Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Vasc Res ; 58(5): 277-285, 2021.
Article in English | MEDLINE | ID: mdl-33951626

ABSTRACT

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.


Subject(s)
Guanine Nucleotide-Releasing Factor 2/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Phosphates/pharmacology , Animals , Cells, Cultured , Guanine Nucleotide-Releasing Factor 2/genetics , Humans , Mice, Inbred C57BL , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Phosphorylation , Signal Transduction , Sodium-Phosphate Cotransporter Proteins, Type III/metabolism
2.
Mol Biol Cell ; 29(9): 1111-1124, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29496966

ABSTRACT

C3G (Crk SH3 domain binding guanine nucleotide releasing factor) (Rap guanine nucleotide exchange factor 1), essential for mammalian embryonic development, is ubiquitously expressed and undergoes regulated nucleocytoplasmic exchange. Here we show that C3G localizes to SC35-positive nuclear speckles and regulates splicing activity. Reversible association of C3G with speckles was seen on inhibition of transcription and splicing. C3G shows partial colocalization with SC35 and is recruited to a chromatin and RNase-sensitive fraction of speckles. Its presence in speckles is dependent on intact cellular actin cytoskeleton and is lost on expression of the kinase Clk1. Rap1, a substrate of C3G, is also present in nuclear speckles, and inactivation of Rap signaling by expression of GFP-Rap1GAP alters speckle morphology and number. Enhanced association of C3G with speckles is seen on glycogen synthase kinase 3 beta inhibition or differentiation of C2C12 cells to myotubes. CRISPR/Cas9-mediated knockdown of C3G resulted in altered splicing activity of an artificial gene as well as endogenous CD44. C3G knockout clones of C2C12 as well as MDA-MB-231 cells showed reduced protein levels of several splicing factors compared with control cells. Our results identify C3G and Rap1 as novel components of nuclear speckles and a role for C3G in regulating cellular RNA splicing activity.


Subject(s)
Guanine Nucleotide-Releasing Factor 2/metabolism , Guanine Nucleotide-Releasing Factor 2/physiology , RNA Splicing/physiology , Animals , Cell Differentiation , Cell Line , Cell Line, Tumor , Cell Nucleus Structures/physiology , Guanine Nucleotide Exchange Factors/metabolism , Humans , Nuclear Proteins , Protein Binding , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases , RNA Processing, Post-Transcriptional/physiology , RNA, Messenger/metabolism , Serine-Arginine Splicing Factors/metabolism , Serine-Arginine Splicing Factors/physiology , Shelterin Complex , Signal Transduction , Spliceosomes , Telomere-Binding Proteins/metabolism
3.
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
4.
Am J Physiol Renal Physiol ; 304(7): F831-9, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23364803

ABSTRACT

Exchange proteins directly activated by cAMP [Epac(s)] were discovered more than a decade ago as new sensors for the second messenger cAMP. The Epac family members, including Epac1 and Epac2, are guanine nucleotide exchange factors for the Ras-like small GTPases Rap1 and Rap2, and they function independently of protein kinase A. Given the importance of cAMP in kidney homeostasis, several molecular and cellular studies using specific Epac agonists have analyzed the role and regulation of Epac proteins in renal physiology and pathophysiology. The specificity of the functions of Epac proteins may depend upon their expression and localization in the kidney as well as their abundance in the microcellular environment. This review discusses recent literature data concerning the involvement of Epac in renal tubular transport physiology and renal glomerular cells where various signaling pathways are known to be operative. In addition, the potential role of Epac in kidney disorders, such as diabetic kidney disease and ischemic kidney injury, is discussed.


Subject(s)
Guanine Nucleotide Exchange Factors/physiology , Kidney Glomerulus/physiology , Kidney Tubules/physiology , Acute Kidney Injury/chemically induced , Acute Kidney Injury/physiopathology , Aquaporin 2/physiology , Cisplatin/adverse effects , Diabetic Nephropathies/physiopathology , Guanine Nucleotide-Releasing Factor 2/physiology , H(+)-K(+)-Exchanging ATPase/physiology , Humans , Kidney/physiology , Kidney Diseases/physiopathology , Kidney Tubules/physiopathology , Membrane Transport Proteins/physiology , Reperfusion Injury/physiopathology , Urea Transporters
5.
Neurochem Int ; 62(6): 848-53, 2013 May.
Article in English | MEDLINE | ID: mdl-23416045

ABSTRACT

Rap1 signaling is important for migration, differentiation, axonal growth, and during neuronal polarity. Rap1 can be activated by external stimuli, which in turn regulates specific guanine nucleotide exchange factors such as C3G, among others. Cdk5 functions are also important to neuronal migration and differentiation. Since we found that pharmacological inhibition of Cdk5 by using roscovitine reduced Rap1 protein levels in COS-7 cells and also C3G contains three putative phosphorylation sites for Cdk5, we examined whether the Cdk5-dependent phosphorylation of C3G could affect Rap1 expression and activity. We co-transfected C3G and tet-OFF system for p35 over-expression, an activator of Cdk5 activity into COS-7 cells, and then we evaluated phosphorylation in serine residues in C3G by immunoprecipitation and Western blot. We found that p35 over-expression increased C3G-serine-phosphorylation while inhibition of p35 expression by tetracycline or inhibition of Cdk5 activity with roscovitine decreased it. Interestingly, we found that MG-132, a proteasome inhibitor, rescue Rap1 protein levels in the presence of roscovitine. Besides, C3G-serine-phosphorylation and Rap1 protein levels were reduced in brain from Cdk5(-/-) as compared with the Cdk5(+/+) brain. Finally, we found that p35 over-expression increased Rap1 activity while inhibition of p35 expression by tetracycline or roscovitine decreased Rap1 activity. These results suggest that Cdk5-mediated serine-phosphorylation of C3G may control Rap1 stability and activity, and this may potentially impact various neuronal functions such as migration, differentiation, and polarity.


Subject(s)
Cyclin-Dependent Kinase 5/physiology , rap1 GTP-Binding Proteins/physiology , Amino Acid Sequence , Animals , Blotting, Western , COS Cells , Chlorocebus aethiops , Cysteine Proteinase Inhibitors , Guanine Nucleotide-Releasing Factor 2/physiology , Immunoprecipitation , Interleukin-12 Subunit p35/biosynthesis , Leupeptins/pharmacology , Membrane Fusion Proteins , Mice , Mice, Knockout , Molecular Sequence Data , Neurons/drug effects , Phosphorylation , Real-Time Polymerase Chain Reaction , Tetracycline/pharmacology , Transfection
6.
Contrib Nephrol ; 172: 50-62, 2011.
Article in English | MEDLINE | ID: mdl-21893988

ABSTRACT

Endothelins (ETs), and particularly ET-1, activate a complex network of interconnected signaling cascades in mesangial glomerular cells, which play an important role in the physiology and pathophysiology of the glomerulus. Excessive stimulation of ET-1 production by mesangial cells results in activation of a wide variety of signaling pathways in the renal mesangium, which is at least partially responsible for glomerular damage in the setting of diabetes, hypertension, and glomerulonephritis. Mesangial cells express both types of ET receptors (ET(A)-R and ET(B)-R), which are G protein-coupled receptors. ET-1 induces mobilization of Ca(2+); activation of phospholipases A, C, and D; activation of protein kinase C; GTP-loading of several families of small GTPases; and activation of intracellular tyrosine kinases resulting in protein tyrosine phosphorylation of adaptor, scaffolding, and signaling proteins. ET-1-triggered posttranslational modification of signaling molecules sets the base for the formation of multiunit signaling complexes which define the specificity of ET signaling. Long-term effects of ET-1 are also mediated via increased expression of particular signaling proteins. It is likely that ET-1 acts via ET(A)-R to trigger the contraction of mesangial cells, which decreases glomerular filtration area and reduces the glomerular filtration rate, promoting impaired renal function. Proliferation of mesangial cells is observed in the progress of several types of glomerulonephritis. ET-1 is a potent mitogen of mesangial cells and the ability of ET-1 to support mesangial cell proliferation is likely to be associated with both recruitment of cytoplasmic tyrosine kinases which activate the Shc-Sos-Ras-Raf-MEK-ERK signaling pathway and transactivation of the EGF receptor. The guanine nucleotide exchange factor ßPix and the adaptor protein p66(Shc) are important players in Akt-independent inactivation of FOXO3a transcription factor. This results in the depletion of the inhibitor of cell cycle progression p27(kip1), and promotion of mesangial cell proliferation. Plentiful evidence suggests an essential role of ET-1-signaling and action in the renal mesangium for renal biology and pathobiology.


Subject(s)
Endothelins/physiology , Glomerular Mesangium/physiology , Signal Transduction/physiology , Animals , Apoptosis , Calcium/metabolism , Cell Proliferation , Extracellular Matrix Proteins/biosynthesis , Glomerular Mesangium/cytology , Guanine Nucleotide-Releasing Factor 2/physiology , Humans , Mesangial Cells/physiology
7.
Oncogene ; 29(48): 6378-89, 2010 Dec 02.
Article in English | MEDLINE | ID: mdl-20729917

ABSTRACT

The v-Crk oncogene product consists of two protein interaction modules, a Src homology 2 (SH2) domain and a Src homology 3 (SH3) domain. Overexpression of CrkI, the cellular homolog of v-Crk, transforms mouse fibroblasts, and elevated CrkI expression is observed in several human cancers. The SH2 and SH3 domains of Crk are required for transformation, but the identity of the critical cellular binding partners is not known. A number of candidate Crk SH3-binding proteins have been identified, including the nonreceptor tyrosine kinases c-Abl and Arg, and the guanine nucleotide exchange proteins C3G, SOS1 and DOCK180. The aim of this study is to determine which of these are required for transformation by CrkI. We found that short hairpin RNA-mediated knockdown of C3G or SOS1 suppressed anchorage-independent growth of NIH-3T3 cells overexpressing CrkI, whereas knockdown of SOS1 alone was sufficient to suppress tumor formation by these cells in nude mice. Knockdown of C3G was sufficient to revert morphological changes induced by CrkI expression. By contrast, knockdown of Abl family kinases or their inhibition with imatinib enhanced anchorage-independent growth and tumorigenesis induced by Crk. These results show that SOS1 is essential for CrkI-induced fibroblast transformation, and also reveal a surprising negative role for Abl kinases in Crk transformation.


Subject(s)
Cell Transformation, Neoplastic , Proto-Oncogene Proteins c-crk/physiology , src Homology Domains , Animals , Apoptosis , Guanine Nucleotide-Releasing Factor 2/physiology , HEK293 Cells , Humans , Male , Mice , Mice, Nude , NIH 3T3 Cells , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/physiology , Proto-Oncogene Proteins c-crk/chemistry , SOS1 Protein/physiology , Signal Transduction
8.
Development ; 135(12): 2139-49, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18506028

ABSTRACT

Neuronal migration is integral to the development of the cerebral cortex and higher brain function. Cortical neuron migration defects lead to mental disorders such as lissencephaly and epilepsy. Interaction of neurons with their extracellular environment regulates cortical neuron migration through cell surface receptors. However, it is unclear how the signals from extracellular matrix proteins are transduced intracellularly. We report here that mouse embryos lacking the Ras family guanine nucleotide exchange factor, C3G (Rapgef1, Grf2), exhibit a cortical neuron migration defect resulting in a failure to split the preplate into marginal zone and subplate and a failure to form a cortical plate. C3G-deficient cortical neurons fail to migrate. Instead, they arrest in a multipolar state and accumulate below the preplate. The basement membrane is disrupted and radial glial processes are disorganised and lack attachment in C3G-deficient brains. C3G is activated in response to reelin in cortical neurons, which, in turn, leads to activation of the small GTPase Rap1. In C3G-deficient cells, Rap1 GTP loading in response to reelin stimulation is reduced. In conclusion, the Ras family regulator C3G is essential for two aspects of cortex development, namely radial glial attachment and neuronal migration.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/embryology , Guanine Nucleotide-Releasing Factor 2/physiology , Neuroglia/physiology , Neurons/physiology , Animals , Cell Adhesion/physiology , Cell Movement/physiology , Cells, Cultured , Crosses, Genetic , Embryo, Mammalian , Guanine Nucleotide-Releasing Factor 2/genetics , Mice , Mice, Inbred CBA , Mice, Inbred Strains , Mutation , Organ Culture Techniques , Reelin Protein , Stem Cells/cytology
9.
Genetics ; 177(4): 2445-56, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17947423

ABSTRACT

In Drosophila melanogaster oocytes, the C(3)G protein comprises the transverse filaments (TFs) of the synaptonemal complex (SC). Like other TF proteins, such as Zip1p in yeast and SCP1 in mammals, C(3)G is composed of a central coiled-coil-rich domain flanked by N- and C-terminal globular domains. Here, we analyze in-frame deletions within the N- and C-terminal regions of C(3)G in Drosophila oocytes. As is the case for Zip1p, a C-terminal deletion of C(3)G fails to attach to the lateral elements of the SC. Instead, this C-terminal deletion protein forms a large cylindrical polycomplex structure. EM analysis of this structure reveals a polycomplex of concentric rings alternating dark and light bands. However, unlike both yeast and mammals, all three proteins deleted for N-terminal regions completely abolished both SC and polycomplex formation. Both the N- and C-terminal deletions significantly reduce or abolish meiotic recombination similarly to c(3)G null homozygotes. To explain these data, we propose that in Drosophila the N terminus, but not the C-terminal globular domain, of C(3)G is critical for the formation of antiparallel pairs of C(3)G homodimers that span the central region and thus for assembly of complete TFs, while the C terminus is required to affix these homodimers to the lateral elements.


Subject(s)
Chromosome Pairing , Drosophila Proteins/physiology , Guanine Nucleotide-Releasing Factor 2/physiology , Recombination, Genetic , Animals , Dimerization , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster , Guanine Nucleotide-Releasing Factor 2/chemistry , Guanine Nucleotide-Releasing Factor 2/genetics , Meiosis , Microscopy, Electron , Oocytes/chemistry , Oocytes/ultrastructure , Protein Structure, Tertiary , Sequence Deletion
10.
EMBO J ; 25(15): 3652-63, 2006 Aug 09.
Article in English | MEDLINE | ID: mdl-16858399

ABSTRACT

The mechanisms regulating the size of the cerebral cortex are poorly understood. Here, we demonstrate that the Rap1 guanine nucleotide exchange factor, C3G (Grf2, Rapgef1), controls the size of the cerebral precursor population. Mice lacking C3G show overproliferation of the cortical neuroepithelium. C3G-deficient neuroepithelial cells accumulate nuclear beta-catenin and fail to exit the cell cycle in vivo. C3G mutant neural precursor cells fail to activate Rap1, exhibit activation of Akt/PKB, inhibition of the beta-catenin-degrading enzyme, Gsk3beta and accumulation of cytosolic and nuclear beta-catenin when exposed to growth factors, in vitro. Our results show that the size of the cortical neural precursor population is controlled by C3G-mediated inhibition of the Ras signalling pathway.


Subject(s)
Cerebral Cortex/embryology , Guanine Nucleotide-Releasing Factor 2/physiology , Neurons/cytology , Animals , Cell Nucleus/metabolism , Cell Proliferation , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Cytosol/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 2/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Guanine Nucleotide-Releasing Factor 2/genetics , Mice , Mitogens/metabolism , Oncogene Protein v-akt/metabolism , Phosphorylation , Signal Transduction , beta Catenin/metabolism , rap1 GTP-Binding Proteins/metabolism
11.
J Neurosci ; 26(6): 1721-9, 2006 Feb 08.
Article in English | MEDLINE | ID: mdl-16467520

ABSTRACT

NMDA-type glutamate receptors (NMDARs) contribute to many forms of long-term potentiation (LTP) and long-term depression (LTD). NMDARs are heteromers containing calcium-permeating neuronal receptor 1 (NR1) subunits and a variety of NR2 subunits. Evidence suggests that, in the CA1 region of the hippocampus, NR2A-containing NMDARs promote LTP whereas NR2B-containing receptors promote LTD. However, the calcium sensors that distinguish between these signals to promote the appropriate form of synaptic plasticity are not known. Ras-guanine nucleotide-releasing factor 1 (Ras-GRF1) and Ras-GRF2 are highly similar calcium-stimulated exchange factors that activate Ras and Rac GTPases. Here, using a set of Ras-GRF knock-out mice, we show that Ras-GRF2 contributes predominantly to the induction of NMDAR-dependent LTP, whereas Ras-GRF1 contributes predominantly to the induction of NMDAR-dependent LTD in the CA1 region of the hippocampus of postpubescent mice (postnatal days 25-36). In contrast, neither Ras-GRF protein influences synaptic plasticity in prepubescent mice (postnatal days 14-18). Ras-GRF2 mediates signaling from (R)-[(S)-1-(4-bromo-phenyl)-ethylamino]-(2,3-dioxo-1,2,3,4-tetrahydroquinoxalin-5-yl)-methyl-phosphonic acid-sensitive (NVP-AAM077-sensitive) (NR2A-containing) NMDARs to the Ras effector extracellular signal-related protein kinase 1/2 (Erk1/2) mitogen-activated protein (MAP) kinase, a promoter of NMDAR-induced LTP at this site. In contrast, Ras-GRF1 mediates signaling from ifenprodil-sensitive (NR2B-containing) NMDARs to the Rac effector p38 MAP kinase, a promoter of LTD. These findings show that, despite their similar functional domain organization, Ras-GRF1 and Ras-GRF2 mediate opposing forms of synaptic plasticity by coupling different classes of NMDARs to distinct MAP kinase pathways. Moreover, the postnatal appearance of Ras-GRF-dependent LTP and LTD coincides with the emergence of hippocampal-dependent behavior, implying that Ras-GRF proteins contribute to forms of synaptic plasticity that are required specifically for mature hippocampal function.


Subject(s)
Guanine Nucleotide-Releasing Factor 2/physiology , Long-Term Potentiation/physiology , Long-Term Synaptic Depression/physiology , ras-GRF1/physiology , Animals , Excitatory Postsynaptic Potentials , Guanine Nucleotide-Releasing Factor 2/deficiency , Guanine Nucleotide-Releasing Factor 2/genetics , Hippocampus/physiology , MAP Kinase Signaling System/physiology , Mice , Mice, Knockout , Neuronal Plasticity/physiology , Receptors, N-Methyl-D-Aspartate/physiology , ras-GRF1/deficiency , ras-GRF1/genetics
12.
Oncogene ; 20(41): 5908-12, 2001 Sep 13.
Article in English | MEDLINE | ID: mdl-11593397

ABSTRACT

We investigated the attachment and spreading of v-Crk-transformed cells, v-Crk3Y1, on fibronectin. Transformation by v-Crk virtually suppressed the spreading, but not the attachment, of cells on fibronectin. This suppression of cell spreading was not correlated with the suppression of integrin alpha5 and beta1 expression. However, the spreading of v-Crk3Y1 on fibronectin was dramatically restored by either expression of dominant-negative Ras or treatment with manumycin A, a Ras farnesyltransferase inhibitor. Moreover, both expression of dominant-negative MEK1 and treatment of cells with U0126, a MEK1 inhibitor, restored the cell spreading of v-Crk3Y1. In contrast, neither treatment with LY294002, a PI3K inhibitor, nor expression of dominant-negative C3G showed no effect on cell spreading on fibronectin. Taken together, our results suggest that, among multiple signaling pathways activated by v-Crk, the Ras-MEK1-MAP kinase cascade plays a pivotal role in the suppression of cell spreading on fibronectin, but C3G and the PI3 kinase do not.


Subject(s)
Cell Adhesion/physiology , Fibronectins/metabolism , Mitogen-Activated Protein Kinase Kinases/physiology , Mitogen-Activated Protein Kinases/physiology , Protein Serine-Threonine Kinases/physiology , Receptors, Fibronectin/metabolism , Retroviridae Proteins, Oncogenic/physiology , Signal Transduction/physiology , ras Proteins/physiology , Animals , Butadienes/pharmacology , Cell Adhesion/drug effects , Cell Line/cytology , Cell Line/drug effects , Cell Line, Transformed/drug effects , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Guanine Nucleotide-Releasing Factor 2/physiology , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Morpholines/pharmacology , Nitriles/pharmacology , Oncogene Protein v-crk , Phosphoinositide-3 Kinase Inhibitors , Polyenes/pharmacology , Polyunsaturated Alkamides , Protein Serine-Threonine Kinases/antagonists & inhibitors , Rats , Signal Transduction/drug effects , ras Proteins/antagonists & inhibitors
13.
J Immunol ; 164(4): 1800-6, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10657627

ABSTRACT

IFN-gamma transduces signals by activating the IFN-gamma receptor-associated Jak-1 and Jak-2 kinases and by inducing tyrosine phosphorylation and activation of the Stat-1 transcriptional activator. We report that IFN-gamma activates a distinct signaling cascade involving the c-cbl protooncogene product, CrkL adapter, and small G protein Rap1. During treatment of NB-4 human cells with IFN-gamma, c-cbl protooncogene product is rapidly phosphorylated on tyrosine and provides a docking site for the src homology 2 domain of CrkL, which also undergoes IFN-gamma-dependent tyrosine phosphorylation. CrkL then regulates activation of the guanine exchange factor C3G, with which it interacts constitutively via its N terminus src homology 3 domain. This results in the IFN-gamma-dependent activation of Rap1, a protein known to exhibit tumor suppressor activity and mediate growth inhibitory responses. In a similar manner, Rap1 is also activated in response to treatment of cells with type I IFNs (IFN-alpha, IFN-beta), which also engage CrkL in their signaling pathways. On the other hand, IFN-gamma does not induce formation of nuclear CrkL-Stat5 DNA-binding complexes, which are induced by IFN-alpha and IFN-beta, indicating that pathways downstream of CrkL are differentially regulated by different IFN subtypes. Taken altogether, our data demonstrate that, in addition to activating the Stat pathway, IFN-gamma activates a distinct signaling cascade that may play an important role in the generation of its growth inhibitory effects on target cells.


Subject(s)
Adaptor Proteins, Signal Transducing , Guanine Nucleotide-Releasing Factor 2/physiology , Interferon-gamma/physiology , Signal Transduction/immunology , Ubiquitin-Protein Ligases , Guanine Nucleotide-Releasing Factor 2/metabolism , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation , Protein Binding/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-cbl , Recombinant Fusion Proteins/metabolism , Tumor Cells, Cultured , src Homology Domains/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...