Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Psychiatry ; 27(12): 5020-5027, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36224258

RESUMEN

Tourette Syndrome (TS) is a heritable, early-onset neuropsychiatric disorder that typically begins in early childhood. Identifying rare genetic variants that make a significant contribution to risk in affected families may provide important insights into the molecular aetiology of this complex and heterogeneous syndrome. Here we present a whole-genome sequencing (WGS) analysis from the 11-generation pedigree (>500 individuals) of a densely affected Costa Rican family which shares ancestry from six founder pairs. By conducting an identity-by-descent (IBD) analysis using WGS data from 19 individuals from the extended pedigree we have identified putative risk haplotypes that were not seen in controls, and can be linked with four of the six founder pairs. Rare coding and non-coding variants present on the haplotypes and only seen in haplotype carriers show an enrichment in pathways such as regulation of locomotion and signal transduction, suggesting common mechanisms by which the haplotype-specific variants may be contributing to TS-risk in this pedigree. In particular we have identified a rare deleterious missense variation in RAPGEF1 on a chromosome 9 haplotype and two ultra-rare deleterious intronic variants in ERBB4 and IKZF2 on the same chromosome 2 haplotype. All three genes play a role in neurodevelopment. This study, using WGS data in a pedigree-based approach, shows the importance of investigating both coding and non-coding variants to identify genes that may contribute to disease risk. Together, the genes and variants identified on the IBD haplotypes represent biologically relevant targets for investigation in other pedigree and population-based TS data.


Asunto(s)
Neurogénesis , Síndrome de Tourette , Preescolar , Humanos , Costa Rica , Haplotipos , Linaje , Transducción de Señal , Síndrome de Tourette/genética , Neurogénesis/genética , Polimorfismo Genético , Secuenciación Completa del Genoma , Factor 2 Liberador de Guanina Nucleótido/genética
2.
Clin Genet ; 100(2): 144-155, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33834495

RESUMEN

RAPGEF1 is a guanine nucleotide exchange factor responsible for transmitting extracellular signals to the Ras family of GTPase located at the inside of membrane. Here, we report for the first time a homozygous mutation of RAPGEF1 in a consanguineous family with two siblings affected by neuropsychiatric disorder. To confirm the correlation of the mutation and the phenotype, we utilized in silico analysis and established a zebrafish model. Survival rate was reduced in the rapgef1a-knockdown model, and the zebrafish showed global morphological abnormalities, particularly of brain and blood vessels. Co-application of human RAPGEF1 wildtype mRNA effectively rescued the abnormal phenotype, while that of RAPGEF1 mRNA carrying the human mutation did not. This work is the first report of a human Mendelian disease associated with RAPGEF1 and the first report of a zebrafish model built for this gene. The phenotype of zebrafish model provides further evidence that defective RAPGEF1 may lead to global developmental delay in human patients.


Asunto(s)
Factor 2 Liberador de Guanina Nucleótido/genética , Mutación , Pez Cebra/embriología , Pez Cebra/genética , Animales , Modelos Animales de Enfermedad , Embrión no Mamífero/anomalías , Embrión no Mamífero/irrigación sanguínea , Femenino , Factor 2 Liberador de Guanina Nucleótido/metabolismo , Semivida , Humanos , Masculino , Trastornos del Humor/genética , Neuronas Motoras/patología , Linaje , Fenotipo , Proteínas de Pez Cebra/genética
3.
J Vasc Res ; 58(5): 277-285, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33951626

RESUMEN

The sodium-dependent phosphate transporter, SLC20A1, is required for elevated inorganic phosphate (Pi) induced vascular smooth muscle cell (VSMC) matrix mineralization and phenotype transdifferentiation. Recently, elevated Pi was shown to induce ERK1/2 phosphorylation through SLC20A1 by Pi uptake-independent functions in VSMCs, suggesting a cell signaling response to elevated Pi. Previous studies identified Rap1 guanine nucleotide exchange factor (RapGEF1) as an SLC20A1-interacting protein and RapGEF1 promotes ERK1/2 phosphorylation through Rap1 activation. In this study, we tested the hypothesis that RapGEF1 is a critical component of the SLC20A1-mediated Pi-induced ERK1/2 phosphorylation pathway. Co-localization of SLC20A1 and RapGEF1, knockdown of RapGEF1 with siRNA, and small molecule inhibitors of Rap1, B-Raf, and Mek1/2 were investigated. SLC20A1 and RapGEF1 were co-localized in peri-membranous structures in VSMCs. Knockdown of RapGEF1 and small molecule inhibitors against Rap1, B-Raf, and Mek1/2 eliminated elevated Pi-induced ERK1/2 phosphorylation. Knockdown of RapGEF1 inhibited SM22α mRNA expression and blocked elevated Pi-induced downregulation of SM22α mRNA. Together, these data suggest that RapGEF1 is required for SLC20A1-mediated elevated Pi signaling through a Rap1/B-Raf/Mek1/2 cell signaling pathway, thereby promoting ERK1/2 phosphorylation and inhibiting SM22α gene expression in VSMCs.


