Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
J Biol Chem ; 289(5): 2658-74, 2014 Jan 31.
Article in English | MEDLINE | ID: mdl-24337580

ABSTRACT

Activation of mammalian target of rapamycin complex 1 (mTORC1) by amino acids is mediated in part by the Rag GTPases, which bind the raptor subunit of mTORC1 in an amino acid-stimulated manner and promote mTORC1 interaction with Rheb-GTP, the immediate activator. Here we examine whether the ability of amino acids to regulate mTORC1 binding to Rag and mTORC1 activation is due to the regulation of Rag guanyl nucleotide charging. Rag heterodimers in vitro exhibit a very rapid, spontaneous exchange of guanyl nucleotides and an inability to hydrolyze GTP. Mutation of the Rag P-loop corresponding to Ras(Ser-17) abolishes guanyl nucleotide binding. Such a mutation in RagA or RagB inhibits, whereas in RagC or RagD it enhances, Rag heterodimer binding to mTORC1. The binding of wild-type and mutant Rag heterodimers to mTORC1 in vitro parallels that seen with transient expression, but binding to mTORC1 in vitro is entirely independent of Rag guanyl nucleotide charging. HeLa cells stably overexpressing wild-type or P-loop mutant RagC exhibit unaltered amino acid regulation of mTORC1. Despite amino acid-independent raptor binding to Rag, mTORC1 is inhibited by amino acid withdrawal as in parental cells. Rag heterodimers extracted from (32)P-labeled whole cells, or just from the pool associated with the lysosomal membrane, exhibit constitutive [(32)P]GTP charging that is unaltered by amino acid withdrawal. Thus, amino acids promote mTORC1 activation without altering Rag GTP charging. Raptor binding to Rag, although necessary, is not sufficient for mTORC1 activation. Additional amino acid-dependent steps couple Rag-mTORC1 to Rheb-GTP.


Subject(s)
Amino Acids/metabolism , GTP Phosphohydrolases/metabolism , Monomeric GTP-Binding Proteins/metabolism , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Dimerization , Enzyme Activation/physiology , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/genetics , HEK293 Cells , HeLa Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Monomeric GTP-Binding Proteins/chemistry , Monomeric GTP-Binding Proteins/genetics , Protein Binding/physiology , Regulatory-Associated Protein of mTOR , Signal Transduction/physiology , Tritium , mTOR Associated Protein, LST8 Homolog
2.
J Neurochem ; 127(1): 66-77, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23841933

ABSTRACT

Growth factors and nutrients, such as amino acids and glucose, regulate mammalian target of rapamycin complex 1 (mTORC1) signaling and subsequent translational control in a coordinated manner. Brain-derived neurotrophic factor (BDNF), the most prominent neurotrophic factor in the brain, activates mTORC1 and induces phosphorylation of its target, p70S6 kinase (p70S6K), at Thr389 in neurons. BDNF also increases mammalian target of rapamycin-dependent novel protein synthesis in neurons. Here, we report that BDNF-induced p70S6K activation is dependent on glucose, but not amino acids, sufficiency in cultured cortical neurons. AMP-activated protein kinase (AMPK) is the molecular background to this specific nutrient dependency. Activation of AMPK, which is induced by glucose deprivation, treatment with pharmacological agents such as 2-deoxy-D-glucose, metformin, and 5-aminoimidazole-4-carboxamide ribonucleoside or forced expression of a constitutively active AMPKα subunit, counteracts BDNF-induced phosphorylation of p70S6K and enhanced protein synthesis in cortical neurons. These results indicate that AMPK inhibits the effects of BDNF on mTORC1-mediated translation in neurons.


Subject(s)
AMP-Activated Protein Kinases/physiology , Brain-Derived Neurotrophic Factor/pharmacology , Multiprotein Complexes/physiology , Neurons/drug effects , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/physiology , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Deoxyglucose/pharmacology , Electrophoresis, Polyacrylamide Gel , Electroporation , Fibroblasts/metabolism , Glucose/deficiency , Glucose/physiology , Hypoglycemic Agents/pharmacology , Immunohistochemistry , Immunoprecipitation , Mechanistic Target of Rapamycin Complex 1 , Metformin/pharmacology , Methionine/metabolism , Oncogene Protein v-akt/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
3.
J Biomed Sci ; 20: 24, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23594158

ABSTRACT

BACKGROUND: mTOR is a genetically conserved serine/threonine protein kinase, which controls cell growth, proliferation, and survival. A multifunctional protein CAD, catalyzing the initial three steps in de novo pyrimidine synthesis, is regulated by the phosphorylation reaction with different protein kinases, but the relationship with mTOR protein kinase has not been known. RESULTS: CAD was recovered as a binding protein with mLST8, a component of the mTOR complexes, from HEK293 cells transfected with the FLAG-mLST8 vector. Association of these two proteins was confirmed by the co-immuoprecipitaiton followed by immunoblot analysis of transfected myc-CAD and FLAG-mLST8 as well as that of the endogenous proteins in the cells. Analysis using mutant constructs suggested that CAD has more than one region for the binding with mLST8, and that mLST8 recognizes CAD and mTOR in distinct ways. The CAD enzymatic activity decreased in the cells depleted of amino acids and serum, in which the mTOR activity is suppressed. CONCLUSION: The results obtained indicate that mLST8 bridges between CAD and mTOR, and plays a role in the signaling mechanism where CAD is regulated in the mTOR pathway through the association with mLST8.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Aspartate Carbamoyltransferase/metabolism , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/metabolism , Dihydroorotase/metabolism , Gene Expression Regulation, Enzymologic , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Amino Acids/metabolism , Aspartate Carbamoyltransferase/genetics , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/genetics , Dihydroorotase/genetics , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Phosphorylation , Protein Binding , Pyrimidines/biosynthesis , Pyrimidines/metabolism , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics , mTOR Associated Protein, LST8 Homolog
4.
J Biol Chem ; 286(44): 38043-38053, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-21914810

ABSTRACT

Insulin activation of mTOR complex 1 is accompanied by enhanced binding of substrates. We examined the mechanism and contribution of this enhancement to insulin activation of mTORC1 signaling in 293E and HeLa cells. In 293E, insulin increased the amount of mTORC1 retrieved by the transiently expressed nonphosphorylatable 4E-BP[5A] to an extent that varied inversely with the amount of PRAS40 bound to mTORC1. RNAi depletion of PRAS40 enhanced 4E-BP[5A] binding to ∼70% the extent of maximal insulin, and PRAS40 RNAi and insulin together did not increase 4E-BP[5A] binding beyond insulin alone, suggesting that removal of PRAS40 from mTORC1 is the predominant mechanism of an insulin-induced increase in substrate access. As regards the role of increased substrate access in mTORC1 signaling, RNAi depletion of PRAS40, although increasing 4E-BP[5A] binding, did not stimulate phosphorylation of endogenous mTORC1 substrates S6K1(Thr(389)) or 4E-BP (Thr(37)/Thr(46)), the latter already ∼70% of maximal in amino acid replete, serum-deprived 293E cells. In HeLa cells, insulin and PRAS40 RNAi also both enhanced the binding of 4E-BP[5A] to raptor but only insulin stimulated S6K1 and 4E-BP phosphorylation. Furthermore, Rheb overexpression in 293E activated mTORC1 signaling completely without causing PRAS40 release. In the presence of Rheb and insulin, PRAS40 release is abolished by Akt inhibition without diminishing mTORC1 signaling. In conclusion, dissociation of PRAS40 from mTORC1 and enhanced mTORC1 substrate binding results from Akt and mTORC1 activation and makes little or no contribution to mTORC1 signaling, which rather is determined by Rheb activation of mTOR catalytic activity, through mechanisms that remain to be fully elucidated.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Gene Expression Regulation , Phosphoproteins/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Glutathione Transferase/metabolism , HeLa Cells , Humans , Insulin/metabolism , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Phosphorylation , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
5.
J Biol Chem ; 285(26): 20109-16, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20427287

ABSTRACT

Mammalian target of rapamycin (mTOR) is a member of the phosphatidylinositol 3-kinase-related kinase (PIKK) family and is a major regulator of translation, cell growth, and autophagy. mTOR exists in two distinct complexes, mTORC1 and mTORC2, that differ in their subunit composition. In this study, we identified KIAA0406 as a novel mTOR-interacting protein. Because it has sequence homology with Schizosaccharomyces pombe Tti1, we named it mammalian Tti1. Tti1 constitutively interacts with mTOR in both mTORC1 and mTORC2. Knockdown of Tti1 suppresses phosphorylation of both mTORC1 substrates (S6K1 and 4E-BP1) and an mTORC2 substrate (Akt) and also induces autophagy. S. pombe Tti1 binds to Tel2, a protein whose mammalian homolog was recently reported to regulate the stability of PIKKs. We confirmed that Tti1 binds to Tel2 also in mammalian cells, and Tti1 interacts with and stabilizes all six members of the PIKK family of proteins (mTOR, ATM, ATR, DNA-PKcs, SMG-1, and TRRAP). Furthermore, using immunoprecipitation and size-exclusion chromatography analyses, we found that knockdown of either Tti1 or Tel2 causes disassembly of mTORC1 and mTORC2. These results indicate that Tti1 and Tel2 are important not only for mTOR stability but also for assembly of the mTOR complexes to maintain their activities.


Subject(s)
Carrier Proteins/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Transcription Factors/metabolism , Carrier Proteins/genetics , Cell Line , Chromatography, Gel , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Intracellular Signaling Peptides and Proteins , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Protein Kinases/metabolism , Proteins , Proto-Oncogene Proteins c-ets/genetics , RNA Interference , TOR Serine-Threonine Kinases , Transcription Factors/genetics , Transfection
6.
Genes Cells ; 14(2): 179-89, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19170765

ABSTRACT

Glutamine : fructose-6-phosphate amidotransferase 1 (GFAT1) was identified as a protein phosphorylated in glucose-deprived cells by immunoprecipitation using the anti-phospho Akt substrates (PAS) antibody, which recognizes the phosphorylation motif site by AMP-activated protein kinase (AMPK), followed by mass fingerprinting analysis. Glucose depletion-induced phosphorylation of endogenous GFAT was potentiated by 2-deoxyglucose (2-DG), an AMPK activator, and the 2-DG-stimulated phosphorylation of FLAG-tagged GFAT1 in transfected cells was suppressed by Compound C, an AMPK inhibitor. The 2-DG induced phosphorylation of GFAT1 was attenuated by the introduction of the kinase-negative mutant of AMPK, and the phosphorylation was observed in the cells expressing the constitutively active mutant of AMPK even in the absence of 2-DG. Subsequent analysis revealed that the PAS antibody recognized GFAT1 phosphorylated at Ser243, which is conserved among different species. The assay of the GFAT enzymatic activity in the cell lysates indicated that the 2-DG-treatment inhibited the enzymatic activity, and Compound C-preincubation partially prevented the 2-DG-induced decrease of the activity. Furthermore, the mutant replacing Ser243 by alanine partially prevented the decrease of GFAT activity by 2-DG treatment. These results indicate that the phosphorylation of GFAT1 at Ser243 by AMPK has an important role in the regulation of the GFAT1 enzymatic activity.


Subject(s)
AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/physiology , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Enzyme Activation , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/antagonists & inhibitors , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/chemistry , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/genetics , Humans , Molecular Sequence Data , Phosphorylation , Point Mutation , Sequence Homology, Amino Acid , Serine/genetics , Serine/metabolism , Transfection
7.
Biochem Soc Trans ; 37(Pt 1): 223-6, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19143636

ABSTRACT

The signalling function of mTOR complex 1 is activated by Rheb-GTP, which controls the catalytic competence of the mTOR (mammalian target of rapamycin) kinase domain by an incompletely understood mechanism. Rheb can bind directly to the mTOR kinase domain, and association with inactive nucleotide-deficient Rheb mutants traps mTOR in a catalytically inactive state. Nevertheless, Rheb-GTP targets other than mTOR, such as FKBP38 (FK506-binding protein 38) and/or PLD1 (phospholipase D(1)), may also contribute to mTOR activation. Once activated, the mTOR catalytic domain phosphorylates substrates only when they are bound to raptor (regulatory associated protein of mTOR), a separate polypeptide within the complex. The mechanism of insulin/nutrient stimulation of mTOR complex 1 signalling, in addition to Rheb-GTP activation of the mTOR catalytic function, also involves a stable modification of the configuration of mTORC1 (mTOR complex 1) that increases access of substrates to their binding site on the raptor polypeptide. The mechanism underlying this second step in the activation of mTORC1 is unknown.


Subject(s)
Guanosine Triphosphate/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neuropeptides/metabolism , Transcription Factors/metabolism , Animals , Catalysis , Enzyme Activation , Humans
8.
Mol Cell Biol ; 24(15): 6710-8, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15254238

ABSTRACT

TOR is a serine-threonine kinase that was originally identified as a target of rapamycin in Saccharomyces cerevisiae and then found to be highly conserved among eukaryotes. In Drosophila melanogaster, inactivation of TOR or its substrate, S6 kinase, results in reduced cell size and embryonic lethality, indicating a critical role for the TOR pathway in cell growth control. However, the in vivo functions of mammalian TOR (mTOR) remain unclear. In this study, we disrupted the kinase domain of mouse mTOR by homologous recombination. While heterozygous mutant mice were normal and fertile, homozygous mutant embryos died shortly after implantation due to impaired cell proliferation in both embryonic and extraembryonic compartments. Homozygous blastocysts looked normal, but their inner cell mass and trophoblast failed to proliferate in vitro. Deletion of the C-terminal six amino acids of mTOR, which are essential for kinase activity, resulted in reduced cell size and proliferation arrest in embryonic stem cells. These data show that mTOR controls both cell size and proliferation in early mouse embryos and embryonic stem cells.


Subject(s)
Embryo, Mammalian/cytology , Protein Kinases/physiology , Stem Cells/physiology , Animals , Blastocyst/metabolism , Blotting, Southern , Cell Cycle , Cell Division , Flow Cytometry , Gene Deletion , Genotype , Heterozygote , Mice , Mice, Knockout , Models, Genetic , Mutation , Protein Kinases/metabolism , Protein Structure, Tertiary , Recombination, Genetic , Ribosomal Protein S6 Kinases/metabolism , Sirolimus/pharmacology , Stem Cells/metabolism , TOR Serine-Threonine Kinases , Time Factors , Tissue Distribution
9.
J Biochem ; 139(1): 129-35, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16428328

ABSTRACT

Heat shock protein 90 (Hsp90) was co-immunoprecipitated with raptor, the binding partner of the mammalian target of rapamycin (mTOR) from HEK293 cells. Hsp90 was detected in the anti-raptor antibody immunoprecipitates prepared from the cell extract by immunoblot analysis using the anti-Hsp90 antibody, and the association of these two proteins was confirmed by immunoprecipitation from the cells co-expressing Hsp90 and raptor as epitope-tagged molecules. Geldanamycin, a potent inhibitor of Hsp90, disrupted the in vivo binding of Hsp90 to raptor without affecting the association of raptor and mTOR, and suppressed the phosphorylation by mTOR of the downstream translational regulators p70 S6 kinase (S6K) and eukaryotic initiation factor 4E-binding protein 1 (4E-BP1). The protein kinase activity of S6K as well as the phosphorylation of the substrate, 40S ribosomal protein S6, were lowered in the geldanamycin-treated cells. These results indicate that Hsp90 is involved in the regulation of protein translation by facilitating the phosphorylation reaction of 4E-BP1 and S6K catalyzed by the mTOR/raptor complex through the association with raptor, and that the mTOR signaling pathway is a novel target of geldanamycin.


Subject(s)
Benzoquinones/pharmacology , Enzyme Inhibitors/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/pharmacology , Protein Kinases/metabolism , Proteins/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Cell Line , Humans , Immunoprecipitation , Molecular Sequence Data , Phosphorylation , Regulatory-Associated Protein of mTOR , TOR Serine-Threonine Kinases
10.
FEBS Lett ; 587(18): 2924-9, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23892082

ABSTRACT

Target of rapamycin complex 1 (TORC1) has a key role in cellular regulations in response to environmental conditions. In yeast, Tip41 downregulates TORC1 signaling via activation of PP2A phosphatase. We show here that overexpression of TIPRL, a mammalian Tip41, suppressed dephosphorylation of mechanistic TORC1 (mTORC1) substrates under amino acid withdrawal, and knockdown of TIPRL conversely attenuated phosphorylation of those substrates after amino acid refeeding. TIPRL associated with the catalytic subunit of PP2A (PP2Ac), which was required for the TIPRL action on mTORC1 signaling. Collectively, unlike yeast TIP41, TIPRL has a positive effect on mTORC1 signaling through the association with PP2Ac.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Multiprotein Complexes/metabolism , Protein Phosphatase 2/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/genetics , Point Mutation , Protein Phosphatase 2/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , TOR Serine-Threonine Kinases/genetics , Transfection
11.
Mol Cell Biol ; 30(4): 1049-58, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19995911

ABSTRACT

Autophagy is a bulk proteolytic process that is indispensable for cell survival during starvation. Autophagy is induced by nutrient deprivation via inactivation of the rapamycin-sensitive Tor complex1 (TORC1), a protein kinase complex regulating cell growth in response to nutrient conditions. However, the mechanism by which TORC1 controls autophagy and the direct target of TORC1 activity remain unclear. Atg13 is an essential regulatory component of autophagy upstream of the Atg1 kinase complex, and here we show that yeast TORC1 directly phosphorylates Atg13 at multiple Ser residues. Additionally, expression of an unphosphorylatable Atg13 mutant bypasses the TORC1 pathway to induce autophagy through activation of Atg1 in cells growing under nutrient-rich conditions. Our findings suggest that the direct control of the Atg1 complex by TORC1 induces autophagy.


Subject(s)
Autophagy , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/enzymology , Amino Acid Sequence , Autophagy-Related Proteins , Microscopy, Electron , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Kinases/chemistry , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
12.
Mol Biol Cell ; 20(7): 1981-91, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19211835

ABSTRACT

Autophagy is an intracellular degradation system, by which cytoplasmic contents are degraded in lysosomes. Autophagy is dynamically induced by nutrient depletion to provide necessary amino acids within cells, thus helping them adapt to starvation. Although it has been suggested that mTOR is a major negative regulator of autophagy, how it controls autophagy has not yet been determined. Here, we report a novel mammalian autophagy factor, Atg13, which forms a stable approximately 3-MDa protein complex with ULK1 and FIP200. Atg13 localizes on the autophagic isolation membrane and is essential for autophagosome formation. In contrast to yeast counterparts, formation of the ULK1-Atg13-FIP200 complex is not altered by nutrient conditions. Importantly, mTORC1 is incorporated into the ULK1-Atg13-FIP200 complex through ULK1 in a nutrient-dependent manner and mTOR phosphorylates ULK1 and Atg13. ULK1 is dephosphorylated by rapamycin treatment or starvation. These data suggest that mTORC1 suppresses autophagy through direct regulation of the approximately 3-MDa ULK1-Atg13-FIP200 complex.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Amino Acids/pharmacology , Autophagy/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Amino Acids/deficiency , Animals , Autophagy-Related Protein-1 Homolog , Autophagy-Related Proteins , Cell Line , Cell Membrane/drug effects , Cell Membrane/enzymology , Cell Membrane/ultrastructure , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Molecular Weight , Multiprotein Complexes , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Kinases/metabolism , Protein Transport/drug effects , Proteins , TOR Serine-Threonine Kinases
13.
J Biol Chem ; 283(7): 4430-8, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18063581

ABSTRACT

Sufficiency and depletion of nutrients regulate the cellular activities through the protein phosphorylation reaction; however, many protein substrates remain to be clarified. GBF1 (Golgi-specific brefeldin A resistance factor 1), a guanine nucleotide exchange factor for the ADP-ribosylation factor family associated with the Golgi apparatus, was isolated as a phosphoprotein from the glucose-depleted cells by using the phospho-Akt-substrate antibody, which recognizes the substrate proteins of several protein kinases. The phosphorylation of GBF1 was induced by 2-deoxyglucose (2-DG), which blocks glucose utilization and increases the intracellular AMP concentration, and by AICAR, an AMP-activated protein kinase (AMPK) activator. This phosphorylation was observed in the cells expressing the constitutively active AMPK. The 2-DG-induced phosphorylation of GBF1 was suppressed by Compound C, an AMPK inhibitor, and by the overexpression of the kinase-negative AMPK. Analysis using the deletion and point mutants identified Thr(1337) as the 2-DG-induced phosphorylation site in GBF1, which is phosphorylated by AMPK in vitro. ATP depletion is known to provoke the Golgi apparatus disassembly. Immunofluorescent microscopic analysis with the Golgi markers indicated that GBF1 associates with the fragmented Golgi apparatus in the cells treated with 2-DG and AICAR. The expression of the kinase-negative AMPK and the GBF1 mutant replacing Thr(1337) by Ala prevented the 2-DG-induced Golgi disassembly. These results indicate that GBF1 is a novel AMPK substrate and that the AMPK-mediated phosphorylation of GBF1 at Thr(1337) has a critical role, presumably by attenuating the function of GBF1, in the disassembly of the Golgi apparatus induced under stress conditions that lower the intracellular ATP concentration.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Golgi Apparatus/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Threonine/metabolism , Animals , Cell Line , Fluorescent Antibody Technique , Guanine Nucleotide Exchange Factors/chemistry , Humans , Phosphorylation
14.
Biochem Biophys Res Commun ; 361(1): 218-23, 2007 Sep 14.
Article in English | MEDLINE | ID: mdl-17658474

ABSTRACT

TBC7, a TBC (Tre-2/Bub2/Cdc16) 1 domain protein, was identified as a novel binding protein to the TSC1-TSC2 tumor suppressor complex by peptide mass fingerprinting analysis of the proteins immunoprecipitated with FLAG-epitope tagged TSC1 and TSC2 from the transfected mammalian cells. The in vivo and in vitro association of TBC7 and the TSC1-TSC2 complex was confirmed by the co-immunoprecipitation and pull-down analysis, respectively, and TBC7 was revealed to bind to the C-terminal half region of TSC1, which is distinct from the binding site with TSC2. The immunofluorescence microscopy and subcellular fractionation showed that TBC7 co-localizes with the tumor suppressor complex in the endomembrane. Overexpression of TBC7 enhanced ubiquitination of TSC1 and increased phosphorylation of S6 protein by S6 kinase, that is located in the mTOR-signaling pathway. These results indicate TBC7 could take a part in the negative regulation of the tumor suppressor complex through facilitating the downregulation of TSC1.


Subject(s)
Carrier Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Binding Sites , Carrier Proteins/analysis , Carrier Proteins/chemistry , Down-Regulation , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Protein Kinases/metabolism , Protein Structure, Tertiary , Signal Transduction , TOR Serine-Threonine Kinases , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein
15.
J Biol Chem ; 282(28): 20329-39, 2007 Jul 13.
Article in English | MEDLINE | ID: mdl-17517883

ABSTRACT

The proline-rich Akt substrate of 40 kilodaltons (PRAS40) was identified as a raptor-binding protein that is phosphorylated directly by mammalian target of rapamycin (mTOR) complex 1 (mTORC1) but not mTORC2 in vitro, predominantly at PRAS40 (Ser(183)). The binding of S6K1 and 4E-BP1 to raptor requires a TOR signaling (TOS) motif, which contains an essential Phe followed by four alternating acidic and small hydrophobic amino acids. PRAS40 binding to raptor was severely inhibited by mutation of PRAS40 (Phe(129) to Ala). Immediately carboxyl-terminal to Phe(129) are two small hydrophobic amino acid followed by two acidic residues. PRAS40 binding to raptor was also abolished by mutation of the major mTORC1 phosphorylation site, Ser(183), to Asp. PRAS40 (Ser(183)) was phosphorylated in intact cells; this phosphorylation was inhibited by rapamycin, by 2-deoxyglucose, and by overexpression of the tuberous sclerosis complex heterodimer. PRAS40 (Ser(183)) phosphorylation was also inhibited reversibly by withdrawal of all or of only the branched chain amino acids; this inhibition was reversed by overexpression of the Rheb GTPase. Overexpressed PRAS40 suppressed the phosphorylation of S6K1 and 4E-BP1 at their rapamycin-sensitive phosphorylation sites, and reciprocally, overexpression of S6K1 or 4E-BP1 suppressed phosphorylation of PRAS40 (Ser(183)) and its binding to raptor. RNA interference-induced depletion of PRAS40 enhanced the amino acid-stimulated phosphorylation of both S6K1 and 4E-BP1. These results establish PRAS40 as a physiological mTORC1 substrate that contains a variant TOS motif. Moreover, they indicate that the ability of raptor to bind endogenous substrates is limiting for the activity of mTORC1 in vivo and is therefore a potential locus of regulation.


Subject(s)
Phosphoproteins/metabolism , Protein Processing, Post-Translational/physiology , Proteins/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Substitution , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Gene Expression , HeLa Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Multiprotein Complexes , Mutation, Missense , Neuropeptides/genetics , Neuropeptides/metabolism , Phosphoproteins/genetics , Phosphorylation , Protein Binding/physiology , Proteins/genetics , RNA Interference , Ras Homolog Enriched in Brain Protein , Regulatory-Associated Protein of mTOR , Ribosomal Protein S6 Kinases/genetics , Ribosomal Protein S6 Kinases/metabolism , TOR Serine-Threonine Kinases , Transcription Factors/genetics
16.
Genes Cells ; 11(7): 757-66, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16824195

ABSTRACT

The translational regulator protein 4E-BP1, that binds to eukaryotic initiation factor-4E (eIF4E) to prevent the formation of the active translation complex, dissociates from eIF4E by phosphorylation through the mammalian target of rapamycin (mTOR) in the cells stimulated by amino acids. 4E-BP1 has been shown to associate with the scaffold protein raptor through its TOS and RAIP motifs to be recognized by mTOR. We revealed that the TOS motif mutant was phosphorylated by mTOR only at the priming sites of Thr37/46 but the RAIP motif mutant was phosphorylated not only at the priming sites but also at the subsequent site of Thr70 in vitro and in response to amino acid treatment in HEK293 cells. Analysis using the phosphorylation site mutants indicated that phosphorylation of the priming and subsequent sites of 4E-BP1 was required for dissociation from raptor as well as for the release of eIF4E. The expression of the 4E-BP1 mutants replacing the TOS motif and phosphorylation sites, that are poor substrates for mTOR and have no or little dissociation ability from raptor and eIF4E, respectively, significantly reduced the size of K562 cells. These results indicate that the the TOS motif has an essential function whereas the RAIP motif has an accessory role in the association with raptor and mTOR-mediated phosphorylation of 4E-BP1 to dissociate it from raptor and release eIF4E in response to amino acid stimulation leading to the control of cell size.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Phosphoproteins/metabolism , Protein Kinases/metabolism , Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Cycle Proteins , Cell Size , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Humans , K562 Cells , Mutagenesis, Site-Directed , Phosphoproteins/genetics , Phosphorylation , Regulatory-Associated Protein of mTOR , TOR Serine-Threonine Kinases
17.
J Biol Chem ; 281(42): 31627-37, 2006 Oct 20.
Article in English | MEDLINE | ID: mdl-16905533

ABSTRACT

Diacylglycerol (DAG) acts as an allosteric activator of protein kinase C (PKC) and is converted to phosphatidic acid by DAG kinase (DGK). Therefore, DGK is thought to be a negative regulator of PKC activation. Here we show molecular mechanisms of functional coupling of the two kinases. gammaPKC directly associated with DGKgamma through its accessory domain (AD), depending on Ca2+ as well as phosphatidylserine/diolein in vitro. Mass spectrometric analysis and mutation studies revealed that gammaPKC phosphorylated Ser-776 and Ser-779 in the AD of DGKgamma. The phosphorylation by gammaPKC resulted in activation of DGKgamma because a DGKgamma mutant in which Ser-776 and Ser-779 were substituted with glutamic acid to mimic phosphorylation exhibited significantly higher activity compared with wild type DGKgamma and an unphosphorylatable DGKgamma mutant. Importantly, the interaction of the two kinases and the phosphorylation of DGKgamma by gammaPKC could be confirmed in vivo, and overexpression of the AD of DGKgamma inhibited re-translocation of gammaPKC. These results demonstrate that localization and activation of the functionally correlated kinases, gammaPKC and DGKgamma, are spatio-temporally orchestrated by their direct association and phosphorylation, contributing to subtype-specific regulation of DGKgamma and DAG signaling.


Subject(s)
Diacylglycerol Kinase/biosynthesis , Gene Expression Regulation, Enzymologic , Protein Kinase C/metabolism , Up-Regulation , Amino Acid Sequence , Animals , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , Molecular Sequence Data , Phosphorylation , Protein Kinase C/chemistry , Rats , Sequence Homology, Amino Acid
18.
J Biol Chem ; 280(15): 15029-37, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15699050

ABSTRACT

Here we describe mass spectrometric identification, molecular cloning, and biochemical characterization of a lipid/membrane raft-associated protein that is tyrosine-phosphorylated upon Xenopus egg fertilization. This protein is homologous to mammalian uroplakin III, a member of the uroplakin family proteins (UPs) that constitute asymmetric unit membranes in the mammalian urothelial tissues, thus termed Xenopus uroplakin III (xUPIII). xUPIII contains N-linked sugars and is highly expressed in Xenopus eggs, ovary, urinary tract, and kidney. In unfertilized eggs, xUPIII is predominantly localized to the lipid/membrane rafts and exposed on the cell surface, as judged by surface biotinylation experiments and indirect immunofluorescent studies. After fertilization or hydrogen peroxide-induced egg activation, xUPIII becomes rapidly phosphorylated on tyrosine residue-249, which locates in the carboxyl-terminal cytoplasmic tail of the molecule. Raft localization and tyrosine phosphorylation of xUPIII can be reconstituted in HEK293 cells by coexpression of xUPIII, and Xenopus c-Src, a tyrosine kinase whose fertilization-induced activation in egg rafts is required for initiation of development. In mammals, UPIII is forming a complex with a tetraspanin molecule uroplakin Ib. As another tetraspanin, CD9, is known to be a critical component for sperm-egg fusion in the mouse, we have assumed that xUPIII is involved in sperm-egg interaction. An antibody against the extracellular domain of xUPIII blocks sperm-egg interaction, as judged by the occurrence of egg activation and first cell cleavage. Thus, xUPIII represents an egg raft-associated protein that is likely involved in sperm-egg interaction as well as subsequent Src-dependent intracellular events of egg activation in Xenopus.


Subject(s)
Membrane Glycoproteins/metabolism , Tyrosine/chemistry , Amino Acid Sequence , Animals , Antigens, CD/chemistry , Biotinylation , CSK Tyrosine-Protein Kinase , Cell Line , Cell Membrane/metabolism , Centrifugation, Density Gradient , Cloning, Molecular , Cytoplasm/metabolism , DNA, Complementary/metabolism , Electrophoresis, Polyacrylamide Gel , Expressed Sequence Tags , Female , Fertilization , Fluorescent Antibody Technique, Indirect , Glutathione Transferase/metabolism , Humans , Hydrogen Peroxide/chemistry , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Lipid Metabolism , Male , Mass Spectrometry , Membrane Glycoproteins/chemistry , Membrane Microdomains/metabolism , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Protein-Tyrosine Kinases/metabolism , Recombinant Fusion Proteins/chemistry , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sperm-Ovum Interactions , Tetraspanin 29 , Tissue Distribution , Uroplakin III , Uroplakin Ib , Xenopus , src-Family Kinases
19.
Biochem Biophys Res Commun ; 313(2): 437-41, 2004 Jan 09.
Article in English | MEDLINE | ID: mdl-14684181

ABSTRACT

The mammalian target of rapamycin (mTOR) controls cell growth in response to amino acids and growth factors, in part by regulating p70 S6 kinase alpha (p70 alpha) and eukaryotic initiation factor 4E binding protein 1 (4EBP1). Raptor (regulatory associated protein of mTOR) is a 150 kDa mTOR binding protein that is essential for TOR signaling in vivo and also binds 4EBP1 and p70alpha through their respective TOS (TOR signaling) motifs, a short conserved segment previously shown to be required for amino acid- and mTOR-dependent regulation of these substrates in vivo. Raptor appears to serve as an mTOR scaffold protein, the binding of which to the TOS motif of mTOR substrates is necessary for effective mTOR-catalyzed phosphorylation. Further understanding of regulation of the mTOR-raptor complex in response to the nutritional environment would require identification of the interplay between the mTOR-raptor complex and its upstream effectors such as the protein products of tumor suppressor gene tuberous sclerosis complexes 1 and 2, and the Ras-related small G protein Rheb.


Subject(s)
Protein Kinases/metabolism , Proteins/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Motifs , Amino Acid Sequence , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins , Conserved Sequence , Humans , Molecular Sequence Data , Phosphoproteins/metabolism , Phosphorylation , Proteins/chemistry , Proteins/genetics , Regulatory-Associated Protein of mTOR , Sequence Alignment , Sequence Homology, Amino Acid , TOR Serine-Threonine Kinases
20.
Eur J Biochem ; 269(21): 5203-14, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12392552

ABSTRACT

Mitochondrial ribosomal proteins (mrps) of the budding yeast, Saccharomyces cerevisiae, have been extensively characterized genetically and biochemically. However, the list of the genes encoding individual mrps is still not complete and quite a few of the mrps are only predicted from their similarity to bacterial ribosomal proteins. We have constructed a yeast strain in which one of the small subunit proteins, termed Mrp4, was tagged with S-peptide and used for affinity purification of mitochondrial ribosome. Mass spectrometric analysis of the isolated proteins detected most of the small subunit mrps which were previously identified or predicted and about half of the large subunit mrps. In addition, several proteins of unknown function were identified. To confirm their identity further, we added tags to these proteins and analyzed their localization in subcellular fractions. Thus, we have newly established Ymr158w (MrpS8), Ypl013c (MrpS16), Ymr188c (MrpS17) and Ygr165w (MrpS35) as small subunit mrps and Img1, Img2, Ydr116c (MrpL1), Ynl177c (MrpL22), Ynr022c (MrpL50) and Ypr100w (MrpL51) as large subunit mrps.


Subject(s)
Mitochondria/chemistry , Multidrug Resistance-Associated Proteins , Ribosomal Proteins/chemistry , Ribosomes/chemistry , Amino Acid Sequence , Base Sequence , Macromolecular Substances , Mass Spectrometry , Mitochondrial Proteins , Molecular Sequence Data , Ribosomal Proteins/genetics , Ribosomal Proteins/isolation & purification , Ribosomal Proteins/metabolism , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/isolation & purification , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL