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










Publication year range
1.
J Med Chem ; 65(14): 9662-9677, 2022 07 28.
Article in English | MEDLINE | ID: mdl-35838760

ABSTRACT

While epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) have changed the treatment landscape for EGFR mutant (L858R and ex19del)-driven non-small-cell lung cancer (NSCLC), most patients will eventually develop resistance to TKIs. In the case of first- and second-generation TKIs, up to 60% of patients will develop an EGFR T790M mutation, while third-generation irreversible TKIs, like osimertinib, lead to C797S as the primary on-target resistance mutation. The development of reversible inhibitors of these resistance mutants is often hampered by poor selectivity against wild-type EGFR, resulting in potentially dose-limiting toxicities and a sub-optimal profile for use in combinations. BLU-945 (compound 30) is a potent, reversible, wild-type-sparing inhibitor of EGFR+/T790M and EGFR+/T790M/C797S resistance mutants that maintains activity against the sensitizing mutations, especially L858R. Pre-clinical efficacy and safety studies supported progression of BLU-945 into clinical studies, and it is currently in phase 1/2 clinical trials for treatment-resistant EGFR-driven NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Drug Resistance, Neoplasm , ErbB Receptors , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
2.
J Am Soc Nephrol ; 33(6): 1087-1104, 2022 06.
Article in English | MEDLINE | ID: mdl-35236775

ABSTRACT

BACKGROUND: Upregulation of cAMP-dependent and cAMP-independent PKA signaling is thought to promote cystogenesis in polycystic kidney disease (PKD). PKA-I regulatory subunit RIα is increased in kidneys of orthologous mouse models. Kidney-specific knockout of RIα upregulates PKA activity, induces cystic disease in wild-type mice, and aggravates it in Pkd1RC/RC mice. METHODS: PKA-I activation or inhibition was compared with EPAC activation or PKA-II inhibition using Pkd1RC/RC metanephric organ cultures. The effect of constitutive PKA (preferentially PKA-I) downregulation in vivo was ascertained by kidney-specific expression of a dominant negative RIαB allele in Pkd1RC/RC mice obtained by crossing Prkar1αR1αB/WT, Pkd1RC/RC , and Pkhd1-Cre mice (C57BL/6 background). The effect of pharmacologic PKA inhibition using a novel, selective PRKACA inhibitor (BLU2864) was tested in mIMCD3 3D cultures, metanephric organ cultures, and Pkd1RC/RC mice on a C57BL/6 × 129S6/Sv F1 background. Mice were sacrificed at 16 weeks of age. RESULTS: PKA-I activation promoted and inhibition prevented ex vivo P-Ser133 CREB expression and cystogenesis. EPAC activation or PKA-II inhibition had no or only minor effects. BLU2864 inhibited in vitro mIMCD3 cystogenesis and ex vivo P-Ser133 CREB expression and cystogenesis. Genetic downregulation of PKA activity and BLU2864 directly and/or indirectly inhibited many pro-proliferative pathways and were both protective in vivo. BLU2864 had no detectable on- or off-target adverse effects. CONCLUSIONS: PKA-I is the main PKA isozyme promoting cystogenesis. Direct PKA inhibition may be an effective strategy to treat PKD and other conditions where PKA signaling is upregulated. By acting directly on PKA, the inhibition may be more effective than or substantially increase the efficacy of treatments that only affect PKA activity by lowering cAMP.


Subject(s)
Polycystic Kidney, Autosomal Dominant , Polycystic Kidney, Autosomal Recessive , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Down-Regulation , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Guanine Nucleotide Exchange Factors/pharmacology , Kidney/metabolism , Mice , Mice, Inbred C57BL , Polycystic Kidney Diseases , Polycystic Kidney, Autosomal Dominant/metabolism , Receptors, Cell Surface/genetics , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism
3.
Blood ; 125(2): 296-303, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25398940

ABSTRACT

Mutations of IDH1 and IDH2, which produce the oncometabolite 2-hydroxyglutarate (2HG), have been identified in several tumors, including acute myeloid leukemia. Recent studies have shown that expression of the IDH mutant enzymes results in high levels of 2HG and a block in cellular differentiation that can be reversed with IDH mutant-specific small-molecule inhibitors. To further understand the role of IDH mutations in cancer, we conducted mechanistic studies in the TF-1 IDH2 R140Q erythroleukemia model system and found that IDH2 mutant expression caused both histone and genomic DNA methylation changes that can be reversed when IDH2 mutant activity is inhibited. Specifically, histone hypermethylation is rapidly reversed within days, whereas reversal of DNA hypermethylation proceeds in a progressive manner over the course of weeks. We identified several gene signatures implicated in tumorigenesis of leukemia and lymphoma, indicating a selective modulation of relevant cancer genes by IDH mutations. As methylation of DNA and histones is closely linked to mRNA expression and differentiation, these results indicate that IDH2 mutant inhibition may function as a cancer therapy via histone and DNA demethylation at genes involved in differentiation and tumorigenesis.


Subject(s)
DNA Methylation/genetics , Enzyme Inhibitors/pharmacology , Histones/genetics , Isocitrate Dehydrogenase/genetics , Mutation , Transcriptome/drug effects , Cell Line, Tumor , Chromatin Immunoprecipitation , Chromatography, Liquid , Histones/drug effects , Humans , Leukemia, Myeloid, Acute/genetics , Phenylurea Compounds/pharmacology , Principal Component Analysis , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , Tandem Mass Spectrometry
4.
Nat Commun ; 5: 4846, 2014 Sep 10.
Article in English | MEDLINE | ID: mdl-25204415

ABSTRACT

Human cancer genomes harbour a variety of alterations leading to the deregulation of key pathways in tumour cells. The genomic characterization of tumours has uncovered numerous genes recurrently mutated, deleted or amplified, but gene fusions have not been characterized as extensively. Here we develop heuristics for reliably detecting gene fusion events in RNA-seq data and apply them to nearly 7,000 samples from The Cancer Genome Atlas. We thereby are able to discover several novel and recurrent fusions involving kinases. These findings have immediate clinical implications and expand the therapeutic options for cancer patients, as approved or exploratory drugs exist for many of these kinases.


Subject(s)
Gene Fusion/genetics , Neoplasms/genetics , Phosphotransferases/genetics , Gene Expression Profiling , Genome, Human , Humans , Molecular Targeted Therapy , Sequence Analysis, RNA
5.
Science ; 340(6132): 626-30, 2013 May 03.
Article in English | MEDLINE | ID: mdl-23558169

ABSTRACT

The recent discovery of mutations in metabolic enzymes has rekindled interest in harnessing the altered metabolism of cancer cells for cancer therapy. One potential drug target is isocitrate dehydrogenase 1 (IDH1), which is mutated in multiple human cancers. Here, we examine the role of mutant IDH1 in fully transformed cells with endogenous IDH1 mutations. A selective R132H-IDH1 inhibitor (AGI-5198) identified through a high-throughput screen blocked, in a dose-dependent manner, the ability of the mutant enzyme (mIDH1) to produce R-2-hydroxyglutarate (R-2HG). Under conditions of near-complete R-2HG inhibition, the mIDH1 inhibitor induced demethylation of histone H3K9me3 and expression of genes associated with gliogenic differentiation. Blockade of mIDH1 impaired the growth of IDH1-mutant--but not IDH1-wild-type--glioma cells without appreciable changes in genome-wide DNA methylation. These data suggest that mIDH1 may promote glioma growth through mechanisms beyond its well-characterized epigenetic effects.


Subject(s)
Benzeneacetamides/pharmacology , Cell Differentiation , Enzyme Inhibitors/pharmacology , Glioma/enzymology , Glioma/pathology , Imidazoles/pharmacology , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Animals , Benzeneacetamides/administration & dosage , Benzeneacetamides/toxicity , Cell Differentiation/drug effects , Cell Transformation, Neoplastic , Enzyme Inhibitors/toxicity , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Glioma/drug therapy , Glioma/genetics , Glutarates/metabolism , Histones/metabolism , Imidazoles/administration & dosage , Imidazoles/toxicity , Isocitrate Dehydrogenase/chemistry , Isocitrate Dehydrogenase/metabolism , Methylation , Mice , Mice, SCID , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Multimerization , RNA Interference , Xenograft Model Antitumor Assays
6.
Science ; 340(6132): 622-6, 2013 May 03.
Article in English | MEDLINE | ID: mdl-23558173

ABSTRACT

A number of human cancers harbor somatic point mutations in the genes encoding isocitrate dehydrogenases 1 and 2 (IDH1 and IDH2). These mutations alter residues in the enzyme active sites and confer a gain-of-function in cancer cells, resulting in the accumulation and secretion of the oncometabolite (R)-2-hydroxyglutarate (2HG). We developed a small molecule, AGI-6780, that potently and selectively inhibits the tumor-associated mutant IDH2/R140Q. A crystal structure of AGI-6780 complexed with IDH2/R140Q revealed that the inhibitor binds in an allosteric manner at the dimer interface. The results of steady-state enzymology analysis were consistent with allostery and slow-tight binding by AGI-6780. Treatment with AGI-6780 induced differentiation of TF-1 erythroleukemia and primary human acute myelogenous leukemia cells in vitro. These data provide proof-of-concept that inhibitors targeting mutant IDH2/R140Q could have potential applications as a differentiation therapy for cancer.


Subject(s)
Enzyme Inhibitors/pharmacology , Hematopoiesis/drug effects , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/enzymology , Phenylurea Compounds/pharmacology , Sulfonamides/pharmacology , Allosteric Site , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Catalytic Domain , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Erythropoiesis/drug effects , Gene Expression Regulation, Leukemic , Glutarates/metabolism , Humans , Isocitrate Dehydrogenase/chemistry , Isocitrate Dehydrogenase/metabolism , Leukemia, Erythroblastic, Acute , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Molecular Targeted Therapy , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Phenylurea Compounds/chemistry , Phenylurea Compounds/metabolism , Point Mutation , Protein Multimerization , Protein Structure, Secondary , Small Molecule Libraries , Sulfonamides/chemistry , Sulfonamides/metabolism
7.
Nature ; 488(7413): 656-9, 2012 Aug 30.
Article in English | MEDLINE | ID: mdl-22763442

ABSTRACT

Mutations in the IDH1 and IDH2 genes encoding isocitrate dehydrogenases are frequently found in human glioblastomas and cytogenetically normal acute myeloid leukaemias (AML). These alterations are gain-of-function mutations in that they drive the synthesis of the 'oncometabolite' R-2-hydroxyglutarate (2HG). It remains unclear how IDH1 and IDH2 mutations modify myeloid cell development and promote leukaemogenesis. Here we report the characterization of conditional knock-in (KI) mice in which the most common IDH1 mutation, IDH1(R132H), is inserted into the endogenous murine Idh1 locus and is expressed in all haematopoietic cells (Vav-KI mice) or specifically in cells of the myeloid lineage (LysM-KI mice). These mutants show increased numbers of early haematopoietic progenitors and develop splenomegaly and anaemia with extramedullary haematopoiesis, suggesting a dysfunctional bone marrow niche. Furthermore, LysM-KI cells have hypermethylated histones and changes to DNA methylation similar to those observed in human IDH1- or IDH2-mutant AML. To our knowledge, our study is the first to describe the generation and characterization of conditional IDH1(R132H)-KI mice, and also the first report to demonstrate the induction of a leukaemic DNA methylation signature in a mouse model. Our report thus sheds light on the mechanistic links between IDH1 mutation and human AML.


Subject(s)
Epigenesis, Genetic/genetics , Hematopoietic Stem Cells/cytology , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Mutant Proteins/metabolism , Mutation/genetics , Aging , Animals , Bone Marrow/pathology , Cell Lineage , CpG Islands/genetics , DNA Methylation , Disease Models, Animal , Female , Gene Knock-In Techniques , Glioma/pathology , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Histones/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Male , Mice , Mutant Proteins/genetics , Myeloid Cells/cytology , Myeloid Cells/metabolism , Spleen/pathology
8.
Proc Natl Acad Sci U S A ; 107(41): 17774-9, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20876099

ABSTRACT

The α-, ß-, and γ-protocadherins (Pcdhα, Pcdhß, and Pcdhγ) comprise a large family of single-pass transmembrane proteins predominantly expressed in the nervous system. These proteins contain six cadherin-like extracellular domains, and proteolysis of Pcdhα and Pcdhγ by the γ-secretase complex releases their intracellular domains into the cytoplasm where they may function locally and/or enter the nucleus and affect gene expression. Thus, cleavage of Pcdhs may function to link intercellular contacts and intracellular signaling. Here we report that shedding of the Pcdhα extracellular domain and subsequent processing by γ-secretase require endocytosis and that Pcdhs interact with the regulator of vesicular sorting ESCRT-0 in undifferentiated cells. We also find that the accumulation of Pcdh cleavage products is regulated during development. Differentiation leads to an increase in the interactions between Pcdh proteins and a decrease in the accumulation of cleavage products. We conclude that Pcdh processing requires endocytosis and that the level of cleavage products is regulated during neuronal differentiation.


Subject(s)
Cadherins/metabolism , Cell Differentiation/physiology , Endocytosis/physiology , Neurons/physiology , Peptide Hydrolases/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Blotting, Western , Cell Line , Endosomal Sorting Complexes Required for Transport/metabolism , Immunohistochemistry , Immunoprecipitation , Mice , Neurons/cytology , Plasmids/genetics , Protein Structure, Tertiary , Signal Transduction/physiology
9.
Proc Natl Acad Sci U S A ; 107(31): 13894-9, 2010 Aug 03.
Article in English | MEDLINE | ID: mdl-20616001

ABSTRACT

The clustered protocadherins (Pcdhs) are a large family of cadherin-like transmembrane proteins expressed in the nervous system. Stochastic expression of Pcdh genes and alternative splicing of their pre-mRNAs have the potential to generate enormous protein diversity at the cell surface of neurons. At present, the regulation and function of Pcdh proteins are largely unknown. Here, we show that Pcdhs form a heteromeric signaling complex(es), consisting of multiple Pcdh isoforms, receptor tyrosine kinases, phosphatases, and cell adhesion molecules. In particular, we find that the receptor tyrosine kinase rearranged during transformation (Ret) binds to Pcdhs in differentiated neuroblastoma cells and is required for stabilization and differentiation-induced phosphorylation of Pcdh proteins. In addition, the Ret ligand glial cell line-derived neurotrophic factor induces phosphorylation of Pcdhgamma in motor neurons and phosphorylation of Pcdhalpha and Pcdhgamma in sympathetic neurons. Conversely, Pcdh proteins are also required for the stabilization of activated Ret in neuroblastoma cells and sympathetic ganglia. Thus, Pcdhs and Ret are functional components of a phosphorylation-dependent signaling complex.


Subject(s)
Cadherins/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Animals , Cell Differentiation , Cells, Cultured , Chromatography, Affinity , Enzyme Activation , Enzyme Stability , Mice , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-ret/genetics , Signal Transduction
10.
J Biol Chem ; 280(12): 11101-6, 2005 Mar 25.
Article in English | MEDLINE | ID: mdl-15659381

ABSTRACT

The mammalian target of rapamycin, mTOR, is a Ser/Thr kinase that promotes cell growth and proliferation by activating ribosomal protein S6 kinase 1 (S6K1). We previously identified a conserved TOR signaling (TOS) motif in the N terminus of S6K1 that is required for its mTOR-dependent activation. Furthermore, our data suggested that the TOS motif suppresses an inhibitory function associated with the C terminus of S6K1. Here, we have characterized the mTOR-regulated inhibitory region within the C terminus. We have identified a conserved C-terminal "RSPRR" sequence that is responsible for an mTOR-dependent suppression of S6K1 activation. Deletion or mutations within this RSPRR motif partially rescue the kinase activity of the S6K1 TOS motif mutant (S6K1-F5A), and this rescued activity is rapamycin resistant. Furthermore, we have shown that the RSPRR motif significantly suppresses S6K1 phosphorylation at two phosphorylation sites (Thr-389 and Thr-229) that are crucial for S6K1 activation. Importantly, introducing both the Thr-389 phosphomimetic and RSPRR motif mutations into the catalytically inactive S6K1 mutant S6K1-F5A completely rescues its activity and renders it fully rapamycin resistant. These data show that the N-terminal TOS motif suppresses an inhibitory function mediated by the C-terminal RSPRR motif. We propose that the RSPRR motif interacts with a negative regulator of S6K1 that is normally suppressed by mTOR.


Subject(s)
Protein Kinases/physiology , Ribosomal Protein S6 Kinases, 70-kDa/chemistry , Sirolimus/pharmacology , Amino Acid Motifs , Amino Acid Sequence , Conserved Sequence , Humans , Molecular Sequence Data , Phosphorylation , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , Structure-Activity Relationship , TOR Serine-Threonine Kinases
11.
Semin Cell Dev Biol ; 15(2): 147-59, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15209374

ABSTRACT

Regulation of growth and proliferation in higher eukaryotic cells results from an integration of nutritional, energy, and mitogenic signals. Biochemical processes underlying cell growth and proliferation are governed by the phosphatidylinositol 3-kinase (PI3K) and target of rapamycin (TOR) signaling pathways. The importance of the interplay between these two pathways is underscored by the discovery that the TOR inhibitor rapamycin is effective against tumors caused by misregulation of the PI3K pathway. We review here recent data concerning the convergence of the PI3K and TOR pathways, the role of these pathways in cell growth and proliferation, and the regulation of growth by downstream TOR targets.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , Animals , Cell Division/drug effects
12.
Curr Biol ; 13(10): 797-806, 2003 May 13.
Article in English | MEDLINE | ID: mdl-12747827

ABSTRACT

BACKGROUND: The mammalian target of rapamycin, mTOR, is a serine/threonine kinase that controls cell growth and proliferation via the translation regulators eukaryotic initiation factor 4E (eIF4E) binding protein 1 (4E-BP1) and ribosomal protein S6 kinase 1 (S6K1). We recently identified a TOR signaling (TOS) motif in the N terminus of S6K1 and the C terminus of 4E-BP1 and demonstrated that in S6K1, the TOS motif is necessary to facilitate mTOR signaling to phosphorylate and activate S6K1. However, it is unclear how the TOS motif in S6K1 and 4E-BP1 mediates mTOR signaling. RESULTS: Here, we show that a functional TOS motif is required for 4E-BP1 to bind to raptor (a recently identified mTOR-interacting protein), for 4E-BP1 to be efficiently phosphorylated in vitro by the mTOR/raptor complex, and for 4E-BP1 to be phosphorylated in vivo at all identified mTOR-regulated sites. mTOR/raptor-regulated phosphorylation is necessary for 4E-BP's efficient release from the translational initiation factor eIF4E. Consistently, overexpression of a mutant of 4E-BP1 containing a single amino acid change in the TOS motif (F114A) reduces cell size, demonstrating that mTOR-dependent regulation of cell growth by 4E-BP1 is dependent on a functional TOS motif. CONCLUSIONS: Our data demonstrate that the TOS motif functions as a docking site for the mTOR/raptor complex, which is required for multisite phosphorylation of 4E-BP1, eIF4E release from 4E-BP1, and cell growth.


Subject(s)
Carrier Proteins/metabolism , Phosphoproteins/metabolism , Proteins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/chemistry , Adaptor Proteins, Signal Transducing , Amino Acid Motifs , Amino Acid Substitution , Binding Sites , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Cycle Proteins , Cell Line , Cell Line, Tumor , Cell Size , Eukaryotic Initiation Factor-4E/metabolism , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Phosphoproteins/chemistry , Phosphoproteins/genetics , Phosphorylation , Protein Kinases/metabolism , Proteins/chemistry , Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Regulatory-Associated Protein of mTOR , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction , TOR Serine-Threonine Kinases , Transfection
13.
Curr Biol ; 12(8): 632-9, 2002 Apr 16.
Article in English | MEDLINE | ID: mdl-11967149

ABSTRACT

BACKGROUND: The mammalian target of rapamycin (mTOR) controls the translation machinery via activation of S6 kinases 1 and 2 (S6K1/2) and inhibition of the eukaryotic initiation factor 4E (eIF4E) binding proteins 1, 2, and 3 (4E-BP1/2/3). S6K1 and 4E-BP1 are regulated by nutrient-sensing and mitogen-activated pathways. The molecular basis of mTOR regulation of S6K1 and 4E-BP1 remains controversial. RESULTS: We have identified a conserved TOR signaling (TOS) motif in the N terminus of all known S6 kinases and in the C terminus of the 4E-BPs that is crucial for phosphorylation and regulation S6K1 and 4E-BP1 activities. Deletion or mutations within the TOS motif significantly inhibit S6K1 activation and the phosphorylation of its hydrophobic motif, Thr389. In addition, this sequence is required to suppress an inhibitory activity mediated by the S6K1 C terminus. The TOS motif is essential for S6K1 activation by mTOR, as mutations in this motif mimic the effect of rapamycin on S6K1 phosphorylation, and render S6K1 insensitive to changes in amino acids. Furthermore, only overexpression of S6K1 with an intact TOS motif prevents 4E-BP1 phosphorylation by a common mTOR-regulated modulator of S6K1 and 4E-BP1. CONCLUSIONS: S6K1 and 4E-BP1 contain a conserved five amino acid sequence (TOS motif) that is crucial for their regulation by the mTOR pathway. mTOR seems to regulate S6K1 by two distinct mechanisms. The TOS motif appears to function as a docking site for either mTOR itself or a common upstream activator of S6K1 and 4E-BP1.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Conserved Sequence , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Protein Kinases/metabolism , Ribosomal Protein S6 Kinases/chemistry , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing , Amino Acid Motifs , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Cell Cycle Proteins , Cell Line , Enzyme Activation , Humans , Molecular Sequence Data , Mutation , Phosphoproteins/genetics , Phosphorylation , Phosphothreonine/metabolism , Protein Kinases/genetics , Ribosomal Protein S6 Kinases/genetics , Sequence Homology, Amino Acid , TOR Serine-Threonine Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...