Asunto(s)
Factor 2 Liberador de Guanina Nucleótido/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Fosfatos/farmacología , Animales , Células Cultivadas , Factor 2 Liberador de Guanina Nucleótido/genética , Humanos , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Fosforilación , Transducción de Señal , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/metabolismo
4.
Int J Mol Sci ; 22(18)2021 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-34576182

RESUMEN

C3G (RAPGEF1) is a guanine nucleotide exchange factor (GEF) for GTPases from the Ras superfamily, mainly Rap1, although it also acts through GEF-independent mechanisms. C3G regulates several cellular functions. It is expressed at relatively high levels in specific brain areas, playing important roles during embryonic development. Recent studies have uncovered different roles for C3G in cancer that are likely to depend on cell context, tumour type, and stage. However, its role in brain tumours remained unknown until very recently. We found that C3G expression is downregulated in GBM, which promotes the acquisition of a more mesenchymal phenotype, enhancing migration and invasion, but not proliferation. ERKs hyperactivation, likely induced by FGFR1, is responsible for this pro-invasive effect detected in C3G silenced cells. Other RTKs (Receptor Tyrosine Kinases) are also dysregulated and could also contribute to C3G effects. However, it remains undetermined whether Rap1 is a mediator of C3G actions in GBM. Various Rap1 isoforms can promote proliferation and invasion in GBM cells, while C3G inhibits migration/invasion. Therefore, other RapGEFs could play a major role regulating Rap1 activity in these tumours. Based on the information available, C3G could represent a new biomarker for GBM diagnosis, prognosis, and personalised treatment of patients in combination with other GBM molecular markers. The quantification of C3G levels in circulating tumour cells (CTCs) in the cerebrospinal liquid and/or circulating fluids might be a useful tool to improve GBM patient treatment and survival.


Asunto(s)
Glioblastoma/metabolismo , Factor 2 Liberador de Guanina Nucleótido/metabolismo , Animales , Glioblastoma/genética , Factor 2 Liberador de Guanina Nucleótido/genética , Humanos , Células Neoplásicas Circulantes/metabolismo , Proteínas de Unión al GTP rap1/genética , Proteínas de Unión al GTP rap1/metabolismo
5.
Clin Exp Pharmacol Physiol ; 46(3): 237-245, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30152875

RESUMEN

Experimental studies have shown that overexpression of Rap guanine nucleotide exchange factor 1 (C3G) plays pro-survival and anti-apoptotic roles through molecule phosphorylated extracellular signal-regulated kinase1/2 (p-ERK1/2) in cardiomyocytes. However, it is still unclear if silencing of C3G may increase cell survival inhibition and apoptosis in cardiomyocytes, and whether C3G silence induced injuries are reduced by the overexpression of C3G through regulation of p-ERK1/2 and pro-apoptotic molecule Bax. In this study, the rat-derived H9C2 cardiomyocytes were infected with C3G small hairpin RNA interference recombinant lentiviruses, which silenced the endogenous C3G expression in the cardiomyocytes. Then, contrary experiments were conducted using C3G overexpression. The cell proliferation and apoptosis were analyzed in the cardiomyocytes which were treated with or without hypoxia/reoxygenation (H/R). Silencing of C3G leaded to significant increase in cell survival inhibition and apoptosis, combined with aggravated the injuries induced by H/R. Overexpression of C3G reduced the injuries induced by the silencing of C3G in the cardiomyocytes via regulation of p-ERK1/2 and Bax. In conclusion, our results provide new experimental evidence that silencing of C3G can increase cell survival inhibition and apoptosis in cardiomyocytes via regulation of p-ERK1/2 and Bax.


Asunto(s)
Apoptosis/genética , Silenciador del Gen , Factor 2 Liberador de Guanina Nucleótido/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Miocitos Cardíacos/citología , Proteína X Asociada a bcl-2/metabolismo , Animales , Línea Celular , Proliferación Celular/genética , Supervivencia Celular/genética , Factor 2 Liberador de Guanina Nucleótido/deficiencia , Fosfoproteínas/metabolismo , Ratas
6.
J Am Soc Nephrol ; 29(8): 2053-2059, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29895552

RESUMEN

Background C3 glomerulopathy (C3G) is a life-threatening kidney disease caused by dysregulation of the alternative pathway of complement (AP) activation. No approved specific therapy is available for C3G, although an anti-C5 mAb has been used off-label in some patients with C3G, with mixed results. Thus, there is an unmet medical need to develop other inhibitors of complement for C3G.Methods We used a murine model of lethal C3G to test the potential efficacy of an Fc fusion protein of complement receptor of the Ig superfamily (CRIg-Fc) in the treatment of C3G. CRIg-Fc binds C3b and inhibits C3 and C5 convertases of the AP. Mice with mutations in the factor H and properdin genes (FHm/mP-/-) develop early-onset C3G, with AP consumption, high proteinuria, and lethal crescentic GN.Results Treatment of FHm/mP-/- mice with CRIg-Fc, but not a control IgG, inhibited AP activation and diminished the consumption of plasma C3, factor B, and C5. CRIg-Fc-treated FHm/mP-/- mice also had significantly improved survival and reduced proteinuria, hematuria, BUN, glomerular C3 fragment, C9 and fibrin deposition, and GN pathology scores.Conclusions Therapeutics developed on the basis of the mechanism of action of soluble CRIg may be effective for the treatment of C3G and should be explored clinically.


Asunto(s)
Complemento C3/antagonistas & inhibidores , Complemento C3/genética , Glomerulonefritis por IGA/genética , Glomerulonefritis por IGA/prevención & control , Factor 2 Liberador de Guanina Nucleótido/genética , Receptores de Complemento/genética , Análisis de Varianza , Animales , Biopsia con Aguja , Western Blotting , Activación de Complemento , Factor B del Complemento/inmunología , Factor B del Complemento/metabolismo , Modelos Animales de Enfermedad , Glomerulonefritis por IGA/patología , Inmunohistoquímica , Pruebas de Función Renal , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria , Receptores de Complemento/metabolismo , Tasa de Supervivencia
7.
Genes Chromosomes Cancer ; 56(10): 750-757, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28639284

RESUMEN

Soft tissue angiofibroma is rare and has characteristic histomorphological and genetic features. For diagnostic purposes, there are no specific antibodies available. Fourteen lesions (6 females, 8 males; age range 7-67 years) of the lower extremities (12) and trunk (2) were investigated by immunohistochemistry, including for the first time NCOA2. NCOA2 was also tested in a control group of other spindle cell lesions. The known fusion-genes (AHRR-NCOA2 and GTF2I-NCOA2) were examined using RT-PCR in order to evaluate their diagnostic value. Cases in which no fusion gene was detected were additionally analysed by RNA sequencing. All cases tested showed nuclear expression of NCOA2. However, this was not specific since other spindle cell neoplasms also expressed this marker in a high percentage of cases. Other variably positive markers were EMA, SMA, desmin and CD34. STAT6 was negative in the cases tested. By RT-PCR for the most frequently observed fusions, an AHRR-NCOA2 fusion transcript was found in 9/14 cases. GTF2I-NCOA2 was not detected in the remaining cases (n = 3). RNA sequencing revealed three additional positive cases; two harbored a AHRR-NCOA2 fusion and one case a novel GAB1-ABL1 fusion. Two cases failed molecular analysis due to poor RNA quality. In conclusion, the AHRR-NCOA2 fusion is a frequent finding in soft tissue angiofibroma, while GTF2I-NCOA2 seems to be a rare genetic event. For the first time, we report a GAB1-ABL1 fusion in a soft tissue angiofibroma of a child. Nuclear expression of NCOA2 is not discriminating when compared with other spindle cell neoplasms.


Asunto(s)
Angiofibroma/genética , Coactivador 2 del Receptor Nuclear/genética , Fusión de Oncogenes/genética , Neoplasias de los Tejidos Blandos/genética , Adolescente , Adulto , Anciano , Angiofibroma/patología , Antígenos CD34/genética , Antígenos CD34/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Niño , Femenino , Genes abl/genética , Factor 2 Liberador de Guanina Nucleótido/genética , Humanos , Masculino , Persona de Mediana Edad , Coactivador 2 del Receptor Nuclear/metabolismo , Proteínas Represoras/genética , Factor de Transcripción STAT6/genética , Factor de Transcripción STAT6/metabolismo , Neoplasias de los Tejidos Blandos/patología
8.
Biochim Biophys Acta ; 1853(10 Pt A): 2629-39, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26133694

RESUMEN

RapGEF1 (C3G) is a ubiquitously expressed protein that is essential for mammalian embryonic development. We have shown earlier that C3G regulates cytoskeletal dynamics and is required for neuronal differentiation. To determine if C3G plays a wider role in differentiation of multiple tissue types, we examined its role in skeletal muscle differentiation using the model system of C2C12 cells in culture. C3G protein is highly expressed in mouse skeletal muscle and its transcript and protein levels increase as C2C12 cells are induced to differentiate. Increase in C3G was predominantly seen in the nuclei of myotubes. Ectopic expression of C3G promoted myotube formation when cells were cultured in growth as well as differentiation medium and, enhanced MHC levels were associated with C3G expression. C3G induced differentiation required its catalytic and protein interaction domains and was dependent on the function of cellular R-Ras. Knockdown of cellular C3G using small hairpin RNA reduced expression of muscle specific markers and ß-catenin, resulting in impaired differentiation. Disabling C3G function also resulted in enhanced cell death suggesting that cellular C3G is required for cell survival. In cells grown in growth medium, over-expressed C3G increased Akt activity, and C3G knockdown reduced it. C3G expression also suppressed cyclin D1 levels, and induced p27 expression, molecules involved in regulating cell proliferation. Endogenous C3G localizes to focal adhesions in myotubes and C3G expressing cells show distinct stress fibers, elongation and parallel alignment. Expression of a dominant negative construct of C3G, disrupts actin cytoskeleton and formation of focal adhesions resulting in detachment of cells from the substratum and inhibition of differentiation. Our results provide evidence that C3G plays an important role in myogenic differentiation by coordinating cell cycle exit, actin dynamics and survival signaling.


Asunto(s)
Actinas/metabolismo , Diferenciación Celular/fisiología , Factor 2 Liberador de Guanina Nucleótido/metabolismo , Células Madre Mesenquimatosas/metabolismo , Desarrollo de Músculos/fisiología , Fibras Musculares Esqueléticas/metabolismo , Actinas/genética , Animales , Antígenos de Diferenciación/biosíntesis , Antígenos de Diferenciación/genética , Línea Celular , Supervivencia Celular/fisiología , Regulación de la Expresión Génica/fisiología , Factor 2 Liberador de Guanina Nucleótido/genética , Células Madre Mesenquimatosas/citología , Ratones , Fibras Musculares Esqueléticas/citología
10.
Biochem J ; 463(2): 257-70, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25028810

RESUMEN

The Ras-related GTPase Rap1 promotes cell adhesion and migration. Although the significance of Rap1 contribution to cell migration is increasingly being recognized, little is known about the biochemical mechanisms driving this process. In the present study, we discovered a previously unidentified regulatory role of insulin-like growth factor type I (IGF-I) receptor (IGF-IR) in CRK Src homology 3 (SH3)-binding guanine-nucleotide-releasing protein (C3G)-Rap1-fascin-actin axis promoting cell movement. We demonstrate that a burst of Rap1 activity, rather than presumed hyperactivation, is imperative for the onset of cell movement. We show that while autophosphorylated IGF-IR signals to C3G to activate Rap1, subsequent IGF-IR internalization promotes gradual inactivation of Rap1 by putative Rap1 GTPase-activating protein (GAP). Additionally, IGF-IR signalling recruits active Rap1 at sites of cell motile protrusions. C3G depletion prevents IGF-I-induced fascin accumulation at actin microspikes and blocks protrusions. In the absence of IGF-IR activity, the wild-type (WT) Rap1 and the constitutively active V12Rap1 mutant remain in cell-cell contacts. Forced inactivation of Rap1 signalling by overexpressing dominant negative N17Rap1, Rap1GAP or by silencing C3G has a detrimental effect on filamentous (F)-actin and cell adhesion irrespective of IGF-IR signalling. We conclude that the basal levels of Rap1 activity holds up cell adhesion, whereas sequential regulation of C3G and GAP by IGF-IR reverses the labile Rap1 function from supporting adhesion to promoting migration.


Asunto(s)
Adhesión Celular , Movimiento Celular , Células Epiteliales/enzimología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Factor 2 Liberador de Guanina Nucleótido/genética , Factor 2 Liberador de Guanina Nucleótido/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Células MCF-7 , Receptores de Somatomedina/genética , Receptores de Somatomedina/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rap1/genética
11.
Biochim Biophys Acta ; 1823(8): 1366-77, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22659131

RESUMEN

We have generated mouse transgenic lineages for C3G (tgC3G) and C3GΔCat (tgC3GΔCat, C3G mutant lacking the GEF domain), where the transgenes are expressed under the control of the megakaryocyte and platelet specific PF4 (platelet factor 4) gene promoter. Transgenic platelet activity has been analyzed through in vivo and in vitro approaches, including bleeding time, aggregation assays and flow cytometry. Both transgenes are expressed (RNA and protein) in purified platelets and megakaryocytes and do not modify the number of platelets in peripheral blood. Transgenic C3G animals showed bleeding times significantly shorter than control animals, while tgC3GΔCat mice presented a remarkable bleeding diathesis as compared to their control siblings. Accordingly, platelets from tgC3G mice showed stronger activation in response to platelet agonists such as thrombin, PMA, ADP or collagen than control platelets, while those from tgC3GΔCat animals had a lower response. In addition, we present data indicating that C3G is a mediator in the PKC pathway leading to Rap1 activation. Remarkably, a significant percentage of tgC3G mice presented a higher level of neutrophils than their control siblings. These results indicate that C3G plays an important role in platelet clotting through a mechanism involving its GEF activity and suggest that it might be also involved in neutrophil development.


Asunto(s)
Plaquetas/metabolismo , Factor 2 Liberador de Guanina Nucleótido/genética , Activación Plaquetaria , Animales , Plaquetas/efectos de los fármacos , Plaquetas/fisiología , Células Cultivadas , Activación Enzimática , Femenino , Ingeniería Genética , Factor 2 Liberador de Guanina Nucleótido/biosíntesis , Humanos , Recuento de Leucocitos , Masculino , Megacariocitos/metabolismo , Megacariocitos/fisiología , Ratones , Ratones Transgénicos , Neutrófilos/fisiología , Recuento de Plaquetas , Factor Plaquetario 4/genética , Regiones Promotoras Genéticas , Proteína Quinasa C/metabolismo , Transducción de Señal , Acetato de Tetradecanoilforbol/farmacología , Proteínas de Unión al GTP rap1/metabolismo
12.
Cell Biol Int ; 37(10): 1106-13, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23686869

RESUMEN

Integrin ß1 subunit and its downstream molecules, such as integrin-linked kinase and focal adhesion kinase, are imperative for promotion of cell proliferation, survival and anti-apoptosis in cardiomyocytes by activation of their downstream pro-survival signalling molecules, such as the phosphorylated extracellular signal-regulated kinase1/2 (p-ERK1/2). As a component of the integrin pathway, C3G (Crk-SH3 domain guanine nucleotide exchange factor) protein may be involved in the promotion of cell proliferation and survival and anti-apoptosis in the H9C2 cardiomyocytes. Rat-derived H9C2 cardiomyocytes were transfected with pCXN2-flag-hC3G, a human C3G overexpression eukaryotic recombinant plasmid. Apoptosis, cell proliferation and survival were analysed in the H9C2 cardiomyocytes either treated with hypoxia/reoxygenation (H/R). Human C3G mRNA overexpression significantly elevated C3G protein expression in H9C2 cardiomyocytes whether treated with H/R or not. C3G overexpression promoted proliferation and survival and anti-apoptosis, and attenuated the proliferative and survival inhibition, and apoptosis induced by H/R by activation of its downstream pro-survival signalling molecule, p-ERK1/2. The results suggest that C3G acts as a pro-survival molecule in H9C2 cardiomyocytes by activation of p-ERK1/2.


Asunto(s)
Factor 2 Liberador de Guanina Nucleótido/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animales , Apoptosis/genética , Proliferación Celular , Supervivencia Celular/genética , Citometría de Flujo , Factor 2 Liberador de Guanina Nucleótido/genética , Humanos , Integrinas/metabolismo , Miocitos Cardíacos/enzimología , Fosforilación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Coloración y Etiquetado
13.
Sci Rep ; 13(1): 14006, 2023 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-37635193

RESUMEN

The epidermis is mostly composed of keratinocytes and forms a protecting barrier against external aggressions and dehydration. Epidermal homeostasis is maintained by a fine-tuned balance between keratinocyte proliferation and differentiation. In the regulation of this process, the keratinocyte-specific miR-203 microRNA is of the outmost importance as it promotes differentiation, notably by directly targeting and down-regulating mRNA expression of genes involved in keratinocyte proliferation, such as ΔNp63, Skp2 and Msi2. We aimed at identifying new miR-203 targets involved in the regulation of keratinocyte proliferation/differentiation balance. To this end, a transcriptome analysis of human primary keratinocytes overexpressing miR-203 was performed and revealed that miR-203 overexpression inhibited functions like proliferation, mitosis and cell cycling, and activated differentiation, apoptosis and cell death. Among the down-regulated genes, 24 putative target mRNAs were identified and 8 of them were related to proliferation. We demonstrated that SRC and RAPGEF1 were direct targets of miR-203. Moreover, both were down-regulated during epidermal morphogenesis in a 3D reconstructed skin model, while miR-203 was up-regulated. Finally silencing experiments showed that SRC or RAPGEF1 contributed to keratinocyte proliferation and regulated their differentiation. Preliminary results suggest their involvement in skin carcinoma hyperproliferation. Altogether this data indicates that RAPGEF1 and SRC could be new mediators of miR-203 in epidermal homeostasis regulation.


Asunto(s)
Epidermis , Factor 2 Liberador de Guanina Nucleótido , MicroARNs , Proteínas Proto-Oncogénicas pp60(c-src) , Humanos , Homeostasis/genética , Queratinocitos , MicroARNs/genética , Mitosis , Piel , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Factor 2 Liberador de Guanina Nucleótido/genética
14.
Biochim Biophys Acta ; 1813(3): 456-65, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21223981

RESUMEN

Cytoskeletal remodeling is responsible for cell plasticity and facilitates differentiation, motility and adherence related functions. C3G (RAPGEF1), an exchange factor for Ras family of small GTPases, regulates cytoskeletal reorganization to induce filopodia in epithelial cells and neurite growth in neuroblastoma cells. Here we show that C3G overexpression induces neurite-like extensions (NLE) in MDA-MB-231 and BT549 breast carcinoma cells and not in a variety of other cancer cell lines examined. These processes were actin-rich with nodes, branches and microspikes. C3G associates with the cytoskeleton and its expression enabled stabilization of microtubules. NLE formation was dependent on Rap, Rac and Cdc42. C3G expression was associated with a decrease in cellular ß-catenin levels specifically in MDA-MB-231 and BT549 cells. ß-Catenin stabilization induced by GSK-3ß inhibition, or coexpression of ß-catenin, reduced C3G induced NLE formation. Time lapse analysis showed reduced motility of C3G expressing cells compared to GFP expressing cells. Our results suggest that C3G overexpression can induce phenotypic characteristics of neuronal cells in highly invasive breast cancer cells and inhibit their motility.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma/patología , Citoesqueleto/patología , Factor 2 Liberador de Guanina Nucleótido/metabolismo , Neuritas/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Línea Celular Tumoral , Movimiento Celular , Citoesqueleto/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/metabolismo , Factor 2 Liberador de Guanina Nucleótido/genética , Humanos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Regulación hacia Arriba , beta Catenina/metabolismo , Proteína de Unión al GTP cdc42/metabolismo
15.
J Cell Biol ; 178(5): 843-60, 2007 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-17724123

RESUMEN

Neurotrophins, such as NGF and BDNF, induce sustained activation of Rap1 small G protein and ERK, which are essential for neurite outgrowth. We show involvement of a GDP/GTP exchange factor (GEF) for Rap1, PDZ-GEF1, in these processes. PDZ-GEF1 is activated by GTP-Rap1 via a positive feedback mechanism. Upon NGF binding, the TrkA neurotrophin receptor is internalized from the cell surface, passes through early endosomes, and arrives in late endosomes. A tetrameric complex forms between PDZ-GEF1, synaptic scaffolding molecule and ankyrin repeat-rich membrane spanning protein which interacts directly with the TrkA receptor. At late endosomes, the complex induces sustained activation of Rap1 and ERK, resulting in neurite outgrowth. In cultured rat hippocampal neurons, PDZ-GEF1 is recruited to late endosomes in a BDNF-dependent manner involved in BDNF-induced neurite outgrowth. Thus, the interaction of PDZ-GEF1 with an internalized neurotrophin receptor transported to late endosomes induces sustained activation of both Rap1 and ERK and neurite outgrowth.


Asunto(s)
Endosomas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuritas/metabolismo , Receptor trkA/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/genética , Retroalimentación Fisiológica , Factores de Intercambio de Guanina Nucleótido/genética , Factor 2 Liberador de Guanina Nucleótido/genética , Factor 2 Liberador de Guanina Nucleótido/metabolismo , Guanosina Trifosfato/metabolismo , Guanilato-Quinasas , Hipocampo/citología , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Complejos Multiproteicos , Factor de Crecimiento Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Neuritas/ultraestructura , Células PC12 , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Receptor trkA/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas de Unión al GTP rap1/genética
16.
Stem Cell Rev Rep ; 17(4): 1465-1477, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33624208

RESUMEN

C3G (RAPGEF1), engaged in multiple signaling pathways, is essential for the early development of the mouse. In this study, we have examined its role in mouse embryonic stem cell self-renewal and differentiation. C3G null cells generated by CRISPR mediated knock-in of a targeting vector exhibited enhanced clonogenicity and long-term self-renewal. They did not differentiate in response to LIF withdrawal when compared to the wild type ES cells and were defective for lineage commitment upon teratoma formation in vivo. Gene expression analysis of C3G KO cells showed misregulated expression of a large number of genes compared with WT cells. They express higher levels of self-renewal factors like KLF4 and ESRRB and show high STAT3 activity, and very low ERK activity compared to WT cells. Reintroduction of C3G expression in a KO line partially reverted expression of ESRRB, and KLF4, and ERK activity similar to that seen in WT cells. The expression of self-renewal factors was persistent for a longer time, and induction of lineage-specific markers was not seen when C3G KO cells were induced to form embryoid bodies. C3G KO cells showed poor adhesion and significantly reduced levels of pFAK, pPaxillin, and Integrin-ß1, in addition to downregulation of the cluster of genes involved in cell adhesion, compared to WT cells. Our results show that C3G is essential for the regulation of STAT3, ERK, and adhesion signaling, to maintain pluripotency of mouse embryonic stem cells and enable their lineage commitment for differentiation.


Asunto(s)
Diferenciación Celular , Factor 2 Liberador de Guanina Nucleótido/genética , Células Madre Embrionarias de Ratones , Transducción de Señal , Animales , Diferenciación Celular/genética , Quinasas MAP Reguladas por Señal Extracelular , Factor Inhibidor de Leucemia , Ratones , Células Madre Embrionarias de Ratones/citología , Factor de Transcripción STAT3 , Transducción de Señal/genética
17.
Biochim Biophys Acta Mol Cell Res ; 1868(5): 118964, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33450305

RESUMEN

GSK3ß, a ubiquitously expressed Ser/Thr kinase, regulates cell metabolism, proliferation and differentiation. Its activity is spatially and temporally regulated dependent on external stimuli and interacting partners, and its deregulation is associated with various human disorders. In this study, we identify C3G (RapGEF1), a protein essential for mammalian embryonic development as an interacting partner and substrate of GSK3ß. In vivo and in vitro interaction assays demonstrated that GSK3ß and Akt are present in complex with C3G. Molecular modelling and mutational analysis identified a domain in C3G that aids interaction with GSK3ß, and overlaps with its nuclear export sequence. GSK3ß phosphorylates C3G on primed as well as unprimed sites, and regulates its subcellular localization. Over-expression of C3G resulted in activation of Akt and inactivation of GSK3ß. Huntingtin aggregate formation, dependent on GSK3ß inhibition, was enhanced upon C3G overexpression. Stable clones of C2C12 cells generated by CRISPR/Cas9 mediated knockdown of C3G, that cannot differentiate, show reduced Akt activity and S9-GSK3ß phosphorylation compared to wild type cells. Co-expression of catalytically active GSK3ß inhibited C3G induced myocyte differentiation. C3G mutant defective for GSK3ß phosphorylation, does not alter S9-GSK3ß phosphorylation and, is compromised for inducing myocyte differentiation. Our results show complex formation and reciprocal regulation between GSK3ß and C3G. We have identified a novel function of C3G as a negative regulator of GSK3ß, a property important for its ability to induce myogenic differentiation.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta/metabolismo , Factor 2 Liberador de Guanina Nucleótido/química , Factor 2 Liberador de Guanina Nucleótido/metabolismo , Mutación , Mioblastos/citología , Animales , Células COS , Diferenciación Celular , Línea Celular , Chlorocebus aethiops , Citoplasma/metabolismo , Regulación de la Expresión Génica , Factor 2 Liberador de Guanina Nucleótido/genética , Células HEK293 , Humanos , Ratones , Desarrollo de Músculos , Mioblastos/metabolismo , Fosforilación
18.
Can J Physiol Pharmacol ; 88(8): 808-16, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20725139

RESUMEN

The guanine nucleotide exchange factor C3G is one of the mediators of endothelin-1 (ET-1) intracellular signaling cascades and is vital for kidney development and homeostasis. The aim of the current study was to analyze the specificity of ET-1-induced signaling via C3G in rat glomerular mesangial cells (GMC) and to investigate the biological significance of C3G during mesangioproliferative glomerulonephritis. In GMC, C3G expression was increased (1) in vivo after induction of the anti-Thy1 model of glomerulonephritis and (2) in cell culture experiments after fetal bovine serum incubation. To examine the consequences of C3G up-regulation, adenovirus-mediated gene transfer of C3G into cultured glomerular cells was done, and the GTP loading of the small G proteins Rap1 and R-Ras was analyzed. Overexpression of C3G in mesangial cells resulted in enhanced activation of Rap1, but failed to affect the GTP-bound status of R-Ras in ET-1-stimulated cells. C3G overexpression led to significant changes in GMC spreading and migration patterns in response to ET-1 stimulation and increased stress fiber formation, which was mimicked by Rap1A overexpression. Together, these findings suggest (1) the existence of regulatory mechanisms resulting in disease-related up-regulation of C3G in GMC and (2) that an increase in the C3G protein level may contribute to the resolution stage of mesangioproliferative glomerulonephritis by reducing GMC sensitivity to ET-1, modulating cellular motility, and actin dynamics.


Asunto(s)
Endotelina-1/farmacología , Glomerulonefritis Membranoproliferativa/metabolismo , Factor 2 Liberador de Guanina Nucleótido/metabolismo , Células Mesangiales/metabolismo , Transducción de Señal/fisiología , Citoesqueleto de Actina/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Forma de la Célula/efectos de los fármacos , Forma de la Célula/genética , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Expresión Génica/genética , Glomerulonefritis Membranoproliferativa/inducido químicamente , Glomerulonefritis Membranoproliferativa/inmunología , Factor 2 Liberador de Guanina Nucleótido/genética , Guanosina Trifosfato/metabolismo , Glomérulos Renales/metabolismo , Masculino , Células Mesangiales/citología , Células Mesangiales/efectos de los fármacos , Ratas , Ratas Wistar , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Suero/fisiología , Antígenos Thy-1/inmunología , Transducción Genética , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/genética , Proteínas de Unión al GTP rap1/genética , Proteínas de Unión al GTP rap1/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
19.
Sci Rep ; 10(1): 18838, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-33139841

RESUMEN

Mice lacking C3G (RapGEF1), a ubiquitously expressed protein essential for neuronal differentiation, show multiple defects in brain development. Function of C3G in neurogenesis is poorly defined. Here, we identify brain specific expression of a novel C3G isoform in mice and humans. This isoform has an insert in the Crk-binding region, generating a polypeptide of 175 kDa, unlike the previously known 140 kDa form expressed in all other tissues. In the adult mouse brain, C3G expression is seen in neurons, but was not detectable in GFAP-positive cells. C3G levels were high in the CA3 region of hippocampus and in mitral cells of olfactory bulb. Neural progenitor cells positive for Doublecortin and Nestin, show expression of C3G. During development, C3G is expressed in precursor cells prior to their differentiation into mature neurons or astrocytes. The 175 kDa as well as 140 kDa forms are seen in embryonic mouse brain, while only the 175 kDa variant is seen in post-natal brain. Human cerebral organoids generated from induced pluripotent stem cells predominantly expressed the 140 kDa polypeptides, and the 175 kDa isoform appeared upon maturation. This study describes developmental regulation and neuronal expression of a brain specific isoform of C3G, a molecule essential for normal development of the mammalian brain.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Expresión Génica , Factor 2 Liberador de Guanina Nucleótido/genética , Factor 2 Liberador de Guanina Nucleótido/metabolismo , Animales , Encéfalo/embriología , Hipocampo/metabolismo , Humanos , Ratones , Bulbo Olfatorio/metabolismo , Organoides/metabolismo , Péptidos/metabolismo , Isoformas de Proteínas/metabolismo
20.
Signal Transduct Target Ther ; 5(1): 29, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32296045

RESUMEN

C3G is a GEF (guanine nucleotide exchange factor) for Rap GTPases, among which the isoform Rap1b is an essential protein in platelet biology. Using transgenic mouse models with platelet-specific overexpression of C3G or mutant C3GΔCat, we have unveiled a new function of C3G in regulating the hemostatic function of platelets through its participation in the thrombin-PKC-Rap1b pathway. C3G also plays important roles in angiogenesis, tumor growth, and metastasis through its regulation of the platelet secretome. In addition, C3G contributes to megakaryopoiesis and thrombopoiesis. Here, we used a platelet-specific C3G-KO mouse model to further support the role of C3G in hemostasis. C3G-KO platelets showed a significant delay in platelet activation and aggregation as a consequence of the defective activation of Rap1, which resulted in decreased thrombus formation in vivo. Additionally, we explored the contribution of C3G-Rap1b to platelet signaling pathways triggered by thrombin, PMA or ADP, in the referenced transgenic mouse model, through the use of a battery of specific inhibitors. We found that platelet C3G is phosphorylated at Tyr504 by a mechanism involving PKC-Src. This phosphorylation was shown to be positively regulated by ERKs through their inhibition of the tyrosine phosphatase Shp2. Moreover, C3G participates in the ADP-P2Y12-PI3K-Rap1b pathway and is a mediator of thrombin-TXA2 activities. However, it inhibits the synthesis of TXA2 through cPLA2 regulation. Taken together, our data reveal the critical role of C3G in the main pathways leading to platelet activation and aggregation through the regulation of Rap1b.


Asunto(s)
Factor 2 Liberador de Guanina Nucleótido/genética , Receptores Purinérgicos P2Y12/genética , Trombina/genética , Proteínas de Unión al GTP rap/genética , Animales , Plaquetas/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Hemostasis/genética , Humanos , Ratones , Ratones Noqueados , Fosforilación , Activación Plaquetaria/genética , Agregación Plaquetaria/genética , Proteína Quinasa C/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Transducción de Señal/genética , Trombopoyesis/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA