Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Biochim Biophys Acta Mol Cell Res ; 1868(5): 118964, 2021 04.
Article in English | MEDLINE | ID: mdl-33450305

ABSTRACT

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.


Subject(s)
Glycogen Synthase Kinase 3 beta/metabolism , Guanine Nucleotide-Releasing Factor 2/chemistry , Guanine Nucleotide-Releasing Factor 2/metabolism , Mutation , Myoblasts/cytology , Animals , COS Cells , Cell Differentiation , Cell Line , Chlorocebus aethiops , Cytoplasm/metabolism , Gene Expression Regulation , Guanine Nucleotide-Releasing Factor 2/genetics , HEK293 Cells , Humans , Mice , Muscle Development , Myoblasts/metabolism , Phosphorylation
2.
Mol Biol Cell ; 28(7): 984-995, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28148649

ABSTRACT

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.


Subject(s)
Euchromatin/physiology , Guanine Nucleotide-Releasing Factor 2/metabolism , Guanine Nucleotide-Releasing Factor 2/physiology , Histone Code/physiology , Actins/metabolism , Active Transport, Cell Nucleus/physiology , Animals , Cell Differentiation , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Euchromatin/metabolism , Fatty Acids, Unsaturated/metabolism , Glycogen Synthase Kinase 3/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Guanine Nucleotide-Releasing Factor 2/genetics , Mice , Muscle Development , Nuclear Localization Signals , Phosphorylation , Signal Transduction , Up-Regulation
3.
Genes Cancer ; 3(9-10): 564-77, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23486661

ABSTRACT

The guanine nucleotide exchange factor C3G (RAPGEF1) regulates proliferation, migration, and differentiation of cells and is essential for mammalian embryonic development. The molecular effectors of C3G dependent functions are poorly understood. Here we report that C3G functions as a negative regulator of ß-catenin, a major player in pathways known to be deregulated in human cancers. In mammalian cells, C3G is present in a complex with cellular ß-catenin. The proline rich Crk binding region of C3G and residues 90-525 of ß-catenin are sufficient for the interaction. Knockdown of cellular C3G stimulated, and its overexpression repressed, ß-catenin/TCF transcription activity. C3G acts by destabilizing ß-catenin protein and inhibiting its nuclear accumulation. Nuclear extracts of C3G overexpressing cells showed reduced binding to TCF consensus oligos. C3G exerts its effects independent of its function as an exchange factor. It also inhibits stability and activity of an N-terminal deletion construct of ß-catenin that is not subject to GSK3ß dependent phosphorylation, suggesting that C3G exerts its effect independent of GSK3ß. ß-catenin repression by C3G was not significantly altered in the presence of proteasome inhibitors, MG132 or lactacystin, suggesting that alternate mechanisms are engaged by C3G to cause ß-catenin turnover. C3G expression represses ß-catenin target gene expression, and stable clones of MCF-7 breast cancer cells expressing C3G showed reduced migration. Activation of cellular ß-catenin or expression of constitutively active ß-catenin resulted in reduced C3G expression, indicating that C3G gene expression is negatively regulated by ß-catenin. Our results identify a novel property of C3G in functioning as a negative regulator of ß-catenin signaling by promoting its degradation. In addition, we show that ß-catenin inhibits C3G expression, forming a feedback loop.

4.
Biosci Rep ; 31(4): 231-44, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21366540

ABSTRACT

C3G (Crk SH3-domain-binding guanine-nucleotide-releasing factor) is a ubiquitously expressed member of a class of molecules called GEFs (guanine-nucleotide-exchange factor) that activate small GTPases and is involved in pathways triggered by a variety of signals. It is essential for mammalian embryonic development and many cellular functions in adult tissues. C3G participates in regulating functions that require cytoskeletal remodelling such as adhesion, migration, maintenance of cell junctions, neurite growth and vesicle traffic. C3G is spatially and temporally regulated to act on Ras family GTPases Rap1, Rap2, R-Ras, TC21 and Rho family member TC10. Increased C3G protein levels are associated with differentiation of various cell types, indicating an important role for C3G in cellular differentiation. In signalling pathways, C3G serves functions dependent on catalytic activity as well as protein interaction and can therefore integrate signals necessary for the execution of more than one cellular function. This review summarizes our current knowledge of the biology of C3G with emphasis on its role as a transducer of signals to the actin cytoskeleton. Deregulated C3G may also contribute to pathogenesis of human disorders and therefore could be a potential therapeutic target.


Subject(s)
Actins/metabolism , Guanine Nucleotide-Releasing Factor 2/metabolism , Signal Transduction , Animals , Apoptosis , Cell Proliferation , Cell Survival , Cytoskeleton/metabolism , Guanine Nucleotide-Releasing Factor 2/chemistry , Guanine Nucleotide-Releasing Factor 2/genetics , Humans , Models, Biological , Monomeric GTP-Binding Proteins/metabolism
5.
Biochim Biophys Acta ; 1813(3): 456-65, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21223981

ABSTRACT

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.


Subject(s)
Breast Neoplasms/pathology , Carcinoma/pathology , Cytoskeleton/pathology , Guanine Nucleotide-Releasing Factor 2/metabolism , Neurites/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma/genetics , Carcinoma/metabolism , Cell Line, Tumor , Cell Movement , Cytoskeleton/metabolism , Female , Gene Expression Regulation, Neoplastic , Guanine Nucleotide Exchange Factors/metabolism , Guanine Nucleotide-Releasing Factor 2/genetics , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Up-Regulation , beta Catenin/metabolism , cdc42 GTP-Binding Protein/metabolism
6.
J Neurochem ; 107(5): 1424-35, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18957052

ABSTRACT

Neuronal differentiation involving neurite growth is dependent on environmental cues which are relayed by signalling pathways to actin cytoskeletal remodelling. C3G, the exchange factor for Rap1, functions in pathways leading to actin reorganization and filopodia formation, processes required during neurite growth. In the present study, we have analyzed the function of C3G, in regulating neuronal cell survival and plasticity. Human neuroblastoma cells, IMR-32 induced to differentiate by serum starvation or by treatment with nerve growth factor (NGF) or forskolin showed enhanced C3G protein levels. Transient over-expression of C3G stimulated neurite growth and also increased responsiveness to NGF and serum deprivation induced differentiation. C3G-induced neurite growth was dependent on both its catalytic and N-terminal regulatory domains, and on the functions of Cdc42 and Rap1. Knockdown of C3G using small hairpin RNA inhibited forskolin and NGF-induced morphological differentiation of IMR-32 cells. Forskolin-induced differentiation was dependent on catalytic activity of C3G. Forskolin and NGF treatment resulted in phosphorylation of C3G at Tyr504 predominantly in the Golgi. C3G expression induced the cell cycle inhibitor p21 and C3G knockdown enhanced cell death in response to serum starvation. These findings demonstrate a novel function for C3G in regulating survival and differentiation of human neuroblastoma cells.


Subject(s)
Cell Differentiation/drug effects , Guanine Nucleotide-Releasing Factor 2/metabolism , Nerve Growth Factor/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Colforsin/pharmacology , Culture Media, Serum-Free/pharmacology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Green Fluorescent Proteins/biosynthesis , Guanine Nucleotide-Releasing Factor 2/genetics , Humans , Neurites/drug effects , Neurites/metabolism , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , RNA, Small Interfering/genetics , Transfection , cdc42 GTP-Binding Protein/metabolism , rap1 GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...