Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
2.
Mol Cell Proteomics ; 19(1): 50-64, 2020 01.
Article in English | MEDLINE | ID: mdl-31678930

ABSTRACT

The RAS/mitogen-activated protein kinase (MAPK) signaling pathway regulates various biological functions, including cell survival, proliferation and migration. This pathway is frequently deregulated in cancer, including melanoma, which is the most aggressive form of skin cancer. RSK (p90 ribosomal S6 kinase) is a MAPK-activated protein kinase required for melanoma growth and proliferation, but relatively little is known about its function and the nature of its cellular partners. In this study, we used a proximity-based labeling approach to identify RSK proximity partners in cells. We identified many potential RSK-interacting proteins, including p120ctn (p120-catenin), which is an essential component of adherens junction (AJ). We found that RSK phosphorylates p120ctn on Ser320, which appears to be constitutively phosphorylated in melanoma cells. We also found that RSK inhibition increases melanoma cell-cell adhesion, suggesting that constitutive RAS/MAPK signaling negatively regulates AJ integrity. Together, our results indicate that RSK plays an important role in the regulation of melanoma cell-cell adhesion.


Subject(s)
Catenins/metabolism , Cell Adhesion/genetics , Melanoma/metabolism , Proteomics/methods , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Catenins/genetics , Cell Line, Tumor , HEK293 Cells , Humans , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , RNA Interference , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Signal Transduction/genetics , Delta Catenin
3.
Sci Transl Med ; 8(365): 365ra159, 2016 11 16.
Article in English | MEDLINE | ID: mdl-27856798

ABSTRACT

Microtubule-targeting agents (MTAs) are widely used anticancer agents, but toxicities such as neuropathy limit their clinical use. MTAs bind to and alter the stability of microtubules, causing cell death in mitosis. We describe DZ-2384, a preclinical compound that exhibits potent antitumor activity in models of multiple cancer types. It has an unusually high safety margin and lacks neurotoxicity in rats at effective plasma concentrations. DZ-2384 binds the vinca domain of tubulin in a distinct way, imparting structurally and functionally different effects on microtubule dynamics compared to other vinca-binding compounds. X-ray crystallography and electron microscopy studies demonstrate that DZ-2384 causes straightening of curved protofilaments, an effect proposed to favor polymerization of tubulin. Both DZ-2384 and the vinca alkaloid vinorelbine inhibit microtubule growth rate; however, DZ-2384 increases the rescue frequency and preserves the microtubule network in nonmitotic cells and in primary neurons. This differential modulation of tubulin results in a potent MTA therapeutic with enhanced safety.


Subject(s)
Antineoplastic Agents/pharmacology , Lactams, Macrocyclic/pharmacology , Microtubules/drug effects , Neurons/drug effects , Oxazoles/pharmacology , Vinca Alkaloids/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Crystallography, X-Ray , Dimerization , Genomics , Humans , Lactams, Macrocyclic/chemistry , Mice , Microscopy, Electron , Mitosis , Neoplasm Transplantation , Oxazoles/chemistry , Tubulin/chemistry , Vinblastine/analogs & derivatives , Vinblastine/chemistry , Vinblastine/pharmacology , Vinca Alkaloids/chemistry , Vinorelbine
4.
Mutagenesis ; 30(2): 169-76, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25688110

ABSTRACT

The mechanistic/mammalian target of rapamycin (mTOR) is a conserved protein kinase that controls several anabolic processes required for cell growth and proliferation. As such, mTOR has been implicated in an increasing number of pathological conditions, including cancer, obesity, type 2 diabetes and neurodegeneration. As part of the mTOR complex 1 (mTORC1), mTOR regulates cell growth by promoting the biosynthesis of proteins, lipids and nucleic acids. Several mTORC1 substrates have been shown to regulate protein synthesis, including the eukaryotic initiation factor 4E (eIF4E)-binding proteins (4E-BPs) and the ribosomal S6 kinases (S6Ks) 1 and 2. In this work, we focus on the signalling pathways that lie both upstream and downstream of mTORC1, as well as their relevance to human pathologies. We further discuss pharmacological approaches that target mTOR and their applications for the treatment of cancer.


