Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Sci Rep ; 13(1): 14006, 2023 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-37635193

RESUMO

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.


Assuntos
Epiderme , Fator 2 de Liberação do Nucleotídeo Guanina , MicroRNAs , Proteínas Proto-Oncogênicas pp60(c-src) , Humanos , Homeostase/genética , Queratinócitos , MicroRNAs/genética , Mitose , Pele , Proteínas Proto-Oncogênicas pp60(c-src)/genética , Fator 2 de Liberação do Nucleotídeo Guanina/genética
2.
Mol Psychiatry ; 27(12): 5020-5027, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36224258

RESUMO

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.


Assuntos
Neurogênese , Síndrome de Tourette , Pré-Escolar , Humanos , Costa Rica , Haplótipos , Linhagem , Transdução de Sinais , Síndrome de Tourette/genética , Neurogênese/genética , Polimorfismo Genético , Sequenciamento Completo do Genoma , Fator 2 de Liberação do Nucleotídeo Guanina/genética
3.
Int J Mol Sci ; 22(18)2021 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-34576182

RESUMO

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.


Assuntos
Glioblastoma/metabolismo , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Animais , Glioblastoma/genética , Fator 2 de Liberação do Nucleotídeo Guanina/genética , Humanos , Células Neoplásicas Circulantes/metabolismo , Proteínas rap1 de Ligação ao GTP/genética , Proteínas rap1 de Ligação ao GTP/metabolismo
4.
J Vasc Res ; 58(5): 277-285, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33951626

RESUMO

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.


Assuntos
Fator 2 de Liberação do Nucleotídeo Guanina/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Fosfatos/farmacologia , Animais , Células Cultivadas , Fator 2 de Liberação do Nucleotídeo Guanina/genética , Humanos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Fosforilação , Transdução de Sinais , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/metabolismo
5.
Clin Genet ; 100(2): 144-155, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33834495

RESUMO

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.


Assuntos
Fator 2 de Liberação do Nucleotídeo Guanina/genética , Mutação , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Animais , Modelos Animais de Doenças , Embrião não Mamífero/anormalidades , Embrião não Mamífero/irrigação sanguínea , Feminino , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Meia-Vida , Humanos , Masculino , Transtornos do Humor/genética , Neurônios Motores/patologia , Linhagem , Fenótipo , Proteínas de Peixe-Zebra/genética
6.
Stem Cell Rev Rep ; 17(4): 1465-1477, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33624208

RESUMO

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.


Assuntos
Diferenciação Celular , Fator 2 de Liberação do Nucleotídeo Guanina/genética , Células-Tronco Embrionárias Murinas , Transdução de Sinais , Animais , Diferenciação Celular/genética , MAP Quinases Reguladas por Sinal Extracelular , Fator Inibidor de Leucemia , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Fator de Transcrição STAT3 , Transdução de Sinais/genética
7.
Biochim Biophys Acta Mol Cell Res ; 1868(5): 118964, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33450305

RESUMO

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.


Assuntos
Glicogênio Sintase Quinase 3 beta/metabolismo , Fator 2 de Liberação do Nucleotídeo Guanina/química , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Mutação , Mioblastos/citologia , Animais , Células COS , Diferenciação Celular , Linhagem Celular , Chlorocebus aethiops , Citoplasma/metabolismo , Regulação da Expressão Gênica , Fator 2 de Liberação do Nucleotídeo Guanina/genética , Células HEK293 , Humanos , Camundongos , Desenvolvimento Muscular , Mioblastos/metabolismo , Fosforilação
8.
Sci Rep ; 10(1): 18838, 2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-33139841

RESUMO

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.


Assuntos
Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Expressão Gênica , Fator 2 de Liberação do Nucleotídeo Guanina/genética , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Animais , Encéfalo/embriologia , Hipocampo/metabolismo , Humanos , Camundongos , Bulbo Olfatório/metabolismo , Organoides/metabolismo , Peptídeos/metabolismo , Isoformas de Proteínas/metabolismo
9.
Signal Transduct Target Ther ; 5(1): 29, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32296045

RESUMO

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.


Assuntos
Fator 2 de Liberação do Nucleotídeo Guanina/genética , Receptores Purinérgicos P2Y12/genética , Trombina/genética , Proteínas rap de Ligação ao GTP/genética , Animais , Plaquetas/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Hemostasia/genética , Humanos , Camundongos , Camundongos Knockout , Fosforilação , Ativação Plaquetária/genética , Agregação Plaquetária/genética , Proteína Quinase C/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Transdução de Sinais/genética , Trombopoese/genética
11.
Neurotox Res ; 35(1): 41-48, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29959729

RESUMO

α-Synuclein is a 140-amino acid protein produced predominantly by neurons in the brain which plays a role in the regulation of neurotransmitter release, synaptic function, and plasticity, thus making it the focus in understanding the etiology of a group of neurodegenerative diseases. We conducted genome-wide association studies (GWAS) of α-synuclein levels in cerebrospinal fluid (CSF) with 209 non-Hispanic white participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI-1) cohort using a linear regression model to identify novel variants associated with α-synuclein concentration. The minor allele (T) of rs7072338 in the long intergenic non-protein coding RNA 1515 (LINC01515) and the minor allele (T) of rs17794023 in clusterin-associated protein 1 (CLUAP1) were associated with higher CSF α-synuclein levels at genome-wide significance (P = 4.167 × 10-9 and 9.56 × 10-9, respectively). In addition, single nucleotide polymorphisms (SNPs) near amyloid beta precursor protein (APP) (rs1394839) (P = 2.31 × 10-7), Rap guanine nucleotide exchange factor 1 (RAPGEF1) (rs10901091) (P = 8.07 × 10-7), and two intergenic loci on chromosome 2 and 14 (rs11687064 P = 2.50 × 10-7and rs7147386 P = 4.05 × 10-7) were identified as suggestive loci associated with CSF α-synuclein levels. We have identified significantly associated SNPs for CSF α-synuclein. These associations have important implications for a better understanding of α-synuclein regulation and allow researchers to further explore the relationships between these SNPs and α-synuclein-related neurodegenerative disorders.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Antígenos de Neoplasias/genética , Fator 2 de Liberação do Nucleotídeo Guanina/genética , RNA Longo não Codificante/genética , alfa-Sinucleína/líquido cefalorraquidiano , Idoso , Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/genética , Biomarcadores/líquido cefalorraquidiano , Disfunção Cognitiva/líquido cefalorraquidiano , Disfunção Cognitiva/genética , Estudos de Coortes , Endofenótipos , Feminino , Estudo de Associação Genômica Ampla , Humanos , Masculino , Polimorfismo de Nucleotídeo Único , População Branca
12.
Clin Exp Pharmacol Physiol ; 46(3): 237-245, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30152875

RESUMO

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.


Assuntos
Apoptose/genética , Inativação Gênica , Fator 2 de Liberação do Nucleotídeo Guanina/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Miócitos Cardíacos/citologia , Proteína X Associada a bcl-2/metabolismo , Animais , Linhagem Celular , Proliferação de Células/genética , Sobrevivência Celular/genética , Fator 2 de Liberação do Nucleotídeo Guanina/deficiência , Fosfoproteínas/metabolismo , Ratos
13.
J Am Soc Nephrol ; 29(8): 2053-2059, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29895552

RESUMO

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.


Assuntos
Complemento C3/antagonistas & inibidores , Complemento C3/genética , Glomerulonefrite por IGA/genética , Glomerulonefrite por IGA/prevenção & controle , Fator 2 de Liberação do Nucleotídeo Guanina/genética , Receptores de Complemento/genética , Análise de Variância , Animais , Biópsia por Agulha , Western Blotting , Ativação do Complemento , Fator B do Complemento/imunologia , Fator B do Complemento/metabolismo , Modelos Animais de Doenças , Glomerulonefrite por IGA/patologia , Imuno-Histoquímica , Testes de Função Renal , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Receptores de Complemento/metabolismo , Taxa de Sobrevida
14.
Sci Signal ; 10(491)2017 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-28790195

RESUMO

The adaptor protein CrkII regulates T cell adhesion by recruiting the guanine nucleotide exchange factor C3G, an activator of Rap1. Subsequently, Rap1 stimulates the integrin LFA-1, which leads to T cell adhesion and interaction with antigen-presenting cells (APCs). The adhesion of T cells to APCs is critical for their proper function and education. The interface between the T cell and the APC is known as the immunological synapse. It is characterized by the specific organization of proteins that can be divided into central supramolecular activation clusters (c-SMACs) and peripheral SMACs (p-SMACs). Through total internal reflection fluorescence (TIRF) microscopy and experiments with supported lipid bilayers, we determined that activated Rap1 was recruited to the immunological synapse and localized to the p-SMAC. C3G and the active (dephosphorylated) form of CrkII also localized to the same compartment. In contrast, inactive (phosphorylated) CrkII was confined to the c-SMAC. Activation of CrkII and its subsequent movement from the c-SMAC to the p-SMAC depended on the phosphatase SHP-1, which acted downstream of the T cell receptor. In the p-SMAC, CrkII recruited C3G, which led to Rap1 activation and LFA-1-mediated adhesion of T cells to APCs. Functionally, SHP-1 was necessary for both the adhesion and migration of T cells. Together, these data highlight a signaling pathway in which SHP-1 acts through CrkII to reshape the pattern of Rap1 activation in the immunological synapse.


Assuntos
Proteínas Ativadoras de GTPase/metabolismo , Sinapses Imunológicas/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Proteínas Proto-Oncogênicas c-crk/metabolismo , Linfócitos T/imunologia , Imunidade Adaptativa , Animais , Adesão Celular , Proteínas Ativadoras de GTPase/genética , Fator 2 de Liberação do Nucleotídeo Guanina/genética , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Células HEK293 , Humanos , Sinapses Imunológicas/metabolismo , Células Jurkat , Ativação Linfocitária/imunologia , Antígeno-1 Associado à Função Linfocitária/metabolismo , Camundongos , Camundongos Transgênicos , Fosforilação , Cultura Primária de Células , Proteína Tirosina Fosfatase não Receptora Tipo 6/genética , Proteínas Proto-Oncogênicas c-crk/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Análise de Célula Única , Linfócitos T/metabolismo
15.
Genes Chromosomes Cancer ; 56(10): 750-757, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28639284

RESUMO

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.


Assuntos
Angiofibroma/genética , Coativador 2 de Receptor Nuclear/genética , Fusão Oncogênica/genética , Neoplasias de Tecidos Moles/genética , Adolescente , Adulto , Idoso , Angiofibroma/patologia , Antígenos CD34/genética , Antígenos CD34/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Criança , Feminino , Genes abl/genética , Fator 2 de Liberação do Nucleotídeo Guanina/genética , Humanos , Masculino , Pessoa de Meia-Idade , Coativador 2 de Receptor Nuclear/metabolismo , Proteínas Repressoras/genética , Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/metabolismo , Neoplasias de Tecidos Moles/patologia
16.
Mol Biol Cell ; 28(7): 984-995, 2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-28148649

RESUMO

C3G (RapGEF1) is a ubiquitously expressed guanine nucleotide exchange factor that functions in signaling pathways regulating cell proliferation, apoptosis, and actin reorganization. It is essential for differentiation and early embryonic development in mice. Overexpressed C3G shows predominant cytoplasmic localization, but endogenous C3G is a component of nuclear fractions in a variety of cell types. Coexpression of importin-α and inhibition of nuclear export by leptomycin B resulted in predominant nuclear localization of C3G. Functional NLSs, NES, and GSK3-ß-dependent phosphorylation regulate its dynamic nuclear localization. C3G translocates to the nucleus in response to myogenic differentiation and sublethal dose of cisplatin. C3G is associated with chromatin and nuclear matrix fractions. Cells with C3G localized in the nucleus showed peripheralization of heterochromatin and reduced histone modifications associated with euchromatin. Short hairpin RNA-mediated depletion of C3G in epithelial cells resulted in reduced expression of CDK inhibitors and the histone demethylase KDM5A. Myoblast clones with CRISPR/Cas9-mediated knockout of C3G failed to show repression of histone marks and did not show up-regulation of myosin heavy chain and myotube formation when grown in differentiation medium. Our results document regulated nucleocytoplasmic exchange of C3G in response to physiological stimuli and provide insights into nuclear functions for C3G.


Assuntos
Eucromatina/fisiologia , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Fator 2 de Liberação do Nucleotídeo Guanina/fisiologia , Código das Histonas/fisiologia , Actinas/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proliferação de Células , Eucromatina/metabolismo , Ácidos Graxos Insaturados/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fator 2 de Liberação do Nucleotídeo Guanina/genética , Camundongos , Desenvolvimento Muscular , Sinais de Localização Nuclear , Fosforilação , Transdução de Sinais , Regulação para Cima
17.
PLoS One ; 11(4): e0154174, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27111087

RESUMO

The establishment of a polarized morphology is essential for the development and function of neurons. During the development of the mammalian neocortex, neurons arise in the ventricular zone (VZ) from radial glia cells (RGCs) and leave the VZ to generate the cortical plate (CP). During their migration, newborn neurons first assume a multipolar morphology in the subventricular zone (SVZ) and lower intermediate zone (IZ). Subsequently, they undergo a multi-to-bipolar (MTB) transition to become bipolar in the upper IZ by developing a leading process and a trailing axon. The small GTPases Rap1A and Rap1B act as master regulators of neural cell polarity in the developing mouse neocortex. They are required for maintaining the polarity of RGCs and directing the MTB transition of multipolar neurons. Here we show that the Rap1 guanine nucleotide exchange factor (GEF) C3G (encoded by the Rapgef1 gene) is a crucial regulator of the MTB transition in vivo by conditionally inactivating the Rapgef1 gene in the developing mouse cortex at different time points during neuronal development. Inactivation of C3G results in defects in neuronal migration, axon formation and cortical lamination. Live cell imaging shows that C3G is required in cortical neurons for both the specification of an axon and the initiation of radial migration by forming a leading process.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Fator 2 de Liberação do Nucleotídeo Guanina/genética , Neocórtex/metabolismo , Neurogênese/genética , Neurônios/metabolismo , Animais , Polaridade Celular , Embrião de Mamíferos , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neocórtex/citologia , Neocórtex/embriologia , Neurônios/citologia , Transdução de Sinais , Imagem com Lapso de Tempo , Proteínas rap de Ligação ao GTP/genética , Proteínas rap de Ligação ao GTP/metabolismo , Proteínas rap1 de Ligação ao GTP/genética , Proteínas rap1 de Ligação ao GTP/metabolismo
18.
Biochim Biophys Acta ; 1853(10 Pt A): 2629-39, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26133694

RESUMO

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.


Assuntos
Actinas/metabolismo , Diferenciação Celular/fisiologia , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Células-Tronco Mesenquimais/metabolismo , Desenvolvimento Muscular/fisiologia , Fibras Musculares Esqueléticas/metabolismo , Actinas/genética , Animais , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/genética , Linhagem Celular , Sobrevivência Celular/fisiologia , Regulação da Expressão Gênica/fisiologia , Fator 2 de Liberação do Nucleotídeo Guanina/genética , Células-Tronco Mesenquimais/citologia , Camundongos , Fibras Musculares Esqueléticas/citologia
19.
Biochem J ; 463(2): 257-70, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25028810

RESUMO

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.


Assuntos
Adesão Celular , Movimento Celular , Células Epiteliais/enzimologia , Fator de Crescimento Insulin-Like I/metabolismo , Proteínas rap1 de Ligação ao GTP/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Fator 2 de Liberação do Nucleotídeo Guanina/genética , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/genética , Células MCF-7 , Receptores de Somatomedina/genética , Receptores de Somatomedina/metabolismo , Transdução de Sinais , Proteínas rap1 de Ligação ao GTP/genética
20.
Mol Cell Biol ; 34(6): 978-88, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24396067

RESUMO

T-lymphocyte adhesion plays a critical role in both inflammatory and autoimmune responses. The small GTPase Rap1 is the key coordinator mediating T-cell adhesion to endothelial cells, antigen-presenting cells, and virus-infected cells. We describe a signaling pathway, downstream of the cytotoxic T-lymphocyte antigen 4 (CTLA-4) receptor, leading to Rap1-mediated adhesion. We identified a role for the Rap1 guanine nucleotide exchange factor C3G in the regulation of T-cell adhesion and showed that this factor is required for both T-cell receptor (TCR)-mediated and CTLA-4-mediated T-cell adhesion. Our data indicated that C3G translocates to the plasma membrane downstream of TCR signaling, where it regulates activation of Rap1. We also showed that CTLA-4 receptor signaling mediates tyrosine phosphorylation in the C3G protein, and that this is required for augmented activation of Rap1 and increased adhesion mediated by leukocyte function-associated antigen type 1 (LFA-1). Zap70 is required for C3G translocation to the plasma membrane, whereas the Src family member Hck facilitates C3G phosphorylation. These findings point to C3G and Hck as promising potential therapeutic targets for the treatment of T-cell-dependent autoimmune disorders.


Assuntos
Antígeno CTLA-4/metabolismo , Adesão Celular/fisiologia , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Transdução de Sinais/fisiologia , Proteínas rap1 de Ligação ao GTP/metabolismo , Animais , Antígenos CD28/genética , Antígenos CD28/metabolismo , Antígeno CTLA-4/genética , Adesão Celular/genética , Linhagem Celular Tumoral , Membrana Celular/genética , Membrana Celular/metabolismo , Feminino , Genes src/genética , Fator 2 de Liberação do Nucleotídeo Guanina/genética , Humanos , Células Jurkat , Antígeno-1 Associado à Função Linfocitária/genética , Antígeno-1 Associado à Função Linfocitária/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação/genética , Fosforilação/fisiologia , Proteínas Proto-Oncogênicas c-hck/genética , Proteínas Proto-Oncogênicas c-hck/metabolismo , Transdução de Sinais/genética , Linfócitos T/metabolismo , Proteínas rap1 de Ligação ao GTP/genética , Proteínas ras/genética , Proteínas ras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA