Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
Cell Mol Life Sci ; 81(1): 84, 2024 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-38345631

RESUMO

C3G is a Rap1 GEF that plays a pivotal role in platelet-mediated processes such as angiogenesis, tumor growth, and metastasis by modulating the platelet secretome. Here, we explore the mechanisms through which C3G governs platelet secretion. For this, we utilized animal models featuring either overexpression or deletion of C3G in platelets, as well as PC12 cell clones expressing C3G mutants. We found that C3G specifically regulates α-granule secretion via PKCδ, but it does not affect δ-granules or lysosomes. C3G activated RalA through a GEF-dependent mechanism, facilitating vesicle docking, while interfering with the formation of the trans-SNARE complex, thereby restricting vesicle fusion. Furthermore, C3G promotes the formation of lamellipodia during platelet spreading on specific substrates by enhancing actin polymerization via Src and Rac1-Arp2/3 pathways, but not Rap1. Consequently, C3G deletion in platelets favored kiss-and-run exocytosis. C3G also controlled granule secretion in PC12 cells, including pore formation. Additionally, C3G-deficient platelets exhibited reduced phosphatidylserine exposure, resulting in decreased thrombin generation, which along with defective actin polymerization and spreading, led to impaired clot retraction. In summary, platelet C3G plays a dual role by facilitating platelet spreading and clot retraction through the promotion of outside-in signaling while concurrently downregulating α-granule secretion by restricting granule fusion.


Assuntos
Actinas , Plaquetas , Retração do Coágulo , Fator 2 de Liberação do Nucleotídeo Guanina , Animais , Actinas/metabolismo , Plaquetas/metabolismo , Exocitose/fisiologia , Hemostasia , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo
2.
Cell Commun Signal ; 21(1): 30, 2023 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-36737758

RESUMO

BACKGROUND: C3G is a guanine nucleotide exchange factor (GEF) that activates Rap1 to promote cell adhesion. Resting C3G is autoinhibited and the GEF activity is released by stimuli that signal through tyrosine kinases. C3G is activated by tyrosine phosphorylation and interaction with Crk adaptor proteins, whose expression is elevated in multiple human cancers. However, the molecular details of C3G activation and the interplay between phosphorylation and Crk interaction are poorly understood. METHODS: We combined biochemical, biophysical, and cell biology approaches to elucidate the mechanisms of C3G activation. Binding of Crk adaptor proteins to four proline-rich motifs (P1 to P4) in C3G was characterized in vitro using isothermal titration calorimetry and sedimentation velocity, and in Jurkat and HEK293T cells by affinity pull-down assays. The nucleotide exchange activity of C3G over Rap1 was measured using nucleotide-dissociation kinetic assays. Jurkat cells were also used to analyze C3G translocation to the plasma membrane and the C3G-dependent activation of Rap1 upon ligation of T cell receptors. RESULTS: CrkL interacts through its SH3N domain with sites P1 and P2 of inactive C3G in vitro and in Jurkat and HEK293T cells, and these sites are necessary to recruit C3G to the plasma membrane. However, direct stimulation of the GEF activity requires binding of Crk proteins to the P3 and P4 sites. P3 is occluded in resting C3G and is essential for activation, while P4 contributes secondarily towards complete stimulation. Tyrosine phosphorylation of C3G alone causes marginal activation. Instead, phosphorylation primes C3G lowering the concentration of Crk proteins required for activation and increasing the maximum activity. Unexpectedly, optimal activation also requires the interaction of CrkL-SH2 domain with phosphorylated C3G. CONCLUSION: Our study revealed that phosphorylation of C3G by Src and Crk-binding form a two-factor mechanism that ensures tight control of C3G activation. Additionally, the simultaneous SH2 and SH3N interaction of CrkL with C3G, required for the activation, reveals a novel adaptor-independent function of Crk proteins relevant to understanding their role in physiological signaling and their deregulation in diseases. Video abstract.


Assuntos
Fator 2 de Liberação do Nucleotídeo Guanina , Proteínas Nucleares , Humanos , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Células HEK293 , Proteínas Nucleares/metabolismo , Nucleotídeos/metabolismo , Proteínas Proto-Oncogênicas c-crk/metabolismo , Domínios de Homologia de src , Tirosina/metabolismo
3.
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
4.
Pathol Int ; 71(4): 255-260, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33709437

RESUMO

The expression of Ras-specific guanine nucleotide-releasing factor 2 (RasGRF2) in lung adenocarcinomas was examined using immunohistochemistry in relation to clinicopathological characteristics and prognosis. In comparison to low expression, high expression of RasGRF2 was more closely associated with poor prognosis. Interestingly, expression of phosphorylated epithelial cell transforming 2 (pECT2), which - like RasGRF2 - is also a guanine-nucleotide exchange factor, was also associated with prognosis, and patients with high expression of both RasGRF2 and pECT2 had a much poorer outcome than those who were negative for both.


Assuntos
Adenocarcinoma de Pulmão/patologia , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Prognóstico , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Fatores ras de Troca de Nucleotídeo Guanina/metabolismo
5.
Int J Mol Sci ; 22(24)2021 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-34947971

RESUMO

Crkl is a protein involved in the onset of several cancer pathologies that exerts its function only through its protein-protein interaction domains, a SH2 domain and two SH3 domains. SH3 domains are small protein interaction modules that mediate the binding and recognition of proline-rich sequences. One of the main physiological interactors of Crkl is C3G (also known as RAPGEF1), an interaction with key implications in regulating cellular growth and differentiation, cell morphogenesis and adhesion processes. Thus, understanding the interaction between Crkl and C3G is fundamental to gaining information about the molecular determinants of the several cancer pathologies in which these proteins are involved. In this paper, through a combination of fast kinetics at different experimental conditions and site-directed mutagenesis, we characterize the binding reaction between the N-SH3 domain of Crkl and a peptide mimicking a specific portion of C3G. Our results show a clear effect of pH on the stability of the complex, due to the protonation of negatively charged residues in the binding pocket of N-SH3. Our results are discussed under the light of previous work on SH3 domains.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Mutagênese Sítio-Dirigida/métodos , Proteínas Adaptadoras de Transdução de Sinal/genética , Sítios de Ligação , Adesão Celular , Diferenciação Celular , Proliferação de Células , Fator 2 de Liberação do Nucleotídeo Guanina/química , Humanos , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Eletricidade Estática
6.
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
7.
Biotechnol Lett ; 42(7): 1275-1286, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32236758

RESUMO

OBJECTIVE: To investigate the functions of eIF3b in chronic myelogenous leukemia (CML). METHODS: The expression of eIF3b was inhibited by transfecting aspecifically designed shRNA into the CML cell lines of TK-6 and K562. The CCK8 assay was conducted to determine cell viability, and flow cytometry was used to examine the change in the cell cycle and cell apoptosis. RNAsequencing was applied to screen the candidate targets of eIF3b to identify the underlying mechanisms of eIF3b.An in vivo tumour xenograft mouse model was established by injecting shRNA transfected cells into the NCG mice. The tumour size and body weight of mice were monitored every other day. The mice were sacrificed 2 weeks after the tumour cell injection. The expression of eIF3b and target genes in the tumour tissues were determined by immunohistochemical staining and Western blotting. RESULTS: The group with inhibited expression of eIF3b led to about 50% lower cell viability compared with that of the control group (P < 0.05). Flow cytometry suggested that the percentage of increase in apoptotic cells was eight times higher than those in control group for TK-6 and K562 cells (P < 0.05). However, the difference between the cell amounts in the S phase for the experiment and control groups was not significant. After RNAsequencing and further validation via qPCR, C3G was screened as the potential target of eIF3b involved in the cell proliferation and apoptosis of CML cell lines. Subsequent in vivo analysis proved that the inhibition of eIF3b suppressed tumour formation and decreased C3G expression, thereby indicating that C3G was the potential target of eIF3b. CONCLUSION: eIF3b is correlated with the cell proliferation and cell apoptosis of CML. Moreover, eIF3b regulation most probably occurs via regulating the expression of C3G.


Assuntos
Apoptose/fisiologia , Proliferação de Células/fisiologia , Fatores de Iniciação em Eucariotos/metabolismo , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Animais , Xenoenxertos , Humanos , Células K562 , Camundongos
8.
Cell Commun Signal ; 16(1): 101, 2018 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-30567575

RESUMO

BACKGROUND: Megakaryopoiesis allows platelet formation, which is necessary for coagulation, also playing an important role in different pathologies. However, this process remains to be fully characterized. C3G, an activator of Rap1 GTPases, is involved in platelet activation and regulates several differentiation processes. METHODS: We evaluated C3G function in megakaryopoiesis using transgenic mouse models where C3G and C3GΔCat (mutant lacking the GEF domain) transgenes are expressed exclusively in megakaryocytes and platelets. In addition, we used different clones of K562, HEL and DAMI cell lines with overexpression or silencing of C3G or GATA-1. RESULTS: We found that C3G participates in the differentiation of immature hematopoietic cells to megakaryocytes. Accordingly, bone marrow cells from transgenic C3G, but not those from transgenic C3GΔCat mice, showed increased expression of the differentiation markers CD41 and CD61, upon thrombopoietin treatment. Furthermore, C3G overexpression increased the number of CD41+ megakaryocytes with high DNA content. These results are supported by data obtained in the different models of megakaryocytic cell lines. In addition, it was uncovered GATA-1 as a positive regulator of C3G expression. Moreover, C3G transgenic megakaryocytes from fresh bone marrow explants showed increased migration from the osteoblastic to the vascular niche and an enhanced ability to form proplatelets. Although the transgenic expression of C3G in platelets did not alter basal platelet counts, it did increase slightly those induced by TPO injection in vivo. Moreover, platelet C3G induced adipogenesis in the bone marrow under pathological conditions. CONCLUSIONS: All these data indicate that C3G plays a significant role in different steps of megakaryopoiesis, acting through a mechanism dependent on its GEF activity.


Assuntos
Plaquetas/citologia , Diferenciação Celular , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Megacariócitos/citologia , Adipogenia , Linhagem Celular Tumoral , Humanos , Megacariócitos/metabolismo , Ploidias
9.
Mol Cell Neurosci ; 85: 127-147, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28966131

RESUMO

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.


Assuntos
Diferenciação Celular/fisiologia , Giro Denteado/citologia , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , ras-GRF1/metabolismo , Animais , Giro Denteado/metabolismo , Proteína Duplacortina , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nestina
11.
Immunity ; 29(4): 578-88, 2008 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-18835194

RESUMO

Many cellular responses, such as autoimmunity and cytotoxicity, are controlled by receptors with cytoplasmic immunoreceptor tyrosine-based inhibition motifs (ITIMs). Here, we showed that binding of inhibitory natural killer (NK) cell receptors to human leukocyte antigen (HLA) class I on target cells induced tyrosine phosphorylation of the adaptor Crk, concomitant with dephosphorylation of the guanine exchange factor Vav1. Furthermore, Crk dissociated from the guanine exchange factor C3G and bound to the tyrosine kinase c-Abl during inhibition. Membrane targeting of a tyrosine-mutated form of Crk could overcome inhibition of NK cell cytotoxicity, providing functional evidence that Crk phosphorylation contributes to inhibition. The specific phosphorylation of Crk and its dissociation from a signaling complex, observed here with two types of inhibitory receptors, expands the signaling potential of the large ITIM-receptor family and reveals an unsuspected component of the inhibitory mechanism.


Assuntos
Células Matadoras Naturais/imunologia , Proteínas Proto-Oncogênicas c-crk/metabolismo , Receptores KIR/metabolismo , Motivos de Aminoácidos/imunologia , Animais , Proteína Substrato Associada a Crk/imunologia , Proteína Substrato Associada a Crk/metabolismo , Fator 2 de Liberação do Nucleotídeo Guanina/imunologia , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Antígenos HLA/imunologia , Antígenos HLA/metabolismo , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Células Matadoras Naturais/metabolismo , Subfamília D de Receptores Semelhantes a Lectina de Células NK/imunologia , Subfamília D de Receptores Semelhantes a Lectina de Células NK/metabolismo , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 6/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Proteínas Proto-Oncogênicas c-abl/imunologia , Proteínas Proto-Oncogênicas c-abl/metabolismo , Proteínas Proto-Oncogênicas c-cbl/imunologia , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Proteínas Proto-Oncogênicas c-crk/imunologia , Proteínas Proto-Oncogênicas c-vav/imunologia , Proteínas Proto-Oncogênicas c-vav/metabolismo , Ratos , Receptores KIR/imunologia , Receptores de Células Matadoras Naturais/imunologia , Receptores de Células Matadoras Naturais/metabolismo , Antígenos HLA-E
12.
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
13.
Cell Commun Signal ; 14(1): 21, 2016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27620165

RESUMO

BACKGROUND: Vasodilator-Stimulated Phosphoprotein (VASP) is involved in the inhibition of agonist-induced platelet aggregation by cyclic nucleotides and the adhesion of platelets to the vascular wall. αIIbß3 is the main integrin responsible for platelet activation and Rap1b plays a key role in integrin signalling. We investigated whether VASP is involved in the regulation of Rap1b in platelets since VASP-null platelets exhibit augmented adhesion to endothelial cells in vivo. METHODS: Washed platelets from wild type and VASP-deficient mice were stimulated with thrombin, the purinergic receptors agonist ADP, or the thromboxane A2 receptor agonist U46619 and Rap1b activation was measured using the GST-RalGDS-RBD binding assay. Interaction of VASP and Crkl was investigated by co-immunoprecipitation, confocal microscopy, and pull-down assays using Crkl domains expressed as GST-fusion proteins. RESULTS: Surprisingly, we found that activation of Rap1b in response to thrombin, ADP, or U46619 was significantly reduced in platelets from VASP-null mice compared to platelets from wild type mice. However, inhibition of thrombin-induced activation of Rap1b by nitric oxide (NO) was similar in platelets from wild type and VASP-null mice indicating that the NO/cGMP/PKG pathway controls inhibition of Rap1b independently from VASP. To understand how VASP regulated Rap1b, we investigated association between VASP and the Crk-like protein (Crkl), an adapter protein which activates the Rap1b guanine nucleotide exchange factor C3G. We demonstrated the formation of a Crkl/VASP complex by showing that: 1) Crkl co-immunoprecipitated VASP from platelet lysates; 2) Crkl and VASP dynamically co-localized at actin-rich protrusions reminiscent of focal adhesions, filopodia, and lamellipodia upon platelet spreading on fibronectin; 3) recombinant VASP bound directly to the N-terminal SH3 domain of Crkl; 4) Protein Kinase A (PKA) -mediated VASP phosphorylation on Ser157 abrogated the binding of Crkl. CONCLUSIONS: We identified Crkl as a novel protein interacting with VASP in platelets. We propose that the C3G/Crkl/VASP complex plays a role in the regulation of Rap1b and this explains, at least in part, the reduced agonist-induced activation of Rap1b in VASP-null platelets. In addition, the fact that PKA-dependent VASP phosphorylation abrogated its interaction with Crkl may provide, at least in part, a rationale for the PKA-dependent inhibition of Rap1b and platelet aggregation.


Assuntos
Plaquetas/metabolismo , Moléculas de Adesão Celular/metabolismo , Proteínas dos Microfilamentos/metabolismo , Fosfoproteínas/metabolismo , Proteínas rap de Ligação ao GTP/metabolismo , Animais , Plaquetas/efeitos dos fármacos , Moléculas de Adesão Celular/genética , Células Cultivadas , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Humanos , Camundongos , Proteínas dos Microfilamentos/genética , Fosfoproteínas/genética , Ligação Proteica , Proteínas Proto-Oncogênicas c-crk/metabolismo , Agonistas Purinérgicos/farmacologia , Trombina/farmacologia
14.
J Immunol ; 193(8): 3966-77, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25225668

RESUMO

Crk adaptor proteins are key players in signal transduction from a variety of cell surface receptors. CrkI and CrkII, the two alternative spliced forms of CRK, possess an N-terminal Src homology 2 domain, followed by a Src homology 3 (SH3) domain, whereas CrkII possesses in addition a C-terminal linker region plus a SH3 domain, which operate as regulatory moieties. In this study, we investigated the ability of immunophilins, which function as peptidyl-prolyl isomerases, to regulate Crk proteins in human T lymphocytes. We found that endogenous CrkII, but not CrkI, associates with the immunophilins, cyclophilin A, and 12-kDa FK506-binding protein, in resting human Jurkat T cells. In addition, cyclophilin A increased Crk SH3 domain-binding guanine-nucleotide releasing factor (C3G) binding to CrkII, whereas inhibitors of immunophilins, such as cyclosporine A (CsA) and FK506, inhibited CrkII, but not CrkI association with C3G. Expression in Jurkat T cells of phosphorylation indicator of Crk chimeric unit plasmid, a plasmid encoding the human CrkII1-236 sandwiched between cyan fluorescent protein and yellow fluorescent protein, demonstrated a basal level of fluorescence resonance energy transfer, which increased in response to cell treatment with CsA and FK506, reflecting increased trans-to-cis conversion of CrkII. Crk-C3G complexes are known to play an important role in integrin-mediated cell adhesion and migration. We found that overexpression of CrkI or CrkII increased adhesion and migration of Jurkat T cells. However, immunophilin inhibitors suppressed the ability of CrkII- but not CrkI-overexpressing cells to adhere to fibronectin-coated surfaces and migrate toward the stromal cell-derived factor 1α chemokine. The present data demonstrate that immunophilins regulate CrkII, but not CrkI activity in T cells and suggest that CsA and FK506 inhibit selected effector T cell functions via a CrkII-dependent mechanism.


Assuntos
Ciclofilina A/antagonistas & inibidores , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Proteínas Proto-Oncogênicas c-crk/metabolismo , Linfócitos T/imunologia , Adesão Celular/imunologia , Movimento Celular/imunologia , Quimiocina CXCL12/imunologia , Ciclofilina A/metabolismo , Ciclosporina/farmacologia , Humanos , Células Jurkat , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Plasmídeos/genética , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Transdução de Sinais/imunologia , Tacrolimo/farmacologia , Domínios de Homologia de src/genética
15.
J Biol Chem ; 289(23): 16551-64, 2014 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-24755227

RESUMO

Ras-GRF1 (GRF1) and Ras-GRF2 (GRF2) constitute a family of similar calcium sensors that regulate synaptic plasticity. They are both guanine exchange factors that contain a very similar set of functional domains, including N-terminal pleckstrin homology, coiled-coil, and calmodulin-binding IQ domains and C-terminal Dbl homology Rac-activating domains, Ras-exchange motifs, and CDC25 Ras-activating domains. Nevertheless, they regulate different forms of synaptic plasticity. Although both GRF proteins transduce calcium signals emanating from NMDA-type glutamate receptors in the CA1 region of the hippocampus, GRF1 promotes LTD, whereas GRF2 promotes θ-burst stimulation-induced LTP (TBS-LTP). GRF1 can also mediate high frequency stimulation-induced LTP (HFS-LTP) in mice over 2-months of age, which involves calcium-permeable AMPA-type glutamate receptors. To add to our understanding of how proteins with similar domains can have different functions, WT and various chimeras between GRF1 and GRF2 proteins were tested for their abilities to reconstitute defective LTP and/or LTD in the CA1 hippocampus of Grf1/Grf2 double knock-out mice. These studies revealed a critical role for the GRF2 CDC25 domain in the induction of TBS-LTP by GRF proteins. In contrast, the N-terminal pleckstrin homology and/or coiled-coil domains of GRF1 are key to the induction of HFS-LTP by GRF proteins. Finally, the IQ motif of GRF1 determines whether a GRF protein can induce LTD. Overall, these findings show that for the three forms of synaptic plasticity that are regulated by GRF proteins in the CA1 hippocampus, specificity is encoded in only one or two domains, and a different set of domains for each form of synaptic plasticity.


Assuntos
Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Transdução de Sinais , ras-GRF1/metabolismo , Animais , Sinalização do Cálcio , Hipocampo/metabolismo , Camundongos
16.
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
17.
Mol Cell Proteomics ; 11(8): 355-69, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22499769

RESUMO

The cellular proto-oncogene c-Src is a nonreceptor tyrosine kinase involved in cell growth and cytoskeletal regulation. Despite being dysregulated in a variety of human cancers, its precise functions are not fully understood. Identification of the substrates of c-Src remains a major challenge, because there is no simple way to directly stimulate its activity. Here we combine the chemical rescue of mutant c-Src and global quantitative phosphoproteomics to obtain the first high resolution snapshot of the range of tyrosine phosphorylation events that occur in the cell immediately after specific c-Src stimulation. After enrichment by anti-phosphotyrosine antibodies, we identified 29 potential novel c-Src substrate proteins. Tyrosine phosphopeptide mapping allowed the identification of 382 nonredundant tyrosine phosphopeptides on 213 phosphoproteins. Stable isotope labeling of amino acids in cell culture-based quantitation allowed the detection of 97 nonredundant tyrosine phosphopeptides whose level of phosphorylation is increased by c-Src. A large number of previously uncharacterized c-Src putative protein targets and phosphorylation sites are presented here, a majority of which play key roles in signaling and cytoskeletal networks, particularly in cell adhesion. Integrin signaling and focal adhesion kinase signaling pathway are two of the most altered pathways upon c-Src activation through chemical rescue. In this context, our study revealed the temporal connection between c-Src activation and the GTPase Rap1, known to stimulate integrin-dependent adhesion. Chemical rescue of c-Src provided a tool to dissect the spatiotemporal mechanism of activation of the Rap1 guanine exchange factor, C3G, one of the identified potential c-Src substrates that plays a role in focal adhesion signaling. In addition to unveiling the role of c-Src in the cell and, specifically, in the Crk-C3G-Rap1 pathway, these results exemplify a strategy for obtaining a comprehensive understanding of the functions of nonreceptor tyrosine kinases with high specificity and kinetic resolution.


Assuntos
Fosfoproteínas/análise , Proteômica/métodos , Tirosina/metabolismo , Quinases da Família src/metabolismo , Animais , Western Blotting , Proteína Tirosina Quinase CSK , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Adesões Focais , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Humanos , Imidazóis/química , Espectrometria de Massas , Camundongos , Camundongos Knockout , Microscopia Confocal , Mutação , Fosfopeptídeos/análise , Fosforilação , Ligação Proteica , Proto-Oncogene Mas , Transdução de Sinais , Especificidade por Substrato , Tirosina/química , Tirosina/genética , Proteínas rap1 de Ligação ao GTP/metabolismo , Quinases da Família src/química , Quinases da Família src/genética
18.
J Cell Sci ; 124(Pt 13): 2153-64, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21628423

RESUMO

Establishing new adhesions at the extended leading edges of motile cells is essential for stable polarity and persistent motility. Despite recent identification of signaling pathways that mediate polarity and chemotaxis in neutrophils, little is known about molecular mechanisms governing cell-extracellular-matrix (ECM) adhesion in these highly polarized and rapidly migrating cells. Here, we describe a signaling pathway in neutrophils that is essential for localized integrin activation, leading edge attachment and persistent migration during chemotaxis. This pathway depends upon G(i)-protein-mediated activation and leading edge recruitment of Lyn, a non-receptor tyrosine kinase belonging to the Src kinase family. We identified the small GTPase Rap1 as a major downstream effector of Lyn to regulate neutrophil adhesion during chemotaxis. Depletion of Lyn in neutrophil-like HL-60 cells prevented chemoattractant-induced Rap1 activation at the leading edge of the cell, whereas ectopic expression of Rap1 largely rescued the defects induced by Lyn depletion. Furthermore, Lyn controls spatial activation of Rap1 by recruiting the CrkL-C3G protein complex to the leading edge. Together, these results provide novel mechanistic insights into the poorly understood signaling network that controls leading edge adhesion during chemotaxis of neutrophils, and possibly other amoeboid cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adesão Celular/fisiologia , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Neutrófilos/citologia , Neutrófilos/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Ligação a Telômeros/metabolismo , Quinases da Família src/metabolismo , Linhagem Celular , Movimento Celular , Quimiotaxia/fisiologia , Matriz Extracelular/metabolismo , Humanos , Ligação Proteica , Complexo Shelterina , Transdução de Sinais , Quinases da Família src/genética
19.
Cell Biol Int ; 37(10): 1106-13, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23686869

RESUMO

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.


Assuntos
Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animais , Apoptose/genética , Proliferação de Células , Sobrevivência Celular/genética , Citometria de Fluxo , Fator 2 de Liberação do Nucleotídeo Guanina/genética , Humanos , Integrinas/metabolismo , Miócitos Cardíacos/enzimologia , Fosforilação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Coloração e Rotulagem
20.
Biochim Biophys Acta ; 1813(3): 456-65, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21223981

RESUMO

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.


Assuntos
Neoplasias da Mama/patologia , Carcinoma/patologia , Citoesqueleto/patologia , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Neuritos/patologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Citoesqueleto/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fator 2 de Liberação do Nucleotídeo Guanina/genética , Humanos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Regulação para Cima , beta Catenina/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa