobulina , Ratones Transgénicos , Células Plasmáticas/citología , Células Plasmáticas/metabolismo , Bazo/metabolismo
13.
Neurochem Res ; 43(10): 2000-2007, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30242550

RESUMEN

Epileptogenesis, induced by status epilepticus (SE), is a chronic process, and intervention in this progress may prevent chronic epilepsy. It has been proposed that DNA methylation might be related with epileptogenesis. RASgrf1 has a differentially methylated region at the promoter which can silence gene expression. We have previously observed the down-regulation of RASgrf1 in epilepsy patients and proved that hypermethylation of RASgrf1 reaches maximal level at the latent period in mice after kainate-induced SE (KA mice), with corresponding alteration of RASgrf1 expression. In the present study, N-phthalyl-L-tryptophan (RG108), a DNA methyltransferase inhibitor, was applied in KA mice at latent phase and the behavior, electroencephalogram and pathological changes were observed in chronic phase. Methylation and expression of RASgrf1 were determined by polymerase chain reaction (PCR), western blotting, and bisulfite sequencing PCR. The results showed that the incidence of spontaneous recurrent seizures (SRS) was significantly lower in the RG108 group than the normal saline (NS) group. Subgroup analysis showed significant hypermethylation and lower expression of RASgrf1 in the RG108-SRS subgroup and the NS-SRS subgroup but not in the RG108-NSRS (no SRS) subgroup and the NS-NSRS subgroup compared with the control group. No significant difference was found between the RG108-SRS and NS-SRS subgroups. Meanwhile, hippocampal neuronal loss was observed in RG108-SRS and NS-SRS subgroups. We thus demonstrated that RG108 could modify the progression of epileptogenesis after KA induced SE and prevent chronic epilepsy. Meanwhile, hypermethylation of RASgrf1 after KA induced SE could be reversed with corresponding changes of RASgrf1 expression. Additionally, we speculated that RASgrf1 might be a potential epigenetic mediator in epileptogenesis and chronic epilepsy.


Asunto(s)
Expresión Génica/efectos de los fármacos , Estado Epiléptico/metabolismo , ras-GRF1/metabolismo , Animales , Metilación de ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Electroencefalografía , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ácido Kaínico/farmacología , Masculino , Ratones Endogámicos C57BL , Ftalimidas/farmacología , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Estado Epiléptico/inducido químicamente , Estado Epiléptico/genética , Estado Epiléptico/fisiopatología , Triptófano/análogos & derivados , Triptófano/farmacología
14.
Arterioscler Thromb Vasc Biol ; 37(1): 98-107, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27856453

RESUMEN

OBJECTIVES: In response to endoplasmic reticulum (ER) stress, endothelial cells initiate corrective pathways such as the unfolded protein response. Recent studies suggest that reactive oxygen species produced on the ER may participate in homeostatic signaling through Ras in response to ER stress. We sought to identify mechanisms responsible for this focal signaling pathway. APPROACH AND RESULTS: In endothelial cells, we found that ER stress induced by tunicamycin activates the NADPH (nicotinamide adenine dinucleotide phosphate) oxidase Nox4 focally on the ER surface but not on the plasma membrane. Ras activation is also restricted to the ER, occurs downstream of Nox4, and is required for activation of the unfolded protein response. In contrast, treatment with the growth factor VEGF (vascular endothelial growth factor) results in Ras activation and reactive oxygen species production confined instead to the plasma membrane and not to the ER, demonstrating local coupling of reactive oxygen species and Ras signals. We further identify the calcium-responsive, ER-resident guanyl exchange factors RasGRF1 and RasGRF2 as novel upstream mediators linking Nox4 with Ras activation in response to ER stress. Oxidation of the sarcoendoplasmic reticulum calcium ATPase and increases in cytosolic calcium caused by ER stress are blocked by Nox4 knockdown, and reduction in cytosolic free calcium prevents both Ras activation and the unfolded protein response. CONCLUSIONS: ER stress triggers a localized signaling module on the ER surface involving Nox4-dependent calcium mobilization, which directs local Ras activation through ER-associated, calcium-responsive RasGRF.


Asunto(s)
Señalización del Calcio , Estrés del Retículo Endoplásmico , Retículo Endoplásmico/enzimología , Células Endoteliales de la Vena Umbilical Humana/enzimología , NADPH Oxidasas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Factores de Intercambio de Guanina Nucleótido ras/metabolismo , ras-GRF1/metabolismo , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , NADPH Oxidasa 4 , NADPH Oxidasas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Transfección , Tunicamicina/farmacología , Respuesta de Proteína Desplegada , Factor A de Crecimiento Endotelial Vascular/farmacología , Factores de Intercambio de Guanina Nucleótido ras/genética , ras-GRF1/genética
15.
Nature ; 486(7403): 415-9, 2012 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-22722204

RESUMEN

The modification of DNA by 5-methylcytosine (5mC) has essential roles in cell differentiation and development through epigenetic gene regulation. 5mC can be converted to another modified base, 5-hydroxymethylcytosine (5hmC), by the tet methylcytosine dioxygenase (Tet) family of enzymes. Notably, the balance between 5hmC and 5mC in the genome is linked with cell-differentiation processes such as pluripotency and lineage commitment. We have previously reported that the maternal factor PGC7 (also known as Dppa3, Stella) is required for the maintenance of DNA methylation in early embryogenesis, and protects 5mC from conversion to 5hmC in the maternal genome. Here we show that PGC7 protects 5mC from Tet3-mediated conversion to 5hmC by binding to maternal chromatin containing dimethylated histone H3 lysine 9 (H3K9me2) in mice. In addition, imprinted loci that are marked with H3K9me2 in mature sperm are protected by PGC7 binding in early embryogenesis. This type of regulatory mechanism could be involved in DNA modifications in somatic cells as well as in early embryos.


Asunto(s)
5-Metilcitosina/metabolismo , Citosina/análogos & derivados , Embrión de Mamíferos/metabolismo , Histonas/química , Histonas/metabolismo , Proteínas Represoras/metabolismo , Animales , Cromatina/química , Cromatina/metabolismo , Proteínas Cromosómicas no Histona , Citosina/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Embrión de Mamíferos/embriología , Desarrollo Embrionario , Femenino , Impresión Genómica/genética , Lisina/química , Lisina/metabolismo , Masculino , Metilación , Ratones , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , ARN Largo no Codificante , ARN no Traducido/genética , Espermatozoides/metabolismo , ras-GRF1/genética
16.
BMC Ophthalmol ; 18(1): 124, 2018 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-29793445

RESUMEN

BACKGROUND: This study aimed to assess heritability of myopia in Lithuania and evaluate both genes GJD2 (Gap Junction Protein, Delta 2) and RASGRF1 (RAS protein-specific guanine nucleotide-releasing factor 1) relation with myopia. METHODS: In this study Lithuanian twin population aged between 18 and 40 (n = 460) were examined. Single-nucleotide polymorphisms of the RASGRF1 (rs8027411) and GJD2 (rs634990) genes were assessed by real-time polymerase chain reaction method. RESULTS: Intrapair correlations for spherical equivalent in all twin pairs were significantly higher in MZ twin pairs r = 0.539 (p < 0.001, 95% CI 0.353-0.684) than in DZ twin pairs r = 0.203 (p < 0.01, 95% CI 0.0633-0.442) in myopia group. Correlations for spherical equivalent in emmetropia group were not significant in MZ twin pairs r = 0.091 (p > 0.05, 95% CI -0.215-0.381) and in DZ twin pairs r = - 0.220 (p > 0.05, 95% CI -0.587-0.222). The odds ratio (95% CI) were 2.7 (1.018-7.460) for combinations of genotypes of rs634990 CC and rs8027411 GT (p = 0.046). CONCLUSIONS: Our studies have shown that the heritability of myopia makes 67.2% in Lithuania. Persons with combinations of genotypes rs634990 CC and rs8027411 GT have 2.7 times higher odds to have myopia.


Asunto(s)
Conexinas/genética , Predisposición Genética a la Enfermedad , Miopía/genética , ras-GRF1/genética , Adolescente , Adulto , Enfermedades en Gemelos , Femenino , Genotipo , Humanos , Lituania , Masculino , Oportunidad Relativa , Polimorfismo de Nucleótido Simple , Reacción en Cadena en Tiempo Real de la Polimerasa , Errores de Refracción/genética , Adulto Joven
17.
Mol Cell Neurosci ; 85: 127-147, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28966131

RESUMEN

Various parameters of neurogenesis were analyzed in parallel in the two neurogenic areas (the Dentate Gyrus[DG] and the Subventricular Zone[SVZ]/Rostral Migratory Stream[RMS]/Main Olfactory Bulb[MOB] neurogenic system) of adult WT and KO mouse strains for the Ras-GRF1/2 genes (Ras-GRF1-KO, Ras-GRF2-KO, Ras-GRF1/2-DKO). Significantly reduced numbers of doublecortin[DCX]-positive cells were specifically observed in the DG, but not the SVZ/RMS/MOB neurogenic region, of Ras-GRF2-KO and Ras-GRF1/2-DKO mice indicating that this novel Ras-GRF2-dependent phenotype is spatially restricted to a specific neurogenic area. Consistent with a role of CREB as mediator of Ras-GRF2 function in neurogenesis, the density of p-CREB-positive cells was also specifically reduced in all neurogenic regions of Ras-GRF2-KO and DKO mice. Similar levels of early neurogenic proliferation markers (Ki67, BrdU) were observed in all different Ras-GRF genotypes analyzed but significantly elevated levels of nestin-immunolabel, particularly of undifferentiated, highly ramified, A-type nestin-positive neurons were specifically detected in the DG but not the SVZ/RMS/MOB of Ras-GRF2-KO and DKO mice. Together with assays of other neurogenic markers (GFAP, Sox2, Tuj1, NeuN), these observations suggest that the deficit of DCX/p-CREB-positive cells in the DG of Ras-GRF2-depleted mice does not involve impaired neuronal proliferation but rather delayed transition from the stem cell stage to the differentiation stages of the neurogenic process. This model is also supported by functional analyses of DG-derived neurosphere cultures and transcriptional characterization of the neurogenic areas of mice of all relevant Ras-GRF genotypes suggesting that the neurogenic role of Ras-GRF2 is exerted in a cell-autonomous manner through a specific transcriptional program.


Asunto(s)
Diferenciación Celular/fisiología , Giro Dentado/citología , Células-Madre Neurales/citología , Neurogénesis/fisiología , ras-GRF1/metabolismo , Animales , Giro Dentado/metabolismo , Proteína Doblecortina , Factor 2 Liberador de Guanina Nucleótido/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nestina
18.
Mol Cell Neurosci ; 82: 88-95, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28495450

RESUMEN

Rho guanine nucleotide exchange factor (RGNEF) is a 190kDa RNA binding protein (RBP) that also contains a Dbl/PH domain capable of RhoA activation. Consistent with a key role in the pathogenesis of amyotrophic lateral sclerosis (ALS), RGNEF forms pathological neuronal cytoplasmic inclusions in degenerating spinal motor neurons. To further understand the role of RGNEF in the stress response, we first observed that the expression of RGNEF is upregulated in murine spinal motor neurons following distal sciatic nerve injury. Secondly, in response to in vitro cellular stress (500µM sodium arsenite for 1h; or 400mM sorbitol 1 hour exposure; as an oxidative or osmotic stress, respectively), we observed a significant survival benefit in RGNEF-transfected HEK293T cells. Using deletion constructs, we found that the NH2-terminus domain is essential for this protective effect. Interestingly, we observed that under stress conditions RGNEF associates with Staufen1 positive granules but not TIA-1-positive stress granules. These findings support the hypothesis that RGNEF plays a critical role both in RNA homeostasis and in the response to cell stress.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Neuronas Motoras/metabolismo , Estrés Fisiológico , ras-GRF1/metabolismo , Animales , Arsenitos/farmacología , Células HEK293 , Homeostasis/fisiología , Humanos , Cuerpos de Inclusión/metabolismo , Ratones Endogámicos C57BL , ARN/metabolismo , Compuestos de Sodio/farmacología
19.
Int J Mol Sci ; 19(10)2018 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-30308936

RESUMEN

BACKGROUND: Diabetic cardiomyopathy (DCM) is characterized by cardiac fibrosis and stiffness, which often develops into heart failure. This study investigated the role of Ras protein-specific guanine nucleotide releasing factor 1 (RasGRF1) in the development of DCM. METHODS: Forty-eight mice were divided into four groups (n = 12 per group): Group 1: Wild-type (WT) mice, Group 2: RasGRF1 deficiency (RasGRF1-/-) mice. Group 3: Streptozotocin (STZ)-induced diabetic WT mice, Group 4: STZ-induced diabetic RasGRF1-/- mice. Myocardial functions were assessed by cardiac echography. Heart tissues from all of the mice were investigated for cardiac fibrosis, inflammation, and oxidative stress markers. RESULTS: Worse impaired diastolic function with elevation serum interleukin (IL)-6 was found in the diabetic group compared with the non-diabetic groups. Serum IL-6 levels were found to be elevated in the diabetic compared with the non-diabetic groups. However, the diabetic RasGRF1-/- mice exhibited lower serum IL-6 levels and better diastolic function than the diabetic WT mice. The diabetic RasGRF1-/- mice were associated with reduced cardiac inflammation, which was shown by lower invading inflammation cells, lower expression of matrix metalloproteinase 9, and less chemokines compared to the diabetic WT mice. Furthermore, less oxidative stress as well as extracellular matrix deposition leading to a reduction in cardiac fibrosis was also found in the diabetic RasGRF1-/- mice compared with the diabetic WT mice. CONCLUSION: The deletion of RasGRF1 attenuated myocardial fibrosis and improved cardiac function in diabetic mice through inhibiting inflammation and oxidative stress.


Asunto(s)
Cardiomiopatías Diabéticas/etiología , Cardiomiopatías Diabéticas/metabolismo , Eliminación de Gen , Inflamación/complicaciones , Inflamación/metabolismo , Estrés Oxidativo , ras-GRF1/genética , Animales , Biomarcadores , Citocinas/metabolismo , Cardiomiopatías Diabéticas/patología , Modelos Animales de Enfermedad , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Fibrosis , Regulación de la Expresión Génica , Glucosa/metabolismo , Mediadores de Inflamación , Ratones , Ratones Noqueados , Miofibroblastos/metabolismo , Estreptozocina/efectos adversos , ras-GRF1/metabolismo
20.
Biochim Biophys Acta ; 1862(2): 284-95, 2016 02.
Artículo en Inglés | MEDLINE | ID: mdl-26675527

RESUMEN

Our recent study demonstrated that an amyloid-ß binding molecule, BTA-EG4, increases dendritic spine number via Ras-mediated signaling. To potentially optimize the potency of the BTA compounds, we synthesized and evaluated an amyloid-ß binding analog of BTA-EG4 with increased solubility in aqueous solution, BTA-EG6. We initially examined the effects of BTA-EG6 on dendritic spine formation and found that BTA-EG6-treated primary hippocampal neurons had significantly increased dendritic spine number compared to control treatment. In addition, BTA-EG6 significantly increased the surface level of AMPA receptors. Upon investigation into the molecular mechanism by which BTA-EG6 promotes dendritic spine formation, we found that BTA-EG6 may exert its effects on spinogenesis via RasGRF1-ERK signaling, with potential involvement of other spinogenesis-related proteins such as Cdc42 and CDK5. Taken together, our data suggest that BTA-EG6 boosts spine and synapse number, which may have a beneficial effect of enhancing neuronal and synaptic function in the normal healthy brain.


Asunto(s)
Benzotiazoles/química , Benzotiazoles/farmacología , Espinas Dendríticas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteínas ras/metabolismo , ras-GRF1/metabolismo , Compuestos de Anilina/química , Compuestos de Anilina/farmacología , Animales , Células Cultivadas , Espinas Dendríticas/metabolismo , Espinas Dendríticas/ultraestructura , Glicol de Etileno/química , Glicol de Etileno/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratas Sprague-Dawley , Receptores AMPA/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA