Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Dev Genes Evol ; 230(3): 227-238, 2020 05.
Article in English | MEDLINE | ID: mdl-32198667

ABSTRACT

Across the Bilateria, FGF/FGFR signaling is critical for normal development, and in both Drosophila and vertebrates, docking proteins are required to connect activated FGFRs with downstream pathways. While vertebrates use Frs2 to dock FGFR to the RAS/MAPK or PI3K pathways, the unrelated protein, downstream of FGFR (Dof/stumps/heartbroken), fulfills the corresponding function in Drosophila. To better understand the evolution of the signaling pathway downstream of FGFR, the available sequence databases were screened to identify Frs2, Dof, and other key pathway components in phyla that diverged early in animal evolution. While Frs2 homologues were detected only in members of the Bilateria, canonical Dof sequences (containing Dof, ankyrin, and SH2/SH3 domains) were present in cnidarians as well as bilaterians (but not in other animals or holozoans), correlating with the appearance of FGFR. Although these data suggested that Dof coupling might be ancestral, gene expression analysis in the cnidarian Hydra revealed that Dof is not upregulated in the zone of strong FGFRa and FGFRb expression at the bud base, where FGFR signaling controls detachment. In contrast, transcripts encoding other, known elements of FGFR signaling in Bilateria, namely the FGFR adaptors Grb2 and Crkl, which are acting downstream of Dof (and Frs2), as well as the guanyl nucleotide exchange factor Sos, and the tyrosine phosphatase Csw/Shp2, were strongly upregulated at the bud base. Our expression analysis, thus, identified transcriptional upregulation of known elements of FGFR signaling at the Hydra bud base indicating a highly conserved toolkit. Lack of transcriptional Dof upregulation raises the interesting question, whether Hydra FGFR signaling requires either of the docking proteins known from Bilateria.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Biological Evolution , Hydra/genetics , Hydra/metabolism , Receptors, Fibroblast Growth Factor/physiology , Animals , Cnidaria/genetics , Cnidaria/metabolism , GRB2 Adaptor Protein/physiology , Gene Expression Regulation, Developmental , Phylogeny , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Signal Transduction , Son of Sevenless Proteins/physiology
2.
PLoS Pathog ; 15(1): e1007535, 2019 01.
Article in English | MEDLINE | ID: mdl-30615681

ABSTRACT

MicroRNAs (miRNAs) are post-transcriptional regulatory RNAs that can modulate cell signaling and play key roles in cell state transitions. Epstein-Barr virus (EBV) expresses >40 viral miRNAs that manipulate both viral and cellular gene expression patterns and contribute to reprogramming of the host environment during infection. Here, we identified a subset of EBV miRNAs that desensitize cells to B cell receptor (BCR) stimuli, and attenuate the downstream activation of NF-kappaB or AP1-dependent transcription. Bioinformatics and pathway analysis of Ago PAR-CLIP datasets identified multiple EBV miRNA targets related to BCR signal transduction, including GRB2, SOS1, MALT1, RAC1, and INPP5D, which we validated in reporter assays. BCR signaling is critical for B cell activation, proliferation, and differentiation, and for EBV, is linked to reactivation. In functional assays, we demonstrate that EBV miR-BHRF1-2-5p contributes to the growth of latently infected B cells through GRB2 regulation. We further determined that activities of EBV miR-BHRF1-2-5p, EBV miR-BART2-5p, and a cellular miRNA, miR-17-5p, directly regulate virus reactivation triggered by BCR engagement. Our findings provide mechanistic insight into some of the key miRNA interactions impacting the proliferation of latently infected B cells and importantly, governing the latent to lytic switch.


Subject(s)
GRB2 Adaptor Protein/metabolism , Herpesvirus 4, Human/genetics , Receptors, Antigen, B-Cell/physiology , B-Lymphocytes/virology , Cell Line , Epstein-Barr Virus Infections/virology , GRB2 Adaptor Protein/physiology , Gene Expression Regulation, Viral/genetics , HEK293 Cells , Herpesvirus 4, Human/immunology , Humans , MicroRNAs/genetics , NF-kappa B/metabolism , Receptors, Antigen, B-Cell/genetics , Signal Transduction , Viral Proteins/metabolism , Virus Latency/genetics
3.
Oncogene ; 34(4): 474-84, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-24469046

ABSTRACT

Sprouty2 (SPRY2) is a potent negative regulator of receptor tyrosine kinase signaling, and is implicated as a tumor suppressor. SPRY2 inhibits FGF-RAS-ERK signaling by binding to growth factor receptor bound protein 2 (GRB2) during fibroblast growth factor receptor (FGFR) activation, disrupting the GRB2-SOS (son of sevenless) complex that transduces signals from FGFR to RAS. SPRY2 binding to GRB2 is modulated by phosphorylation but the key regulatory kinase(s) are not known. Prior studies identified the frequent presence of CK1 phosphorylation motifs on SPRY2. We therefore tested if CK1 has a role in SPRY2 phosphorylation and function. Loss of CK1 binding and inhibition of CK1 activity by two structurally distinct small molecules abrogated SPRY2 inhibition of FGF-ERK signaling, leading to decreased SPRY2 interaction with GRB2. Moreover, CK1 activity and binding are necessary for SPRY2 inhibition of FGF-stimulated neurite outgrowth in PC12 cells. Consistent with its proposed role as an inhibitor of FGF signaling, we find that CSNK1E transcript abundance negatively correlates with FGF1/FGF7 message in human gastric cancer samples. Modulation of CK1 activity may be therapeutically useful in the treatment of FGF/SPRY2-related diseases.


Subject(s)
Casein Kinase I/physiology , Fibroblast Growth Factors/physiology , Intracellular Signaling Peptides and Proteins/physiology , MAP Kinase Signaling System/physiology , Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , GRB2 Adaptor Protein/physiology , Humans , Neurites/physiology , Phosphorylation , Rats , Receptors, Fibroblast Growth Factor/physiology , Stomach Neoplasms/metabolism
4.
J Biol Chem ; 289(52): 36048-58, 2014 Dec 26.
Article in English | MEDLINE | ID: mdl-25381250

ABSTRACT

The non-receptor isoform of protein-tyrosine phosphatase ϵ (cyt-PTPe) supports adhesion of bone-resorbing osteoclasts by activating Src downstream of integrins. Loss of cyt-PTPe reduces Src activity in osteoclasts, reduces resorption of mineralized matrix both in vivo and in cell culture, and induces mild osteopetrosis in young female PTPe KO mice. Activation of Src by cyt-PTPe is dependent upon this phosphatase undergoing phosphorylation at its C-terminal Tyr-638 by partially active Src. To understand how cyt-PTPe activates Src, we screened 73 Src homology 2 (SH2) domains for binding to Tyr(P)-638 of cyt-PTPe. The SH2 domain of GRB2 bound Tyr(P)-638 of cyt-PTPe most prominently, whereas the Src SH2 domain did not bind at all, suggesting that GRB2 may link PTPe with downstream molecules. Further studies indicated that GRB2 is required for activation of Src by cyt-PTPe in osteoclast-like cells (OCLs) in culture. Overexpression of GRB2 in OCLs increased activating phosphorylation of Src at Tyr-416 and of cyt-PTPe at Tyr-638; opposite results were obtained when GRB2 expression was reduced by shRNA or by gene inactivation. Phosphorylation of cyt-PTPe at Tyr-683 and its association with GRB2 are integrin-driven processes in OCLs, and cyt-PTPe undergoes autodephosphorylation at Tyr-683, thus limiting Src activation by integrins. Reduced GRB2 expression also reduced the ability of bone marrow precursors to differentiate into OCLs and reduced the fraction of OCLs in which podosomal adhesion structures assume organization typical of active, resorbing cells. We conclude that GRB2 physically links cyt-PTPe with Src and enables cyt-PTPe to activate Src downstream of activated integrins in OCLs.


Subject(s)
GRB2 Adaptor Protein/physiology , Osteoclasts/enzymology , Receptor-Like Protein Tyrosine Phosphatases, Class 4/physiology , src-Family Kinases/metabolism , Amino Acid Sequence , Animals , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cell Differentiation , HEK293 Cells , Humans , Mice, 129 Strain , Mice, Knockout , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational
5.
Sci Signal ; 7(330): ra58, 2014 Jun 17.
Article in English | MEDLINE | ID: mdl-24939894

ABSTRACT

Lemur tyrosine kinase 3 (LMTK3) is associated with cell proliferation and endocrine resistance in breast cancer. We found that, in cultured breast cancer cell lines, LMTK3 promotes the development of a metastatic phenotype by inducing the expression of genes encoding integrin subunits. Invasive behavior in various breast cancer cell lines positively correlated with the abundance of LMTK3. Overexpression of LMTK3 in a breast cancer cell line with low endogenous LMTK3 abundance promoted actin cytoskeleton remodeling, focal adhesion formation, and adhesion to collagen and fibronectin in culture. Using SILAC (stable isotope labeling by amino acids in cell culture) proteomic analysis, we found that LMTK3 increased the abundance of integrin subunits α5 and ß1, encoded by ITGA5 and ITGB1. This effect depended on the CDC42 Rho family guanosine triphosphatase, which was in turn activated by the interaction between LMTK3 and growth factor receptor-bound protein 2 (GRB2), an adaptor protein that mediates receptor tyrosine kinase-induced activation of RAS and downstream signaling. Knockdown of GRB2 suppressed LMTK3-induced CDC42 activation, blocked ITGA5 and ITGB1 expression promoted by the transcription factor serum response factor (SRF), and reduced invasive activity. Furthermore, abundance of LMTK3 positively correlated with that of the integrin ß1 subunit in breast cancer patient's tumors. Our findings suggest a role for LMTK3 in promoting integrin activity during breast cancer progression and metastasis.


Subject(s)
Breast Neoplasms/pathology , GRB2 Adaptor Protein/physiology , Integrin beta1/physiology , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Female , Humans , Neoplasm Invasiveness
7.
Nat Struct Mol Biol ; 21(2): 180-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24440983

ABSTRACT

FGFR2-expressing human cancer cells with low concentrations of the adaptor protein Grb2 show high prevalence for metastatic outcome. In nonstimulated cells, the SH3 domain (and not the SH2 domains) of Plcγ1 directly competes for a binding site at the very C terminus of FGFR2 with the C-terminal SH3 domain of Grb2. Reduction of Grb2 concentration permits Plcγ1 access to the receptor. Recruitment of Plcγ1 in this way is sufficient to upregulate phospholipase activity. This results in elevated phosphatidylinositol 4,5-bisphosphate turnover and intracellular calcium levels, thus leading to increased cell motility and promotion of cell-invasive behavior in the absence of extracellular receptor stimulation. Therefore, metastatic outcome can be dictated by the constitutive competition between Grb2 and Plcγ1 for the phosphorylation-independent binding site on FGFR2.


Subject(s)
GRB2 Adaptor Protein/physiology , Phospholipase C gamma/physiology , Phospholipases/physiology , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Binding Sites , Binding, Competitive , Cell Line, Tumor , GRB2 Adaptor Protein/metabolism , HEK293 Cells , Humans , Models, Genetic , Neoplasm Invasiveness/genetics , Phospholipase C gamma/metabolism , Protein Structure, Tertiary
8.
Circ Res ; 114(3): 444-453, 2014 Jan 31.
Article in English | MEDLINE | ID: mdl-24265393

ABSTRACT

RATIONALE: Platelets are anuclear cell fragments derived from bone marrow megakaryocytes (MKs) that safeguard vascular integrity but may also cause pathological vessel occlusion. One major pathway of platelet activation is triggered by 2 receptors that signal through an (hem)immunoreceptor tyrosine-based activation motif (ITAM), the activating collagen receptor glycoprotein (GP) VI and the C-type lectin-like receptor 2 (CLEC-2). Growth factor receptor-bound protein 2 (Grb2) is a ubiquitously expressed adapter molecule involved in signaling processes of numerous receptors in different cell types, but its function in platelets and MKs is unknown. OBJECTIVE: We tested the hypothesis that Grb2 is a crucial adapter protein in (hem)immunoreceptor tyrosine-based activation motif signaling in platelets. METHODS AND RESULTS: Here, we show that genetic ablation of Grb2 in MKs and platelets did not interfere with MK differentiation or platelet production. However, Grb2-deficiency severely impaired glycoprotein VI-mediated platelet activation because of defective stabilization of the linker of activated T-cell (LAT) signalosome and activation of downstream signaling proteins that resulted in reduced adhesion, aggregation, and coagulant activity on collagen in vitro. Similarly, CLEC-2-mediated signaling was impaired in Grb2-deficient platelets, whereas the cells responded normally to stimulation of G protein-coupled receptors. In vivo, this selective (hem)immunoreceptor tyrosine-based activation motif signaling defect resulted in prolonged bleeding times but affected arterial thrombus formation only after concomitant treatment with acetylsalicylic acid, indicating that defective glycoprotein VI signaling in the absence of Grb2 can be compensated through thromboxane A2-induced G protein-coupled receptor signaling pathways. CONCLUSIONS: These results reveal an important contribution of Grb2 in (hem)immunoreceptor tyrosine-based activation motif signaling in platelets in hemostasis and thrombosis by stabilizing the LAT signalosome.


Subject(s)
Blood Platelets/metabolism , GRB2 Adaptor Protein/physiology , Immunoreceptor Tyrosine-Based Activation Motif/genetics , Signal Transduction/genetics , Amino Acid Motifs/genetics , Animals , Cells, Cultured , GRB2 Adaptor Protein/genetics , Hemostasis/genetics , Immunoreceptor Tyrosine-Based Inhibition Motif/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Activation/genetics , Thrombosis/genetics
9.
J Alzheimers Dis ; 38(4): 881-95, 2014.
Article in English | MEDLINE | ID: mdl-24100123

ABSTRACT

The ascertainment of elevated levels of amyloid-ß protein precursor intracellular domain (AICD) in Alzheimer's disease (AD) brains and the fact that it contributes to AD-like pathology has geared the search toward a new paradigm. While studying endogenous as well as overexpressed Grb2-AICD interaction in AD cell models, it was found that Grb2 co-localized to compartments along with AICD. We report now that these vesicles form in a clathrin and dynamin independent manner. Both types of vesicles mature into autophagosomes, merge with lysosomes, and relieve the cells of AICD overload. Inhibiting autophagosome formation results in vesicle accumulation. AICD-level is reduced in Grb2 excess condition in Cycloheximide Chase setup. Reduced caspase activity and apoptosis point toward the fact that the cytotoxic effect of AICD is alleviated by its sequestration in autolysosomes. Hence we state that the entrapping of AICD in Grb2 vesicles and its clearance via autophagosomes is a survival contrivance on the part of the cell. This study unravels, for the first time, the roles of Grb2 in autophagy and in handling toxic protein overload in an AD-like scenario.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Autophagy/physiology , GRB2 Adaptor Protein/physiology , Intracellular Fluid/metabolism , Neurons/metabolism , Amyloid beta-Protein Precursor/toxicity , Animals , Cell Line, Tumor , GRB2 Adaptor Protein/toxicity , Humans , Neurons/pathology
10.
J Biol Chem ; 288(41): 29934-42, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-24003223

ABSTRACT

Grb2-associated regulator of Erk/MAPK1 (GAREM) is an adaptor molecule in the EGF-mediated signaling pathway. GAREM is expressed ubiquitously in human organs and cultured cells. Two GAREM homologues are encoded by the human genome. Therefore, previously identified GAREM is named GAREM1. Here we characterized a new subtype of GAREM, GAREM2, that is specifically expressed in the mouse, rat, and human brain. Three GAREM2 tyrosines (Tyr-102, Tyr-429, and Tyr-551) are phosphorylated upon EGF stimulation and are necessary for binding to Grb2. Furthermore, GAREM2 and Shp2 regulate Erk activity in EGF-stimulated cells. These characteristics are similar to those of GAREM1. GAREM2 is expressed in some neuroblastoma cell lines and is also tyrosine-phosphorylated and bound to Grb2 after treatment with EGF. Eventually, GAREM2 regulates Erk activation in the presence of EGF or insulin like growth factor 1. GAREM2 also regulates insulin-like growth factor 1-induced neuronal differentiation of the SH-SY5Y neuroblastoma cell line. Although the structure and function of both GAREM subtypes are similar, GAREM1 is recruited into the nucleus and GAREM2 is not. Nuclear localization of GAREM1 might be controlled by a GAREM1-specific nuclear localization sequence and 14-3-3ε binding. The N-terminal 20 amino acids of GAREM1 make up its nuclear localization sequence that is also a 14-3-3ε binding site. The GAREM family is a new class of adaptor molecules with subtype-specific biological functions.


Subject(s)
Brain/metabolism , GRB2 Adaptor Protein/physiology , MAP Kinase Signaling System/physiology , Neurites/physiology , Amino Acid Sequence , Animals , COS Cells , Cell Line, Tumor , Cell Nucleus/metabolism , Chlorocebus aethiops , Epidermal Growth Factor/pharmacology , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , MAP Kinase Signaling System/drug effects , Mice , Microscopy, Fluorescence , Molecular Sequence Data , Neurites/metabolism , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neurons/cytology , Neurons/metabolism , Phosphorylation/drug effects , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , RNA Interference , Rats , Rats, Wistar , Sequence Homology, Amino Acid
11.
Chirality ; 25(10): 628-42, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23925889

ABSTRACT

Protein-protein interactions are crucial for signal transductions required for cell differentiation and proliferation. Their modulation is therefore key to the development of therapeutic alternatives, particularly in the context of cancer. According to literature data, the polyproline-rich nuclear receptor coactivators PNRC and PNRC2 interact with estrogen receptor (ERα) through their PxxP SH3-binding motifs. In a search to identify the molecular features governing this interaction, we explored using electronic circular dichroism (ECD) spectroscopy and molecular dynamics (MD) calculations, the capacity of a range of putative biologically active peptides derived from these proteins and containing this PxxP motif(s) to form polyproline II (PPII) domains. An additional more exhaustive structural study on a lead PPII peptide was also performed using 2D nuclear magnetic resonance (NMR) spectroscopy. With the exception of one of all the investigated peptides (PNRC-D), binding assays failed to detect any affinity for Grb2 SH3 domains, suggesting that PPII motifs issued from Grb2 antagonists have a binding mode distinct from those derived from Grb2 agonists. Instead, the peptides revealed a competitive binding ability against a synthetic peptide (ERα17p) with a putative PPII-cognate domain located within a coregulator recruitment region of ERα (AF-2 site). Our work, which constitutes the first structure-related interaction study concerning PNRC and PNRC2, supports not only the existence of PxxP-induced PPII sequences in these coregulators, but also confirms the presence of a PPII recognition site in the AF-2 of the steroid receptor ERα, a region important for transcription regulation.


Subject(s)
Estrogen Receptor alpha/chemistry , Nuclear Proteins , Nuclear Receptor Coactivators/chemistry , Peptides/chemistry , Proline/chemistry , Receptors, Cytoplasmic and Nuclear , Trans-Activators , Transcription Factors , src Homology Domains/physiology , Amino Acid Motifs/physiology , Amino Acid Sequence , Circular Dichroism , Estrogen Receptor alpha/physiology , GRB2 Adaptor Protein/chemistry , GRB2 Adaptor Protein/physiology , Humans , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Protein Binding , Protein Structure, Tertiary , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Sequence Alignment , Trans-Activators/genetics , Trans-Activators/physiology , Transcription Factors/genetics , Transcription Factors/physiology
12.
Circulation ; 127(21): 2097-106, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23625957

ABSTRACT

BACKGROUND: Several microRNAs (miRs) have been shown to regulate gene expression in the heart, and dysregulation of their expression has been linked to cardiac disease. miR-378 is strongly expressed in the mammalian heart but so far has been studied predominantly in cancer, in which it regulates cell survival and tumor growth. METHODS AND RESULTS: Here, we report tight control of cardiomyocyte hypertrophy through miR-378. In isolated primary cardiomyocytes, miR-378 was found to be both necessary and sufficient to repress cardiomyocyte hypertrophy. Bioinformatic prediction suggested that factors of the mitogen-activated protein kinase (MAPK) pathway are enriched among miR-378 targets. Using mRNA and protein expression analysis along with luciferase assays, we validated 4 key components of the MAPK pathway as targets of miR-378: MAPK1 itself, insulin-like growth factor receptor 1, growth factor receptor-bound protein 2, and kinase suppressor of ras 1. RNA interference with these targets prevented the prohypertrophic effect of antimiR-378, suggesting their functional relation with miR-378. Because miR-378 significantly decreases in cardiac disease, we sought to compensate for its loss through adeno-associated virus-mediated, cardiomyocyte-targeted expression of miR-378 in an in vivo model of cardiac hypertrophy (pressure overload by thoracic aortic constriction). Restoration of miR-378 levels significantly attenuated thoracic aortic constriction-induced cardiac hypertrophy and improved cardiac function. CONCLUSIONS: Our data identify miR-378 as a regulator of cardiomyocyte hypertrophy, which exerts its activity by suppressing the MAPK signaling pathway on several distinct levels. Restoration of disease-associated loss of miR-378 through cardiomyocyte-targeted adeno-associated virus-miR-378 may prove to be an effective therapeutic strategy in myocardial disease.


Subject(s)
Cardiomegaly/pathology , Cardiomegaly/physiopathology , MicroRNAs/physiology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/physiology , Signal Transduction/physiology , Adenoviridae/genetics , Animals , Cells, Cultured , Disease Models, Animal , Down-Regulation/physiology , GRB2 Adaptor Protein/antagonists & inhibitors , GRB2 Adaptor Protein/physiology , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/physiology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Protein Kinases/physiology , RNA Interference , Rats , Rats, Sprague-Dawley , Receptors, Somatomedin/antagonists & inhibitors , Receptors, Somatomedin/physiology
13.
J Cell Biol ; 200(4): 493-504, 2013 Feb 18.
Article in English | MEDLINE | ID: mdl-23420874

ABSTRACT

Constitutive receptor tyrosine kinase phosphorylation requires regulation of kinase and phosphatase activity to prevent aberrant signal transduction. A dynamic mechanism is described here in which the adaptor protein, growth factor receptor-bound protein 2 (Grb2), controls fibroblast growth factor receptor 2 (FGFR2) signaling by regulating receptor kinase and SH2 domain-containing protein tyrosine phosphatase 2 (Shp2) phosphatase activity in the absence of extracellular stimulation. FGFR2 cycles between its kinase-active, partially phosphorylated, nonsignaling state and its Shp2-dephosphorylated state. Concurrently, Shp2 cycles between its FGFR2-phosphorylated and dephosphorylated forms. Both reciprocal activities of FGFR2 and Shp2 were inhibited by binding of Grb2 to the receptor. Phosphorylation of Grb2 by FGFR2 abrogated its binding to the receptor, resulting in up-regulation of both FGFR2's kinase and Shp2's phosphatase activity. Dephosphorylation of Grb2 by Shp2 rescued the FGFR2-Grb2 complex. This cycling of enzymatic activity results in a homeostatic, signaling-incompetent state. Growth factor binding perturbs this background cycling, promoting increased FGFR2 phosphorylation and kinase activity, Grb2 dissociation, and downstream signaling. Grb2 therefore exerts constitutive control over the mutually dependent activities of FGFR2 and Shp2.


Subject(s)
GRB2 Adaptor Protein/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Receptor, Fibroblast Growth Factor, Type 2/metabolism , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , Gene Knockdown Techniques , HEK293 Cells , Humans , MAP Kinase Signaling System , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism
15.
Biochem Mol Biol Educ ; 40(5): 333-6, 2012.
Article in English | MEDLINE | ID: mdl-22987556

ABSTRACT

Terms to be familiar with before you start to solve the test: reciprocal translocation, proto-oncogene, gene expression, c-Abl, tyrosine protein kinases, transmembrane protein, G protein, Src protein, malignant transformation, transfection, immunoprecipitation, pre-immune serum, [γ-(32)P]ATP, cAMP, SDS-polyacrylamide gel electrophoresis, autoradiography, Western blot (immunoblot), co-immunoprecipitation, expression vector, cDNA, transient transfection, reporter gene, reporter plasmid, promoter, Ras protein, transformed foci, extracellular signal-regulated kinase.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Philadelphia Chromosome , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Problem-Based Learning/methods , Signal Transduction , Electrophoresis, Polyacrylamide Gel , Fusion Proteins, bcr-abl/physiology , GRB2 Adaptor Protein/physiology , Humans , Immunoprecipitation , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Mas
16.
Blood ; 120(13): 2669-78, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22806893

ABSTRACT

Intracellular mechanism(s) that contribute to promiscuous signaling via oncogenic KIT in systemic mastocytosis and acute myelogenous leukemia are poorly understood. We show that SHP2 phosphatase is essential for oncogenic KIT-induced growth and survival in vitro and myeloproliferative disease (MPD) in vivo. Genetic disruption of SHP2 or treatment of oncogene-bearing cells with a novel SHP2 inhibitor alone or in combination with the PI3K inhibitor corrects MPD by disrupting a protein complex involving p85α, SHP2, and Gab2. Importantly, a single tyrosine at position 719 in oncogenic KIT is sufficient to develop MPD by recruiting p85α, SHP2, and Gab2 complex to oncogenic KIT. Our results demonstrate that SHP2 phosphatase is a druggable target that cooperates with lipid kinases in inducing MPD.


Subject(s)
Cell Transformation, Neoplastic/pathology , GRB2 Adaptor Protein/physiology , Mutation/genetics , Myeloproliferative Disorders/etiology , Myeloproliferative Disorders/prevention & control , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Proto-Oncogene Proteins c-kit/genetics , Animals , Apoptosis , Blotting, Western , Bone Marrow Transplantation , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Immunoprecipitation , Integrases/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Myeloproliferative Disorders/mortality , Phosphorylation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction/drug effects , Survival Rate , Tyrosine/metabolism
17.
Biochim Biophys Acta ; 1813(10): 1836-44, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21745501

ABSTRACT

The Src-homology 3 (SH3) domain is one of the most frequent protein recognition modules (PRMs), being represented in signal transduction pathways and in several pathologies such as cancer and AIDS. Grb2 (growth factor receptor-bound protein 2) is an adaptor protein that contains two SH3 domains and is involved in receptor tyrosine kinase (RTK) signal transduction pathways. The HIV-1 transactivator factor Tat is required for viral replication and it has been shown to bind directly or indirectly to several host proteins, deregulating their functions. In this study, we show interaction between the cellular factor Grb2 and the HIV-1 trans-activating protein Tat. The binding is mediated by the proline-rich sequence of Tat and the SH3 domain of Grb2. As the adaptor protein Grb2 participates in a wide variety of signaling pathways, we characterized at least one of the possible downstream effects of the Tat/Grb2 interaction on the well-known IGF-1R/Raf/MAPK cascade. We show that the binding of Tat to Grb2 impairs activation of the Raf/MAPK pathway, while potentiating the PKA/Raf inhibitory pathway. The Tat/Grb2 interaction affects also viral function by inhibiting the Tat-mediated transactivation of HIV-1 LTR and viral replication in infected primary microglia.


Subject(s)
GRB2 Adaptor Protein/metabolism , HIV Infections/virology , HIV-1/physiology , Microglia/physiology , src Homology Domains , tat Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Sequence , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , GRB2 Adaptor Protein/chemistry , GRB2 Adaptor Protein/physiology , HIV Infections/metabolism , HIV Infections/pathology , HIV-1/metabolism , Host-Pathogen Interactions/physiology , Humans , Microglia/metabolism , Microglia/pathology , Microglia/virology , Molecular Sequence Data , Protein Binding , Signal Transduction , Virus Replication/physiology , tat Gene Products, Human Immunodeficiency Virus/physiology
18.
Am J Physiol Gastrointest Liver Physiol ; 301(2): G368-76, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21617115

ABSTRACT

Upon ligand binding, epidermal growth factor (EGF) receptor (R) autophosphorylates on COOH-terminal tyrosines, generating docking sites for signaling partners that stimulate proliferation, restitution, and chemotaxis. Specificity for individual EGFR tyrosines in cellular responses has been hypothesized but not well documented. Here we tested the requirement for particular tyrosines, and associated downstream pathways, in mouse colon epithelial cell chemotactic migration. We compared these requirements to those for the phenotypically distinct restitution (wound healing) migration. Wild-type, Y992/1173F, Y1045F, Y1068F, and Y1086F EGFR constructs were expressed in EGFR(-/-) cells; EGF-induced chemotaxis or restitution were determined by Boyden chamber or modified scratch wound assay, respectively. Pharmacological inhibitors of p38, phospholipase C (PLC), Src, MEK, JNK/SAPK, phosphatidylinositol 3-kinase (PI 3-kinase), and protein kinase C (PKC) were used to block EGF-stimulated signaling. Pathway activation was determined by immunoblot analysis. Unlike wild-type EGFR, Y992/1173F and Y1086F EGFR did not stimulate colon epithelial cell chemotaxis toward EGF; Y1045F and Y1068F EGFR partially stimulated chemotaxis. Only wild-type EGFR promoted colonocyte restitution. Inhibition of p38, PLC, and Src, or Grb2 knockdown, blocked chemotaxis; JNK, PI 3-kinase, and PKC inhibitors or c-Cbl knockdown blocked restitution but not chemotaxis. All four EGFR mutants stimulated downstream signaling in response to EGF, but Y992/1173F EGFR was partially defective in PLCγ activation whereas both Y1068F and Y1086F EGFR failed to activate Src. We conclude that specific EGFR tyrosines play key roles in determining cellular responses to ligand. Chemotaxis and restitution, which have different migration phenotypes and physiological consequences, have overlapping but not identical EGFR signaling requirements.


Subject(s)
Chemotaxis/physiology , Epidermal Growth Factor/physiology , Epithelial Cells/physiology , ErbB Receptors/metabolism , Signal Transduction/physiology , Tyrosine/physiology , Animals , Binding Sites/physiology , Cell Line , Chemotaxis/drug effects , Colon/metabolism , Epidermal Growth Factor/metabolism , Epithelial Cells/metabolism , ErbB Receptors/deficiency , ErbB Receptors/genetics , ErbB Receptors/physiology , GRB2 Adaptor Protein/deficiency , GRB2 Adaptor Protein/physiology , Mice , Mutation , Phospholipase C gamma/physiology , Phosphorylation , Proto-Oncogene Proteins c-cbl/deficiency , Proto-Oncogene Proteins c-cbl/physiology , RNA, Small Interfering , Transfection , Wound Healing/drug effects , Wound Healing/physiology , p38 Mitogen-Activated Protein Kinases/physiology , src Homology Domains/physiology , src-Family Kinases
19.
EMBO J ; 30(8): 1621-33, 2011 Apr 20.
Article in English | MEDLINE | ID: mdl-21427701

ABSTRACT

Grb2 is a ubiquitously expressed adaptor protein, which activates Ras and MAP kinases in growth factor receptor signalling, while in B-cell receptor (BCR) signalling this role is controversial. In B cell lines it was shown that Grb2 can inhibit BCR-induced Ca(2+) signalling. Nonetheless, the physiological role of Grb2 in primary B cells is still unknown. We generated a B-cell-specific Grb2-deficient mouse line, which had a severe reduction of mature follicular B cells in the periphery due to a differentiation block and decreased B-cell survival. Moreover, we found several changes in important signalling pathways: enhanced BCR-induced Ca(2+) signalling, alterations in mitogen-activated protein kinase activation patterns and strongly impaired Akt activation, the latter pointing towards a defect in PI3K signalling. Interestingly, B-cell-specific Grb2-deficient mice showed impaired IgG and B-cell memory responses, and impaired germinal centre formation. Thus, Grb2-dependent signalling pathways are crucial for lymphocyte differentiation processes, as well as for control of secondary humoral immune responses.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Calcium/metabolism , GRB2 Adaptor Protein/physiology , Immunologic Memory , Signal Transduction , Adoptive Transfer , Animals , B-Cell Activation Factor Receptor , B-Lymphocytes/immunology , Blotting, Western , Bone Marrow/metabolism , Cell Cycle , Cell Differentiation , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Immunization , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Phosphorylation , Proto-Oncogene Proteins c-akt
SELECTION OF CITATIONS
SEARCH DETAIL
...