Subject(s)
Multiprotein Complexes/metabolism , Neoplasms/enzymology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/physiology , Antineoplastic Agents/therapeutic use , Cell Growth Processes/drug effects , Cell Proliferation/drug effects , Mechanistic Target of Rapamycin Complex 1 , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology
5.
Cell ; 158(2): 368-382, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25036633

ABSTRACT

Adenomatous polyposis coli (APC) is a microtubule plus-end scaffolding protein important in biology and disease. APC is implicated in RNA localization, although the mechanisms and functional significance remain unclear. We show APC is an RNA-binding protein and identify an RNA interactome by HITS-CLIP. Targets were highly enriched for APC-related functions, including microtubule organization, cell motility, cancer, and neurologic disease. Among the targets is ß2B-tubulin, known to be required in human neuron and axon migration. We show ß2B-tubulin is synthesized in axons and localizes preferentially to dynamic microtubules in the growth cone periphery. APC binds the ß2B-tubulin 3' UTR; experiments interfering with this interaction reduced ß2B-tubulin mRNA axonal localization and expression, depleted dynamic microtubules and the growth cone periphery, and impaired neuron migration. These results identify APC as a platform binding functionally related protein and RNA networks, and suggest a self-organizing model for the microtubule to localize synthesis of its own subunits.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Microtubules/metabolism , Neurogenesis , RNA-Binding Proteins/metabolism , Animals , Axons/metabolism , Base Sequence , Brain/cytology , Brain/metabolism , Cell Line , Cell Movement , Ganglia, Spinal/cytology , Genome-Wide Association Study , Growth Cones/metabolism , Mice , Molecular Sequence Data , Neurons/metabolism , Protein Interaction Maps , RNA, Messenger/metabolism , Rats , Sequence Alignment , Tubulin/metabolism
6.
Proc Natl Acad Sci U S A ; 111(29): E2918-27, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-25002506

ABSTRACT

The Ras/MAPK signaling cascade regulates various biological functions, including cell growth and proliferation. As such, this pathway is frequently deregulated in several types of cancer, including most cases of melanoma. RSK (p90 ribosomal S6 kinase) is a MAPK-activated protein kinase required for melanoma growth and proliferation, but relatively little is known about its exact function and the nature of its substrates. Herein, we used a quantitative phosphoproteomics approach to define the signaling networks regulated by RSK in melanoma. To more accurately predict direct phosphorylation substrates, we defined the RSK consensus phosphorylation motif and found significant overlap with the binding consensus of 14-3-3 proteins. We thus characterized the phospho-dependent 14-3-3 interactome in melanoma cells and found that a large proportion of 14-3-3 binding proteins are also potential RSK substrates. Our results show that RSK phosphorylates the tumor suppressor PDCD4 (programmed cell death protein 4) on two serine residues (Ser76 and Ser457) that regulate its subcellular localization and interaction with 14-3-3 proteins. We found that 14-3-3 binding promotes PDCD4 degradation, suggesting an important role for RSK in the inactivation of PDCD4 in melanoma. In addition to this tumor suppressor, our results suggest the involvement of RSK in a vast array of unexplored biological functions with relevance in oncogenesis.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Phosphoproteins/metabolism , Proteomics/methods , RNA-Binding Proteins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Tumor Suppressor Proteins/metabolism , 14-3-3 Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Cell Line , Cell Nucleus/metabolism , Consensus Sequence , Humans , Melanoma/metabolism , Melanoma/pathology , Models, Biological , Molecular Sequence Data , Peptide Library , Phosphorylation , Phosphoserine/metabolism , Protein Binding , Protein Transport , Proteolysis , Proteome/metabolism , Substrate Specificity
7.
Genes Dev ; 28(4): 357-71, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24532714

ABSTRACT

The mammalian target of rapamycin (mTOR) promotes cell growth and proliferation by promoting mRNA translation and increasing the protein synthetic capacity of the cell. Although mTOR globally promotes translation by regulating the mRNA 5' cap-binding protein eIF4E (eukaryotic initiation factor 4E), it also preferentially regulates the translation of certain classes of mRNA via unclear mechanisms. To help fill this gap in knowledge, we performed a quantitative proteomic screen to identify proteins that associate with the mRNA 5' cap in an mTOR-dependent manner. Using this approach, we identified many potential regulatory factors, including the putative RNA-binding protein LARP1 (La-related protein 1). Our results indicate that LARP1 associates with actively translating ribosomes via PABP and that LARP1 stimulates the translation of mRNAs containing a 5' terminal oligopyrimidine (TOP) motif, encoding for components of the translational machinery. We found that LARP1 associates with the mTOR complex 1 (mTORC1) and is required for global protein synthesis as well as cell growth and proliferation. Together, these data reveal important molecular mechanisms involved in TOP mRNA translation and implicate LARP1 as an important regulator of cell growth and proliferation.


Subject(s)
Autoantigens/metabolism , Gene Expression Regulation , Proteomics , Pyrimidines/metabolism , RNA, Messenger/genetics , Ribonucleoproteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autoantigens/genetics , Cell Line , Cell Line, Tumor , Cells, Cultured , HEK293 Cells , Humans , Mice , RNA Cap-Binding Proteins/metabolism , Ribonucleoproteins/genetics , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , SS-B Antigen
8.
Cytoskeleton (Hoboken) ; 70(11): 741-54, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24136886

ABSTRACT

The cellular actin cortex is the cytoskeletal structure primarily responsible for the control of animal cell shape and as such plays a central role in cell division, migration, and tissue morphogenesis. Due to the lack of experimental systems where the cortex can be investigated independently from other organelles, little is known about its composition, assembly, and homeostasis. Here, we describe novel tools to resolve the composition and regulation of the cortex. We report and validate a protocol for cortex purification based on the separation of cellular blebs. Mass spectrometry analysis of purified cortices provides a first extensive list of cortical components. To assess the function of identified proteins, we design an automated imaging assay for precise quantification of cortical actomyosin assembly dynamics. We show subtle changes in cortex assembly dynamics upon depletion of the identified cortical component profilin. Our widely applicable integrated method paves the way for systems-level investigations of the actomyosin cortex and its regulation during morphogenesis.


Subject(s)
Actins/metabolism , Cellular Structures/metabolism , Homeostasis , Imaging, Three-Dimensional/methods , Proteomics/methods , Actomyosin/metabolism , Cellular Structures/ultrastructure , HeLa Cells , Humans , Profilins/metabolism
9.
J Med Genet ; 50(11): 740-4, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23687350

ABSTRACT

BACKGROUND: Mutations in TSC1 or TSC2 cause the tuberous sclerosis complex (TSC), a disorder characterised by the development of hamartomas or benign tumours in various organs as well as the variable presence of epilepsy, intellectual disability (ID) and autism. TSC1, TSC2 and the recently described protein TBC1D7 form a complex that inhibits mTORC1 signalling and limits cell growth. Although it has been proposed that mutations in TBC1D7 might also cause TSC, loss of its function has not yet been documented in humans. METHODS AND RESULTS: We used homozygosity mapping and exome sequencing to study a consanguineous family with ID and megalencephaly but without any specific features of TSC. We identified only one rare coding variant, c.538delT:p.Y180fsX1 in TBC1D7, in the regions of homozygosity shared by the affected siblings. We show that this mutation abolishes TBC1D7 expression and is associated with increased mTORC1 signalling in cells of the affected individuals. CONCLUSIONS: Our study suggests that disruption of TBC1D7 causes ID but without the other typical features found in TSC. Although megalencephaly is not commonly observed in TSC, it has been associated with mTORC1 activation. Our observation thus reinforces the relationship between this pathway and the development of megalencephaly.


Subject(s)
Carrier Proteins/genetics , Intellectual Disability/genetics , Megalencephaly/genetics , Tuberous Sclerosis/genetics , Child , Child, Preschool , Female , Humans , Intracellular Signaling Peptides and Proteins , Male , Mutation , Pedigree
10.
Mol Cell Biol ; 33(8): 1657-70, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23401857

ABSTRACT

The scaffolding adapter protein Gab2 (Grb2-associated binder) participates in the signaling response evoked by various growth factors and cytokines. Gab2 is overexpressed in several human malignancies, including breast cancer, and was shown to promote mammary epithelial cell migration. The role of Gab2 in the activation of different signaling pathways is well documented, but less is known regarding the feedback mechanisms responsible for its inactivation. We now demonstrate that activation of the Ras/mitogen-activated protein kinase (MAPK) pathway promotes Gab2 phosphorylation on basic consensus motifs. More specifically, we show that RSK (p90 ribosomal S6 kinase) phosphorylates Gab2 on three conserved residues, both in vivo and in vitro. Mutation of these phosphorylation sites does not alter Gab2 binding to Grb2, but instead, we show that Gab2 phosphorylation inhibits the recruitment of the tyrosine phosphatase Shp2 in response to growth factors. Expression of an unphosphorylatable Gab2 mutant in mammary epithelial cells promotes an invasion-like phenotype and increases cell motility. Taken together, these results suggest that RSK is part of a negative-feedback loop that restricts Gab2-dependent epithelial cell motility. On the basis of the widespread role of Gab2 in receptor signaling, these findings also suggest that RSK plays a regulatory function in diverse receptor systems.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Movement , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Animals , Benzamides/pharmacology , Breast Neoplasms/metabolism , Cell Line , Female , GRB2 Adaptor Protein/metabolism , HEK293 Cells , Humans , Mice , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mutation , Phosphorylation , RNA Interference , RNA, Small Interfering , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Signal Transduction
11.
Mol Cell Biol ; 32(22): 4572-84, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22966201

ABSTRACT

Processing bodies (PBs, or P bodies) are cytoplasmic granules involved in mRNA storage and degradation that participate in the regulation of gene expression. PBs concentrate nontranslated mRNAs and several factors involved in mRNA decay and translational repression, including the eukaryotic translation initiation factor 4E-transporter (4E-T). 4E-T is required for PB assembly, but little is known about the molecular mechanisms that regulate its function. Here, we demonstrate that oxidative stress promotes multisite 4E-T phosphorylation. We show that the c-Jun N-terminal kinase (JNK) is targeted to PBs in response to oxidative stress and promotes the phosphorylation of 4E-T. Quantitative mass spectrometry analysis reveals that JNK phosphorylates 4E-T on six proline-directed sites that are required for the formation of the 4E-T complex upon stress. We have developed an image-based computational method to quantify the size, number, and density of PBs in cells, and we find that while 4E-T is required for steady-state PB assembly, its phosphorylation facilitates the formation of larger PBs upon oxidative stress. Using polysomal mRNA profiling, we assessed global and specific mRNA translation but did not find that 4E-T phosphorylation impacts translational control. Collectively, these data support a model whereby PB assembly is regulated by a two-step mechanism involving a 4E-T-dependent assembly stage in unstressed cells and a 4E-T phosphorylation-dependent aggregation stage in response to stress stimuli.


Subject(s)
Cytoplasmic Granules/metabolism , Gene Expression Regulation , JNK Mitogen-Activated Protein Kinases/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , RNA, Messenger/metabolism , Cell Line , Cytoplasmic Granules/genetics , Humans , Image Processing, Computer-Assisted , JNK Mitogen-Activated Protein Kinases/genetics , Mass Spectrometry , Molecular Imaging , Nucleocytoplasmic Transport Proteins/genetics , Oxidative Stress , Phosphorylation , Plasmids , Proline/metabolism , Protein Biosynthesis , Protein Transport , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Signal Transduction/genetics , Transfection
12.
Cell ; 141(4): 632-44, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20434207

ABSTRACT

Extracellular signals regulate protein translation in many cell functions. A key advantage of control at the translational level is the opportunity to regulate protein synthesis within specific cellular subregions. However, little is known about mechanisms that may link extracellular cues to translation with spatial precision. Here, we show that a transmembrane receptor, DCC, forms a binding complex containing multiple translation components, including eukaryotic initiation factors, ribosomal large and small subunits, and monosomes. In neuronal axons and dendrites DCC colocalizes in particles with translation machinery, and newly synthesized protein. The extracellular ligand netrin promoted DCC-mediated translation and disassociation of translation components. The functional and physical association of a cell surface receptor with the translation machinery leads to a generalizable model for localization and extracellular regulation of protein synthesis, based on a transmembrane translation regulation complex.


Subject(s)
Neurons/metabolism , Protein Biosynthesis , Receptors, Cell Surface/metabolism , Ribosomal Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Motifs , Animals , Axons/metabolism , Cells, Cultured , Chick Embryo , Dendrites/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Humans , Mice , Nerve Growth Factors/metabolism , Netrin-1 , Protein Structure, Tertiary , Rats , Receptors, Cell Surface/chemistry , Ribosomes/metabolism , Tumor Suppressor Proteins/chemistry
13.
Biol Cell ; 99(2): 67-86, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17222083

ABSTRACT

The Rho GTPases are implicated in almost every fundamental cellular process. They act as molecular switches that cycle between an active GTP-bound and an inactive GDP-bound state. Their slow intrinsic GTPase activity is greatly enhanced by RhoGAPs (Rho GTPase-activating proteins), thus causing their inactivation. To date, more than 70 RhoGAPs have been identified in eukaryotes, ranging from yeast to human, and based on sequence homology of their RhoGAP domain, we have grouped them into subfamilies. In the present Review, we discuss their regulation, biological functions and implication in human diseases.


Subject(s)
GTPase-Activating Proteins/metabolism , Animals , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/genetics , Gene Expression Regulation , Humans , Phylogeny , Protein Structure, Tertiary
14.
J Biol Chem ; 282(6): 3624-31, 2007 Feb 09.
Article in English | MEDLINE | ID: mdl-17158447

ABSTRACT

Rho GTPases regulate a multitude of cellular processes from cytoskeletal reorganization to gene transcription and are negatively regulated by GTPase-activating proteins (GAPs). Cdc42 GTPase-activating protein (CdGAP) is a ubiquitously expressed GAP for Rac1 and Cdc42. In this study, we set out to identify CdGAP-binding partners and, using a yeast two-hybrid approach, glycogen synthase kinase 3alpha (GSK-3alpha) was identified as a partner for CdGAP. GSK-3 exists in two isoforms, alpha and beta, and is involved in regulating many cellular functions from insulin response to tumorigenesis. We show that GSK-3alpha and -beta interact with CdGAP in mammalian cells. We also demonstrate that GSK-3 phosphorylates CdGAP both in vitro and in vivo on Thr-776, which we have previously shown to be an ERK 1/2 phosphorylation site involved in CdGAP regulation. We report that the mRNA and protein levels of CdGAP are increased upon serum stimulation and that GSK-3 activity is necessary for the up-regulation of the protein levels of CdGAP but not for the increase in mRNA. We conclude that GSK-3 is an important regulator of CdGAP and that regulation of CdGAP protein levels by serum presents a novel mechanism for cells to control Cdc42/Rac1 GTPase signaling pathways.


Subject(s)
GTPase-Activating Proteins/metabolism , Glycogen Synthase Kinase 3/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphoproteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Line, Tumor , Consensus Sequence , Glycogen Synthase Kinase 3 beta , Humans , Mice , Molecular Sequence Data , NIH 3T3 Cells , Phosphorylation , Proline/metabolism , cdc42 GTP-Binding Protein/metabolism
15.
Biol Cell ; 98(8): 445-56, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16519628

ABSTRACT

BACKGROUND INFORMATION: Rho GTPases regulate a wide range of cellular functions affecting both cell proliferation and cytoskeletal dynamics. They cycle between inactive GDP- and active GTP-bound states. This cycle is tightly regulated by GEFs (guanine nucleotide-exchange factors) and GAPs (GTPase-activating proteins). Mouse CdGAP (mCdc42 GTPase-activating protein) has been previously identified and characterized as a specific GAP for Rac1 and Cdc42, but not for RhoA. It consists of an N-terminal RhoGAP domain and a C-terminal proline-rich region. In addition, CdGAP-related genes are present in both vertebrates and invertebrates. We have recently reported that two predominant isoforms of CdGAP (250 and 90 kDa) exist in specific mouse tissues. RESULTS: In the present study, we have identified and characterized human CdGAP (KIAA1204) which shares 76% sequence identity to the long isoform of mCdGAP (mCdGAP-l). Similar to mCdGAP, it is active in vitro and in vivo on both Cdc42 and Rac1, but not RhoA, and is phosphorylated in vivo on serine and threonine residues. In contrast with mCdGAP-l, human CdGAP interacts with ERK1/2 (extracellular-signal-regulated kinase 1/2) through a region that does not involve a DEF (docking site for ERK Phe-Xaa-Phe-Pro) domain. Also, the tissue distribution of CdGAP proteins appears to be different between human and mouse species. Interestingly, we found that CdGAP proteins cause membrane blebbing in COS-7 cells. CONCLUSIONS: Our results suggest that CdGAP properties are well conserved between human and mouse species, and that CdGAP may play an unexpected role in apoptosis.


Subject(s)
GTPase-Activating Proteins/metabolism , Phosphoproteins/metabolism , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Extracellular Signal-Regulated MAP Kinases/metabolism , Fetus/metabolism , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/physiology , Gene Expression Regulation, Developmental/genetics , Humans , Mice , Molecular Sequence Data , Phosphoproteins/genetics , Phosphoproteins/physiology , Phosphorylation , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Serine/metabolism , Threonine/metabolism , Transfection , cdc42 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/genetics
16.
Mol Cell Biol ; 25(15): 6314-29, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16024771

ABSTRACT

Rho GTPases regulate multiple cellular processes affecting both cell proliferation and cytoskeletal dynamics. Their cycling between inactive GDP- and active GTP-bound states is tightly regulated by guanine nucleotide exchange factors and GTPase-activating proteins (GAPs). We have previously identified CdGAP (for Cdc42 GTPase-activating protein) as a specific GAP for Rac1 and Cdc42. CdGAP consists of an N-terminal RhoGAP domain and a C-terminal proline-rich region. In addition, CdGAP is a member of the impressively large number of mammalian RhoGAP proteins that is well conserved among both vertebrates and invertebrates. In mice, we find two predominant isoforms of CdGAP differentially expressed in specific tissues. We report here that CdGAP is highly phosphorylated in vivo on serine and threonine residues. We find that CdGAP is phosphorylated downstream of the MEK-extracellular signal-regulated kinase (ERK) pathway in response to serum or platelet-derived growth factor stimulation. Furthermore, CdGAP interacts with and is phosphorylated by ERK-1 and RSK-1 in vitro. A putative DEF (docking for ERK FXFP) domain located in the proline-rich region of CdGAP is required for efficient binding and phosphorylation by ERK1/2. We identify Thr776 as an in vivo target site of ERK1/2 and as an important regulatory site of CdGAP activity. Together, these data suggest that CdGAP is a novel substrate of ERK1/2 and mediates cross talk between the Ras/mitogen-activated protein kinase pathway and regulation of Rac1 activity.


Subject(s)
GTPase-Activating Proteins/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Signal Transduction/physiology , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Fibroblasts/metabolism , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/physiology , Male , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase 1/metabolism , Molecular Sequence Data , Mutation , Phosphorylation , Proline/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , Protein Structure, Tertiary , rac1 GTP-Binding Protein/metabolism , ras Proteins/metabolism
17.
J Cell Biol ; 167(4): 687-98, 2004 Nov 22.
Article in English | MEDLINE | ID: mdl-15557120

ABSTRACT

Netrin-1 acts as a chemoattractant molecule to guide commissural neurons (CN) toward the floor plate by interacting with the receptor deleted in colorectal cancer (DCC). The molecular mechanisms underlying Netrin-1-DCC signaling are still poorly characterized. Here, we show that DCC is phosphorylated in vivo on tyrosine residues in response to Netrin-1 stimulation of CN and that the Src family kinase inhibitors PP2 and SU6656 block both Netrin-1-dependent phosphorylation of DCC and axon outgrowth. PP2 also blocks the reorientation of Xenopus laevis retinal ganglion cells that occurs in response to Netrin-1, which suggests an essential role of the Src kinases in Netrin-1-dependent orientation. Fyn, but not Src, is able to phosphorylate the intracellular domain of DCC in vitro, and we demonstrate that Y1418 is crucial for DCC axon outgrowth function. Both DCC phosphorylation and Netrin-1-induced axon outgrowth are impaired in Fyn(-/-) CN and spinal cord explants. We propose that DCC is regulated by tyrosine phosphorylation and that Fyn is essential for the response of axons to Netrin-1.


Subject(s)
Cell Adhesion Molecules/metabolism , Growth Cones/metabolism , Nerve Growth Factors/metabolism , Proto-Oncogene Proteins/metabolism , Retina/embryology , Spinal Cord/embryology , Tumor Suppressor Proteins/metabolism , src-Family Kinases/metabolism , Animals , Cell Differentiation/physiology , Cells, Cultured , DCC Receptor , Enzyme Inhibitors/pharmacology , Female , Growth Cones/drug effects , Growth Cones/ultrastructure , Male , Mice , Mice, Knockout , Nerve Growth Factors/pharmacology , Netrin-1 , Phosphorylation/drug effects , Proto-Oncogene Proteins c-fyn , Rats , Receptors, Cell Surface , Retina/cytology , Retina/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Spinal Cord/cytology , Spinal Cord/metabolism , Tyrosine/metabolism , Xenopus Proteins , Xenopus laevis , src-Family Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...