Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Mol Cancer ; 9: 104, 2010 May 11.
Article in English | MEDLINE | ID: mdl-20459822

ABSTRACT

BACKGROUND: Intestinal cell kinase (ICK; GeneID 22858) is a conserved MAPK and CDK-like kinase that is widely expressed in human tissues. Data from the Cancer Genome Anatomy Project indicated ICK mRNA is increased in cancer, and that its expression correlated with expression of mRNA for an uncharacterized F-box protein, FBX9 (GeneID: 26268). ICK and FBX9 genes are arranged head-to-head on opposite strands, with start sites for transcription separated by approximately 3.3 kb. We hypothesized ICK and FBX9 are potentially important genes in cancer controlled by a bidirectional promoter. RESULTS: We assessed promoter activity of the intergenic region in both orientations in cancer cell lines derived from breast (AU565, SKBR3), colon (HCT-15, KM12), and stomach (AGS) cancers, as well as in embryonic human kidney (HEK293T) cells. The intergenic segment was active in both orientations in all of these lines, and ICK promoter activity was greater than FBX9 promoter activity. Results from deletions and truncations defined a minimal promoter for ICK, and revealed that repressors and enhancers differentially regulate ICK versus FBX9 promoter activity. The ICK promoter contains consensus motifs for several FOX-family transcription factors that align when mouse and human are compared using EMBOSS. FOXA1 and FOXA2 increase luciferase activity of a minimal promoter 10-20 fold in HEK293T cells. Consensus sites for TCF7L2 (TCF4) (Gene Id: 6934) are also present in both mouse and human. The expression of beta-catenin increased activity of the minimal promoter approximately 10 fold. ICK reference mRNAs (NM_014920.3, NM_016513) are expressed in low copy number and increased in some breast cancers, using a ten base tag 5'-TCAACCTTAT-3' specific for both ICK transcripts. CONCLUSION: ICK and FBX9 are divergently transcribed from a bidirectional promoter that is GC-rich and contains a CpG island. A minimal promoter for ICK contains functional sites for beta-catenin/TCF7L2 and FOXA. These data are consistent with functions that have been proposed for ICK in development and in proliferation or survival of some breast and colon cancers.


Subject(s)
F-Box Proteins/genetics , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic/genetics , Promoter Regions, Genetic/genetics , Protein Serine-Threonine Kinases/genetics , TCF Transcription Factors/genetics , Animals , Base Sequence , Blotting, Western , Cell Line, Tumor , Consensus Sequence , Conserved Sequence , F-Box Proteins/metabolism , Forkhead Transcription Factors/metabolism , Humans , Mice , Molecular Sequence Data , Protein Serine-Threonine Kinases/metabolism , Sequence Alignment , TCF Transcription Factors/metabolism , Transcription Factor 7-Like 2 Protein , beta Catenin/genetics , beta Catenin/metabolism
2.
ACS Chem Biol ; 4(12): 999-1015, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-19902965

ABSTRACT

TOR (Target of Rapamycin) is a highly conserved Ser/Thr kinase and a central controller of cell growth. Using the crystal structure of the related lipid kinase PI3KCgamma, we built a model of the catalytic region of TOR, from the FAT domain to near the end of the FATC domain. The model reveals that activating mutations in TOR, identified in yeast in a genetic selection for Rheb-independence, correspond to hotspots for oncogenic mutations in PI3KCalpha. The activating mutations are in the catalytic domain (helices kalpha3, kalpha9, kalpha11) and the helical domain of TOR. Docking studies with small molecule inhibitors (PP242, NVP-BEZ235, and Ku-0063794) show that drugs currently in development utilize a novel pharmacophore space to achieve specificity. Thus, our model provides insight on the regulation of TOR and may be useful in the design of new anticancer drugs.


Subject(s)
Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/genetics , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Catalytic Domain , Crystallography, X-Ray , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Protein Conformation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Schizosaccharomyces/enzymology , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Sequence Alignment , TOR Serine-Threonine Kinases
3.
Mol Cell ; 35(5): 563-73, 2009 Sep 11.
Article in English | MEDLINE | ID: mdl-19748353

ABSTRACT

The target of rapamycin complex 1 (TORC1) is a central regulator of eukaryotic cell growth that is activated by a variety of hormones (e.g., insulin) and nutrients (e.g., amino acids) and is deregulated in various cancers. Here, we report that the yeast Rag GTPase homolog Gtr1, a component of the vacuolar-membrane-associated EGO complex (EGOC), interacts with and activates TORC1 in an amino-acid-sensitive manner. Expression of a constitutively active (GTP-bound) Gtr1(GTP), which interacted strongly with TORC1, rendered TORC1 partially resistant to leucine deprivation, whereas expression of a growth inhibitory, GDP-bound Gtr1(GDP), caused constitutively low TORC1 activity. We also show that the nucleotide-binding status of Gtr1 is regulated by the conserved guanine nucleotide exchange factor (GEF) Vam6. Thus, in addition to its regulatory role in homotypic vacuolar fusion and vacuole protein sorting within the HOPS complex, Vam6 also controls TORC1 function by activating the Gtr1 subunit of the EGO complex.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Intracellular Membranes/enzymology , Monomeric GTP-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Vacuoles/enzymology , Adaptor Proteins, Vesicular Transport/genetics , Amino Acid Transport Systems/metabolism , Amino Acids/metabolism , Cycloheximide/pharmacology , DNA-Binding Proteins/metabolism , Endosomes/enzymology , Guanine Nucleotide Exchange Factors/genetics , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Intracellular Membranes/drug effects , Monomeric GTP-Binding Proteins/genetics , Multiprotein Complexes , Mutation , Protein Binding , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Synthesis Inhibitors/pharmacology , Protein Transport , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction , Sirolimus/pharmacology , Time Factors , Transcription Factors/metabolism , Vacuoles/drug effects
4.
Am J Physiol Gastrointest Liver Physiol ; 297(4): G632-40, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19696144

ABSTRACT

Intestinal cell kinase (ICK), originally cloned from the intestine and expressed in the intestinal crypt epithelium, is a highly conserved serine/threonine protein kinase that is similar to mitogen-activated protein kinases (MAPKs) in the catalytic domain and requires dual phosphorylation within a MAPK-like TDY motif for full activation. Despite these similarities to MAPKs, the biological functions of ICK remain unknown. In this study, we report that suppression of ICK expression in cultured intestinal epithelial cells by short hairpin RNA (shRNA) interference significantly impaired cellular proliferation and induced features of gene expression characteristic of colonic or enterocytic differentiation. Downregulation of ICK altered expression of cell cycle regulators (cyclin D1, c-Myc, and p21(Cip1/WAF1)) of G(1)-S transition, consistent with the G(1) cell cycle delay induced by ICK shRNA. ICK deficiency also led to a significant decrease in the expression and/or activity of p70 ribosomal protein S6 kinase (S6K1) and eukaryotic initiation factor 4E (eIF4E), concomitant with reduced expression of their upstream regulators, the mammalian target of rapamycin (mTOR) and the regulatory associated protein of mTOR (Raptor). Furthermore, ICK interacts with the mTOR/Raptor complex in vivo and phosphorylates Raptor in vitro. These results suggest that disrupting ICK function may downregulate protein translation of specific downstream targets of eIF4E and S6K1 such as cyclin D1 and c-Myc through the mTOR/Raptor signaling pathway. Taken together, our findings demonstrate an important role for ICK in proliferation and differentiation of intestinal epithelial cells.


Subject(s)
Cell Differentiation/genetics , Cell Proliferation , Epithelial Cells/enzymology , G1 Phase , Intestinal Mucosa/enzymology , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Caco-2 Cells , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Epithelial Cells/pathology , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Regulation , Humans , Intestinal Mucosa/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proteins , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , Regulatory-Associated Protein of mTOR , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , TOR Serine-Threonine Kinases , Transcription Factors/metabolism , Transfection
5.
J Biol Chem ; 284(22): 14693-7, 2009 May 29.
Article in English | MEDLINE | ID: mdl-19346248

ABSTRACT

mTORC1 contains multiple proteins and plays a central role in cell growth and metabolism. Raptor (regulatory-associated protein of mammalian target of rapamycin (mTOR)), a constitutively binding protein of mTORC1, is essential for mTORC1 activity and critical for the regulation of mTORC1 activity in response to insulin signaling and nutrient and energy sufficiency. Herein we demonstrate that mTOR phosphorylates raptor in vitro and in vivo. The phosphorylated residues were identified by using phosphopeptide mapping and mutagenesis. The phosphorylation of raptor is stimulated by insulin and inhibited by rapamycin. Importantly, the site-directed mutation of raptor at one phosphorylation site, Ser(863), reduced mTORC1 activity both in vitro and in vivo. Moreover, the Ser(863) mutant prevented small GTP-binding protein Rheb from enhancing the phosphorylation of S6 kinase (S6K) in cells. Therefore, our findings indicate that mTOR-mediated raptor phosphorylation plays an important role on activation of mTORC1.


Subject(s)
Phosphoproteins/metabolism , Protein Kinases/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Humans , Mice , Phosphorylation/drug effects , Phosphoserine/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sirolimus/pharmacology , Substrate Specificity/drug effects , TOR Serine-Threonine Kinases
6.
Eukaryot Cell ; 7(10): 1819-30, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18723607

ABSTRACT

TOR is a structurally and functionally conserved Ser/Thr kinase found in two multiprotein complexes that regulate many cellular processes to control cell growth. Although extensively studied, the localization of TOR is still ambiguous, possibly because endogenous TOR in live cells has not been examined. Here, we examined the localization of green fluorescent protein (GFP) tagged, endogenous TOR1 and TOR2 in live S. cerevisiae cells. A DNA cassette encoding three copies of green fluorescent protein (3XGFP) was inserted in the TOR1 gene (at codon D330) or the TOR2 gene (at codon N321). The TORs were tagged internally because TOR1 or TOR2 tagged at the N or C terminus was not functional. The TOR1(D330-3XGFP) strain was not hypersensitive to rapamycin, was not cold sensitive, and was not resistant to manganese toxicity caused by the loss of Pmr1, all indications that TOR1-3XGFP was expressed and functional. TOR2-3XGFP was functional, as TOR2 is an essential gene and TOR2(N321-3XGFP) haploid cells were viable. Thus, TOR1 and TOR2 retain function after the insertion of 748 amino acids in a variable region of their noncatalytic domain. The localization patterns of TOR1-3XGFP and TOR2-3XGFP were documented by imaging of live cells. TOR1-3XGFP was diffusely cytoplasmic and concentrated near the vacuolar membrane. The TOR2-3XGFP signal was cytoplasmic but predominately in dots at the plasma membrane. Thus, TOR1 and TOR2 have distinct localization patterns, consistent with the regulation of cellular processes as part of two different complexes.


Subject(s)
Cell Cycle Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Membrane/chemistry , Cell Membrane/genetics , Cell Membrane/metabolism , Cytoplasm/chemistry , Cytoplasm/genetics , Cytoplasm/metabolism , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/genetics , Protein Transport , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sequence Alignment
7.
Am J Physiol Gastrointest Liver Physiol ; 295(3): G421-30, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18467502

ABSTRACT

Tristetraprolin (TTP) is the prototype for a family of RNA binding proteins that bind the tumor necrosis factor (TNF) messenger RNA AU-rich element (ARE), causing deadenylation of the TNF poly(A) tail, RNA decay, and silencing of TNF protein production. Using mass spectrometry sequencing we identified poly(A) binding proteins-1 and -4 (PABP1 and PABP4) in high abundance and good protein coverage from TTP immunoprecipitates. PABP1 significantly enhanced TNF ARE binding by RNA EMSA and prevented TTP-initiated deadenylation in an in vitro macrophage assay of TNF poly(A) stability. Neomycin inhibited TTP-promoted deadenylation at concentrations shown to inhibit the deadenylases poly(A) ribonuclease and CCR4. Stably transfected RAW264.7 macrophages overexpressing PABP1 do not oversecrete TNF; instead they upregulate TTP protein without increasing TNF protein production. The PABP1 inhibition of deadenylation initiated by TTP does not require the poly(A) binding regions in RRM1 and RRM2, suggesting a more complicated interaction than simple masking of the poly(A) tail from a 3'-exonuclease. Like TTP, PABP1 is a substrate for p38 MAP kinase. Finally, PABP1 stabilizes cotransfected TTP in 293T cells and prevents the decrease in TTP levels seen with p38 MAP kinase inhibition. These findings suggest several levels of functional antagonism between TTP and PABP1 that have implications for regulation of unstable mRNAs like TNF.


Subject(s)
Gene Silencing , Poly(A)-Binding Protein I/metabolism , RNA Stability , RNA, Messenger/metabolism , Tristetraprolin/metabolism , Tumor Necrosis Factor-alpha/metabolism , 3' Untranslated Regions , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Enzyme Inhibitors/pharmacology , Exoribonucleases/antagonists & inhibitors , Exoribonucleases/metabolism , Gene Silencing/drug effects , Humans , Immunoprecipitation , Mass Spectrometry , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Neomycin/pharmacology , Phosphorylation , Poly(A)-Binding Protein I/genetics , Ribonucleoside Diphosphate Reductase/metabolism , Time Factors , Transfection , Tumor Necrosis Factor-alpha/genetics , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Biochem Biophys Res Commun ; 371(1): 1-4, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18406346

ABSTRACT

The review highlights evidence for different functions in the cell cycle of the two MAP kinase kinases, MEK1 and MEK2, and the two MAP kinases, ERK1 and ERK2. Functional differences may explain why instances of cell cycle arrest can be MEK1 or MEK2 dependent.


Subject(s)
Cell Cycle , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Animals , Humans , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Phosphorylation
9.
Am J Physiol Gastrointest Liver Physiol ; 294(2): G452-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18032482

ABSTRACT

The MNK kinases are downstream of both the p38 and ERK MAP kinase pathways and act to increase gene expression. MNK inhibition using the compound CGP57380 has recently been reported to inhibit tumor necrosis factor (TNF) production in macrophage cell lines stimulated with Escherichia coli lipopolysaccharide (LPS). However, the range of receptors that signal through the MNK kinases and the extent of the resultant cytokine response are not known. We found that TNF production was inhibited in RAW264.7 macrophage cells by CGP57380 in a dose-responsive manner with agonists for Toll-like receptor (TLR) 2 (HKLM), TLR4 (Salmonella LPS), TLR6/2 (FSL), TLR7 (imiquimod), and TLR9 (CpG DNA). CGP57380 also inhibited the peak of TNF mRNA production and increased the rate of TNF mRNA decay, effects not due to the destabilizing RNA binding protein tristetraprolin (TTP). Similar to its effects on TNF, CGP57380 caused dose-responsive inhibition of TTP production from stimulation with either LPS or CpG DNA. MNK inhibition also blocked IL-6 but permitted IL-10 production in response to LPS. Studies using bone marrow-derived macrophages (BMDM) isolated from a spontaneous mouse model of Crohn's disease-like ileitis (SAMP1/YitFc strain) revealed significant inhibition by CGP57380 of the proinflammatory cytokines TNF, IL-6, and monocyte chemoattractant protein-1 at 4 and 24 h after LPS stimulation. IL-10 production was higher in CGP53870-treated BMDM at 4 h but was similar to the controls by 24 h. Taken together, these data demonstrate that MNK kinases signal through a variety of TLR agonists and mediate a potent innate, proinflammatory cytokine response.


Subject(s)
Cytokines/metabolism , Inflammation/metabolism , Macrophages/metabolism , Protein Serine-Threonine Kinases/physiology , Toll-Like Receptors/physiology , Aniline Compounds/pharmacology , Animals , Blotting, Western , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Crohn Disease/metabolism , Enzyme-Linked Immunosorbent Assay , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred Strains , Purines/pharmacology , RNA/biosynthesis , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/biosynthesis
11.
Genes Cells ; 12(10): 1133-40, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17903173

ABSTRACT

Map kinase-interacting protein kinases 1 and 2 (MNK1, MNK2) function downstream of p38 and ERK MAP kinases, but there are large gaps in our knowledge of how MNKs are regulated and function. Mice deleted of both genes are apparently normal, suggesting that MNKs function in adaptive pathways during stress. Here, we show that mouse embryo fibroblasts (MEFs) obtained from mnk1 (-/-)/mnk2 (-/-) as well as mnk1 (-/-) and mnk2 (-/-) mice are sensitized to caspase-3 activation upon withdrawal of serum in comparison to wild-type cells. Caspase-3 cleavage occurs with all cells in the panel, but most rapidly and robustly in cells derived from mice lacking both MNK genes. Treatment of wild-type MEFs in the panel with a compound (CGP57380) that inhibits MNK1 and MNK2 sensitizes wild-type cells for serum-withdrawal induced apoptosis, suggesting that sensitization is due to loss of MNK function and not to a secondary event. Reintroduction of wild-type MNK1 in the double knockout MEFs results in decreased sensitivity to serum withdrawal that is not observed for wild-type MNK2, or the kinase dead variant. Our work identifies MNKs as kinases involved in anti-apoptotic signaling in response to serum withdrawal.


Subject(s)
Apoptosis , Fibroblasts/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Aniline Compounds/pharmacology , Animals , Annexin A5/pharmacology , Blotting, Western , Cell Separation , Cells, Cultured , Culture Media, Serum-Free/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Flow Cytometry , Mice , Mice, Knockout , Purines/pharmacology
12.
Genetics ; 177(1): 231-8, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17603109

ABSTRACT

The Pmr1 Golgi Ca2+/Mn2+ ATPase negatively regulates target of rapamycin complex (TORC1) signaling, the rapamycin-sensitive TOR complex in Saccharomyces cerevisiae. Since pmr1 causes resistance to rapamycin and tor1 causes hypersensitivity, we looked for genetic interactions of pmr1 with tor1. Deletion of TOR1 restored two wild-type phenotypes. Loss of TOR1 restored the ability of the pmr1 strain to grow on media containing 2 mm MnCl2 and conferred wild type as well as the wild-type sensitivity to rapamycin. Mn2+ additions to media partially suppressed rapamycin resistance of wild type and pmr1 tor1, suggesting that Tor1 and Tor2 are regulated by manganese. We parsed the roles of Ca2+ and Mn2+ transport and the compartments in rapamycin response using separation-of-function mutants available for Pmr1. A strain containing the D53A mutant (Mn2+ transporting) of Pmr1 is rapamycin sensitive, but the Q783A mutant (Ca2+ transporting) strain is rapamycin resistant. Mn2+ transport into the Golgi lumen appears to be required for rapamycin sensitivity. Overexpression of Ca2+ pump SERCA1, Ca2+/H+ antiporter Vcx1, or a Mn2+ transporting mutant of Vcx1 (Vcx1-M1) failed to restore rapamycin sensitivity, and loss of Pmr1 but not other transporters of Ca2+ or Mn2+ results in rapamycin resistance. Overexpression of Ccc1, a Fe2+ and Mn2+ transporter that has been localized to Golgi and the vacuole, does restore rapamycin sensitivity to pmr1Delta. We conclude that Mn2+ in the Golgi inhibits TORC1 signaling.


Subject(s)
Calcium-Transporting ATPases/metabolism , Golgi Apparatus/metabolism , Immunosuppressive Agents/pharmacology , Manganese/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Signal Transduction/physiology , Sirolimus/pharmacology , Biological Transport , Blotting, Western , Calcium-Transporting ATPases/genetics , Cation Transport Proteins , Drug Resistance , Molecular Chaperones , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae Proteins/genetics
13.
J Biol Chem ; 282(7): 4243-4252, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-17130135

ABSTRACT

MAPK-interacting protein kinases 1 and 2 (MNK1 and MNK2) function downstream of p38 and ERK MAPK, but there are large gaps in our knowledge of how MNKs are regulated and function. As proteins activated in the HER2/Ras/Raf/ERK pathway, the MNKs are of potential interest in HER2-overexpressing cancers. We utilized a panel of breast cell lines (HCC1419, AU565, SKBR3, MCF7, and MCF10A), three of which overexpress HER2, to characterize the amounts and activation status of MNKs and other pathway enzymes (ERKs and RSKs) in these cells. We generated a phosphospecific antibody to Thr(P)-214 in the T-loop of MNKs and found that phosphorylations of both Thr-209 and Thr-214 in human MNK1 are required for activation. Increased phosphorylation and activity of the MNKs correlate with HER2 overexpression, and inhibition of the MNKs reduces colony formation in soft agar. Our work identifies the MNKs as potential therapeutic targets for breast cancer treatments.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Cell Line, Tumor , Enzyme Activation , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Phosphorylation , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/genetics , Receptor, ErbB-2/genetics , Tyrosine/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Proc Natl Acad Sci U S A ; 103(47): 17840-5, 2006 Nov 21.
Article in English | MEDLINE | ID: mdl-17095607

ABSTRACT

The rapamycin.FKBP12 complex inhibits target of rapamycin (TOR) kinase in TORC1. We screened the yeast nonessential gene deletion collection to identify mutants that conferred rapamycin resistance, and we identified PMR1, encoding the Golgi Ca2+/Mn2+ -ATPase. Deleting PMR1 in two genetic backgrounds confers rapamycin resistance. Epistasis analyses show that Pmr1 functions upstream from Npr1 and Gln-3 in opposition to Lst8, a regulator of TOR. Npr1 kinase is largely cytoplasmic, and a portion localizes to the Golgi where amino acid permeases are modified and sorted. Nuclear translocation of Gln-3 and Gln-3 reporter activity in pmr1 cells are impaired, but expression of functional Gap1 in the plasma membrane of a pmr1 strain in response to nitrogen limitation is enhanced. These two phenotypes suggest up-regulation of Npr1 function in the absence of Pmr1. Together, our results establish that Pmr1-dependent Ca2+ and/or Mn2+ ion homeostasis is necessary for TOR signaling.


Subject(s)
Calcium-Transporting ATPases/metabolism , Golgi Apparatus/enzymology , Molecular Chaperones/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae , Signal Transduction/physiology , Amino Acid Transport Systems/genetics , Amino Acid Transport Systems/metabolism , Antifungal Agents/metabolism , Calcium-Transporting ATPases/genetics , Epistasis, Genetic , Gene Expression Regulation, Fungal , Molecular Chaperones/genetics , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/genetics , Sirolimus/metabolism , Transcription Factors/metabolism
15.
Mol Cell Biol ; 26(22): 8639-54, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16954377

ABSTRACT

MAK (male germ cell-associated protein kinase) and MRK/ICK (MAK-related kinase/intestinal cell kinase) are human homologs of Ime2p in Saccharomyces cerevisiae and of Mde3 and Pit1 in Schizosaccharomyces pombe and are similar to human cyclin-dependent kinase 2 (CDK2) and extracellular signal-regulated kinase 2 (ERK2). MAK and MRK require dual phosphorylation in a TDY motif catalyzed by an unidentified human threonine kinase and tyrosine autophosphorylation. Herein, we establish that human CDK-related kinase CCRK (cell cycle-related kinase) is an activating T157 kinase for MRK, whereas active CDK7/cyclin H/MAT1 complexes phosphorylate CDK2 but not MRK. Protein phosphatase 5 (PP5) interacts with MRK in a complex and dephosphorylates MRK at T157 in vitro and in situ. Thus, CCRK and PP5 are yin-yang regulators of T157 phosphorylation. To determine a substrate consensus, we screened a combinatorial peptide library with active MRK. MRK preferentially phosphorylates R-P-X-S/T-P sites, with the preference for arginine at position -3 (P-3) being more stringent than for prolines at P-2 and P+1. Using the consensus, we identified a putative phosphorylation site (RPLT(1080)S) for MRK in human Scythe, an antiapoptotic protein that interacts with MRK. MRK phosphorylates Scythe at T1080 in vitro as determined by site-directed mutagenesis and mass spectrometry, supporting the consensus and suggesting Scythe as a physiological substrate for MRK.


Subject(s)
Consensus Sequence , Cyclin-Dependent Kinases/metabolism , Gene Expression Regulation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Cell Line , Down-Regulation , Humans , Hydrogen Peroxide/pharmacology , Molecular Chaperones , Molecular Sequence Data , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Structure, Tertiary , Proteins/metabolism , Transfection , Cyclin-Dependent Kinase-Activating Kinase
16.
Arch Biochem Biophys ; 449(1-2): 8-16, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16626623

ABSTRACT

We showed previously that p90 RSK was activated in cells expressing an activated mutant of MEK5, the activator of the MAP kinase ERK5. Based on the following evidence, we suggest that ERK5 can directly activate RSK in cells. ERK5 binds to RSK in vitro and co-immunoprecipitates from cell extracts; activation of ERK5 weakens its binding to RSK, suggesting that RSK is released upon activation. Phosphorylation of RSK by ERK5 in vitro causes its activation, indicating that RSK is a substrate of ERK5. In cells activation of ERK5 but not p38 or the c-Jun N-terminal kinase is associated with RSK activation. The large C-terminal domain of ERK5 is not required for binding or activation of RSK by ERK5; however, the common docking or CD domain of ERK5 and the docking or D domain of RSK are important for their association.


Subject(s)
Mitogen-Activated Protein Kinase 7/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Extracellular Signal-Regulated MAP Kinases/metabolism , HeLa Cells , Humans , Phosphorylation , Protein Binding
17.
Mol Cell Biol ; 25(14): 6047-64, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15988018

ABSTRACT

Male germ cell-associated kinase (MAK) and intestinal cell kinase (ICK) are nuclear Cdc2-related kinases with nearly identical N-terminal catalytic domains and more divergent C-terminal noncatalytic domains. The catalytic domain is also related to mitogen-activated protein kinases (MAPKs) and contains a corresponding TDY motif. Nuclear localization of ICK requires subdomain XI and interactions of the conserved Arg-272, but not kinase activity or, surprisingly, any of the noncatalytic domain. Further, nuclear localization of ICK is required for its activation. ICK is activated by dual phosphorylation of the TDY motif. Phosphorylation of Tyr-159 in the TDY motif requires ICK autokinase activity but confers only basal kinase activity. Full activation requires additional phosphorylation of Thr-157 in the TDY motif. Coexpression of ICK with constitutively active MEK1 or MEK5 fails to increase ICK phosphorylation or activity, suggesting that MEKs are not involved. ICK and MAK are related to Ime2p in budding yeast, and cyclin-dependent protein kinase-activating kinase Cak1p has been placed genetically upstream of Ime2p. Recombinant Cak1p phosphorylates Thr-157 in the TDY motif of recombinant ICK and activates its activity in vitro. Coexpression of ICK with wild-type CAK1 but not kinase-inactive CAK1 in cells also increases ICK phosphorylation and activity. Our studies establish ICK as the prototype for a new group of MAPK-like kinases requiring dual phosphorylation at TDY motifs.


Subject(s)
Cell Nucleus/enzymology , Cyclin-Dependent Kinases/physiology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , CDC2-CDC28 Kinases/genetics , CDC2-CDC28 Kinases/metabolism , Catalytic Domain , Cell Nucleus/chemistry , Cells, Cultured , Enzyme Activation , Humans , Mice , Molecular Sequence Data , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Rats , Cyclin-Dependent Kinase-Activating Kinase
18.
J Biol Chem ; 279(30): 31930-6, 2004 Jul 23.
Article in English | MEDLINE | ID: mdl-15145956

ABSTRACT

We have shown that chronic elevated glucose (25 mm) increases monocyte adhesion to human aortic endothelial cells (EC). This increased adhesion is mediated primarily through induction of interleukin (IL)-8 via activation of the transcription factor AP-1 (Srinivasan, S., Yeh, M., Danziger, E. C., Hatley, M. E., Riggan, A. E., Leitinger, N., Berliner, J. A., and Hedrick, C. C. (2003) Circ. Res. 92, 371-377). In the current study, we identified the elements in the AP-1 transcriptional complex that are activated by glucose. These elements include c-Jun, c-Fos, and Fra-1. AP-1 is activated by cellular oxidative stress, and we have reported significant production of ROS by high glucose-cultured cells. We examined signaling pathways upstream of AP-1 in EC that lead to AP-1 activation by HG. EC cultured in 25 mm glucose had a 2-fold increase in p38 phosphorylation compared with control normal glucose-cultured EC. Inhibition of the p38 pathway using 5 microm SB203580 significantly reduced glucose-mediated IL-8 mRNA production by 60%. Furthermore, blocking p38 pathway activation using a dominant-negative p38 construct significantly reduced glucose-mediated monocyte adhesion by 50%. Thus, glucose-stimulated monocyte adhesion is primarily regulated through phosphorylation of p38 with subsequent activation of AP-1, leading to IL-8 production. To study this pathway in the setting of diabetes, we used the db/db mouse. P38 phosphorylation was increased in diabetic db/db mice compared with control mice. We found a dramatic elevation in plasma levels of KC, the mouse ortholog of IL-8 in diabetic db/db mice (1800 +/- 100 pg/ml KC in db/db versus 300 +/- 75 pg/ml in C57BL/6J control mice, p < 0.0001). Inhibition of the p38 pathway in diabetic db/db mice significantly reduced monocyte adhesion by 50%. Taken together, these data indicate that chronic elevated glucose in diabetes activates the p38 MAP kinase pathway to increase inflammatory IL-8 gene induction and monocyte/endothelial adhesion.


Subject(s)
Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/immunology , Endothelium, Vascular/enzymology , Endothelium, Vascular/immunology , Glucose/pharmacology , Interleukin-8/biosynthesis , Mitogen-Activated Protein Kinases/metabolism , Animals , Base Sequence , Cell Adhesion/drug effects , Cells, Cultured , DNA Primers/genetics , Diabetes Mellitus, Type 2/genetics , Endothelium, Vascular/drug effects , Humans , In Vitro Techniques , Interleukin-8/genetics , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Models, Biological , Monocytes/drug effects , Monocytes/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , p38 Mitogen-Activated Protein Kinases
19.
J Biol Chem ; 279(11): 10176-84, 2004 Mar 12.
Article in English | MEDLINE | ID: mdl-14688255

ABSTRACT

MAPKAP kinase 2 (MK2) is required for tumor necrosis factor synthesis. Tristetraprolin (TTP) binds to the 3'-untranslated region of tumor necrosis factor mRNA and regulates its fate. We identified in vitro and in vivo phosphorylation sites in TTP using nanoflow high pressure liquid chromatography microelectrospray ionization tandem mass spectrometry and novel methods for direct digestion of TTP bound to affinity matrices (GSH-beads or anti-Myc linked to magnetic beads). MK2Delta3B, activated in Escherichia coli by p38alpha, phosphorylates TTP in vitro at major sites Ser(52) and Ser(178) (>10-fold in abundance) as well as at several minor sites that were detected after enriching for phosphopeptides with immobilized metal affinity chromatography. MK2 phosphorylation of TTP creates a functional 14-3-3 binding site. In cells, TTP was phosphorylated at Ser(52), Ser(178), Thr(250), and Ser(316) and at SP sites in a cluster (Ser(80)/Ser(82)/Ser(85)). Anisomycin treatment of NIH 3T3 cells increased phosphorylation of Ser(52) and Ser(178). Overexpression of MK2 sufficed to increase phosphorylation of Ser(52) and Ser(178) but not Ser(80)/Ser(82)/Ser(85) or Thr(250). Thus, Ser(52) and Ser(178) are putative MK2 sites in vivo. Identified phosphosite(s) may be biologic switches controlling mRNA stability and translation.


Subject(s)
DNA-Binding Proteins , Immediate-Early Proteins/metabolism , Protein Serine-Threonine Kinases/physiology , Serine/chemistry , Tyrosine 3-Monooxygenase/chemistry , 14-3-3 Proteins , 3' Untranslated Regions , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Chromatography , Chromatography, High Pressure Liquid , Cricetinae , Dose-Response Relationship, Drug , Glutathione/metabolism , Glutathione Transferase/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Kidney/cytology , Magnetics , Mice , Mitogen-Activated Protein Kinase 14 , Mitogen-Activated Protein Kinases/metabolism , Models, Chemical , Molecular Sequence Data , NIH 3T3 Cells , Peptides/chemistry , Phosphorylation , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Biosynthesis , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Recombinant Fusion Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization , Time Factors , Transfection , Tristetraprolin , Tyrosine 3-Monooxygenase/metabolism
20.
Brain Res ; 952(1): 98-110, 2002 Oct 11.
Article in English | MEDLINE | ID: mdl-12363409

ABSTRACT

The parkinsonian neurotoxin methylpyridinium (MPP(+)) mimics the neuropathology of Parkinson's disease (PD) and likely kills neurons by inhibiting complex I of the electron transport chain and increasing oxidative stress. We examined the time course of activation/inactivation of multiple pro- and anti-apoptotic signaling pathways in MPP(+)-induced apoptotic death of SH-SY5Y neuroblastoma cells. We found an early increase and later decrease of transcriptional activity of the generally anti-apoptotic nuclear factor kappa-beta (NF-kappa B) and early increases in activating phosphorylation of the anti-apoptotic upstream kinase protein kinase B (PKB, also known as AKT). Sequestration-inducing phosphorylation of pro-apoptotic BAD protein increased early then declined. A small biphasic increase in the generally pro-apoptotic p38 kinase activity paralleled the biphasic rise in NF-kappa B-mediated transcription. Inhibition of p38 kinase with 5 micro M SB203540, inhibition of MEK-ERK with 50 micro M U0126, or inhibition of phosphatidylinositol-3-kinase (PI3K) with 10 micro M LY294002 reduced cell viability by 4, 18 or 37%, respectively, after 24 h. All three kinase inhibitors increased cell death in response to 24 h of MPP(+), with the greatest effect shown by LY294002. Nerve growth factor (NGF) caused an early increase in activating phosphorylation of PKB/AKT and MEK-ERK and increased cell survival during MPP(+) exposure. We found that acute MPP(+) exposure activates multiple interacting death- and survival-promoting pathways. Survival-promoting MEK-ERK and PI3K pathways contribute to viability during MPP(+) exposure, both are activated by NGF, and loss of PI3K-mediated signaling and NF-kappa B-mediated transcription may commit cells irreversibly to apoptosis in this model. It remains unknown to what extent these signaling pathways modulate dopamine neuronal death in PD.


Subject(s)
1-Methyl-4-phenylpyridinium/pharmacology , Apoptosis/drug effects , MAP Kinase Kinase Kinase 1 , Nerve Growth Factor/pharmacology , Neuroblastoma , Neurons/cytology , Neurotoxins/pharmacology , Carrier Proteins/metabolism , Drug Interactions , Humans , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Neurons/drug effects , Neurons/enzymology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Tumor Cells, Cultured , bcl-Associated Death Protein , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...