Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
PLoS One ; 10(3): e0114363, 2015.
Article in English | MEDLINE | ID: mdl-25781882

ABSTRACT

Constitutive activation of the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) axis is fundamental to the molecular pathogenesis of a host of hematological disorders, including acute leukemias and myeloproliferative neoplasms (MPN). We demonstrate here that the major JAK2 mutation observed in these diseases (JAK2V617F) enforces Mcl-1 transcription via STAT3 signaling. Targeting this lesion with JAK inhibitor I (JAKi-I) attenuates STAT3 binding to the Mcl-1 promoter and suppresses Mcl-1 transcript and protein expression. The neutralization of Mcl-1 in JAK2V617F-harboring myelodyssplastic syndrome cell lines sensitizes them to apoptosis induced by the BH3-mimetic and Bcl-xL/Bcl-2 inhibitor, ABT-263. Moreover, simultaneously targeting JAK and Bcl-xL/-2 is synergistic in the presence of the JAK2V617F mutation. These findings suggest that JAK/Bcl-xL/-2 inhibitor combination therapy may have applicability in a range of hematological disorders characterized by activating JAK2 mutations.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Janus Kinase 2/antagonists & inhibitors , Leukemia, Myeloid, Acute/pathology , Mutation , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , bcl-X Protein/antagonists & inhibitors , Aniline Compounds/pharmacology , Cell Line, Tumor , Drug Synergism , Humans , Janus Kinase 2/genetics , Protein Kinase Inhibitors/pharmacology , Sulfonamides/pharmacology
2.
PLoS One ; 7(7): e36961, 2012.
Article in English | MEDLINE | ID: mdl-22859932

ABSTRACT

Three p160 family members, p/CIP, SRC1, and TIF2, have been identified as transcriptional coactivators for nuclear hormone receptors and other transcription factors in vitro. In a previous study, we reported initial characterization of the obesity-resistant phenotypes of p/CIP and SRC-1 double knockout (DKO) mice, which exhibit increased energy expenditure, and suggested that nuclear hormone receptor target genes were involved in these phenotypes. In this study, we demonstrate that p/CIP and SRC1 control insulin signaling in a cell-autonomous manner both in vitro and in vivo. Genetic deletion of p/CIP and SRC-1 increases glucose uptake and enhances insulin sensitivity in both regular chow- and high fat diet-fed DKO mice despite increased food intake. Interestingly, we discover that loss of p/CIP and SRC-1 results in resistance to age-related obesity and glucose intolerance. We show that expression levels of a key insulin signaling component, insulin receptor substrate 1 (IRS1), are significantly increased in two cell lines representing fat and muscle lineages with p/CIP and SRC-1 deletions and in white adipose tissue and skeletal muscle of DKO mice; this may account for increased glucose metabolism and insulin sensitivity. This is the first evidence that the p160 coactivators control insulin signaling and glucose metabolism through IRS1. Therefore, our studies indicate that p/CIP and SRC-1 are potential therapeutic targets not only for obesity but also for diabetes.


Subject(s)
Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance , Nuclear Receptor Coactivator 1/physiology , Nuclear Receptor Coactivator 3/physiology , Obesity/metabolism , Adiponectin/blood , Adipose Tissue, White/metabolism , Animals , Blood Glucose , Diet, High-Fat/adverse effects , Disease Models, Animal , Gene Expression , Gene Knockdown Techniques , Glucose Tolerance Test , Insulin Receptor Substrate Proteins/genetics , Male , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , NIH 3T3 Cells , Nuclear Receptor Coactivator 1/genetics , Nuclear Receptor Coactivator 1/metabolism , Nuclear Receptor Coactivator 3/genetics , Nuclear Receptor Coactivator 3/metabolism , Obesity/blood , Obesity/etiology , RNA, Small Interfering/genetics , Signal Transduction
3.
Proc Natl Acad Sci U S A ; 107(28): 12634-9, 2010 Jul 13.
Article in English | MEDLINE | ID: mdl-20616035

ABSTRACT

Aurora kinase B inhibitors induce apoptosis secondary to polyploidization and have entered clinical trials as an emerging class of neocytotoxic chemotherapeutics. We demonstrate here that polyploidization neutralizes Mcl-1 function, rendering cancer cells exquisitely dependent on Bcl-XL/-2. This "addiction" can be exploited therapeutically by combining aurora kinase inhibitors and the orally bioavailable BH3 mimetic, ABT-263, which inhibits Bcl-XL, Bcl-2, and Bcl-w. The combination of ABT-263 with aurora B inhibitors produces a synergistic loss of viability in a range of cell lines of divergent tumor origin and exhibits more sustained tumor growth inhibition in vivo compared with aurora B inhibitor monotherapy. These data demonstrate that Bcl-XL/-2 is necessary to support viability during polyploidization in a variety of tumor models and represents a druggable molecular vulnerability with potential therapeutic utility.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Aniline Compounds , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Aurora Kinase B , Aurora Kinases , Enzyme Inhibitors/therapeutic use , Male , Mice , Neoplasms/genetics , Protein Serine-Threonine Kinases , Sulfonamides
4.
Mol Cell Biol ; 26(17): 6425-34, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16914728

ABSTRACT

The TSC1-TSC2/Rheb/Raptor-mTOR/S6K1 cell growth cassette has recently been shown to regulate cell autonomous insulin and insulin-like growth factor I (IGF-I) sensitivity by transducing a negative feedback signal that targets insulin receptor substrates 1 and 2 (IRS1 and -2). Using two cell culture models of the familial hamartoma syndrome, tuberous sclerosis, we show here that Raptor-mTOR and S6K1 are required for phosphorylation of IRS1 at a subset of serine residues frequently associated with insulin resistance, including S307, S312, S527, S616, and S636 (of human IRS1). Using loss- and gain-of-function S6K1 constructs, we demonstrate a requirement for the catalytic activity of S6K1 in both direct and indirect regulation of IRS1 serine phosphorylation. S6K1 phosphorylates IRS1 in vitro on multiple residues showing strong preference for RXRXXS/T over S/T,P sites. IRS1 is preferentially depleted from the high-speed pellet fraction in TSC1/2-deficient mouse embryo fibroblasts or in HEK293/293T cells overexpressing Rheb. These studies suggest that, through serine phosphorylation, Raptor-mTOR and S6K1 cell autonomously promote the depletion of IRS1 from specific intracellular pools in pathological states of insulin and IGF-I resistance and thus potentially in lesions associated with tuberous sclerosis.


Subject(s)
Phosphoproteins/metabolism , Phosphoserine/metabolism , Protein Kinases/metabolism , Proteins/metabolism , Ribosomal Protein S6 Kinases/metabolism , Tuberous Sclerosis/pathology , Adaptor Proteins, Signal Transducing , Animals , Cell Culture Techniques , Cells, Cultured , Humans , Insulin Receptor Substrate Proteins , Mice , Monomeric GTP-Binding Proteins/metabolism , Neuropeptides/metabolism , Phosphoproteins/deficiency , Ras Homolog Enriched in Brain Protein , Regulatory-Associated Protein of mTOR , Subcellular Fractions , TOR Serine-Threonine Kinases , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/deficiency
5.
Cell Cycle ; 4(1): 46-51, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15611656

ABSTRACT

The TSC1-TSC2 tumor suppressor complex serves as an interface between insulin and nutrient signaling pathways and the cell growth machinery. Recent work has indicated that the TSC1-TSC2 complex plays a role in the pathobiology of a number of tumor predisposition syndromes, including tuberous sclerosis (TSC1/2), Peutz-Jeghers syndrome (LKB1), and Cowden's syndrome (PTEN), in which the TSC/Rheb/mTOR axis is inappropriately active secondary to loss of tumor suppressor function. Recent work has demonstrated that TSC deficiency imposes a negative autoregulatory loop that suppresses insulin signaling at the post-receptor level, effectively resulting in cell autonomous insulin resistance. Exploitation of this insulin signaling deficiency may hold promise among tailored clinical therapies designed to manage tuberous sclerosis.


Subject(s)
Genetic Predisposition to Disease , Insulin Resistance/genetics , Receptors, Retinoic Acid/physiology , Receptors, Thyroid Hormone/physiology , Tuberous Sclerosis/genetics , Tumor Suppressor Proteins/physiology , Animals , Apoptosis/genetics , Apoptosis/physiology , Gene Expression Regulation , Hamartoma Syndrome, Multiple/genetics , Hamartoma Syndrome, Multiple/physiopathology , Humans , Insulin Receptor Substrate Proteins , Insulin Resistance/physiology , Nuclear Receptor Subfamily 1, Group F, Member 3 , Peutz-Jeghers Syndrome/genetics , Peutz-Jeghers Syndrome/physiopathology , Phosphoproteins/genetics , Phosphoproteins/physiology , Protein Processing, Post-Translational , Receptors, Retinoic Acid/genetics , Receptors, Thyroid Hormone/genetics , Transcription, Genetic , Tuberous Sclerosis/physiopathology , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics
6.
Curr Biol ; 14(18): 1650-6, 2004 Sep 21.
Article in English | MEDLINE | ID: mdl-15380067

ABSTRACT

Tuberous sclerosis is a largely benign tumor syndrome derived from the acquisition of somatic lesions in genes encoding the tumor suppressor products, TSC1 or TSC2. Loss of function of the TSC1-TSC2 complex, which acts as a Rheb GAP, yields constitutive, unrestrained signaling from the cell growth machinery comprised of Rheb, mTOR, and S6K. We demonstrate herein that constitutive activation of the Rheb/mTOR/S6K cassette, whether by genetic deletion of TSC1 or TSC2 or by ectopic expression of Rheb, is sufficient to induce insulin resistance. This is the result of downregulation of the insulin receptor substrates, IRS1 and IRS2, which become limiting for signal transmission from the insulin receptor to PI3K. Downstream of PI3K, the survival kinase, Akt, is completely refractory to activation by IRS-dependent growth factor pathways such as insulin or IGF-I in TSC1- or TSC2-deficient cells but not to activation by IRS-independent pathways such as those utilized by PDGF. The antiapoptotic program induced by IGF-I but not PDGF is severely compromised in TSC2 null cells. Our results suggest that inappropriate activation of the Rheb/mTOR/S6K pathway imposes a negative feedback program to attenuate IRS-dependent processes such as cell survival.


Subject(s)
Down-Regulation , Insulin Resistance/physiology , Phosphoproteins/metabolism , Signal Transduction , Tuberous Sclerosis/physiopathology , Blotting, Northern , Cell Survival/physiology , Cells, Cultured , Flow Cytometry , Humans , Immunoblotting , Immunoprecipitation , Insulin Receptor Substrate Proteins , Insulin-Like Growth Factor I/metabolism , Monomeric GTP-Binding Proteins/metabolism , Neuropeptides/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , Proteins/metabolism , Ras Homolog Enriched in Brain Protein , Repressor Proteins/metabolism , Ribosomal Protein S6 Kinases/metabolism , TOR Serine-Threonine Kinases , Tuberous Sclerosis/metabolism , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins
7.
J Biol Chem ; 279(20): 20816-23, 2004 May 14.
Article in English | MEDLINE | ID: mdl-14993219

ABSTRACT

The tuberous sclerosis gene products Tsc1 and Tsc2 behave as tumor suppressors by restricting cell growth, a function conserved among metazoans. Recent evidence has indicated that hyperactivation of S6 kinase 1 (S6K1) may represent an important biochemical change in the development of tuberous sclerosis-associated lesions. We show here that deletion of either Tsc1 or Tsc2 or expression of the Rheb (Ras homolog enriched in brain) GTPase leads to hyperphosphorylation of S6K1 at a subset of regulatory sites, particularly those of two essential residues functionally conserved among AGC superfamily serine/threonine kinases, i.e. the activation loop (T-loop; Thr-229) and the hydrophobic motif (H-motif; Thr-389). These sites are reciprocally and dose-dependently regulated when S6K1 is coexpressed with the Tsc1-Tsc2 complex. Mutations that render S6K1 mTOR (mammalian target of rapamycin)-resistant also protect S6K1 activity and phosphorylation from down-regulation by Tsc1/2. We demonstrate that two disease-associated mutations in Tsc2 fail to negatively regulate S6K1 activity concomitant with a failure to modify T-loop and H-motif phosphorylation. Finally, we identify one pathological Tsc2 mutation that retains its ability to negatively regulate S6K1, suggesting that, in some cases, tuberous sclerosis may develop independently of S6K1 hyperactivation. These results also highlight the importance of dual control of T-loop and H-motif phosphorylation of S6K1 by the Tsc1-Tsc2 complex.


Subject(s)
Chemokines, CC/metabolism , Gene Expression Regulation, Enzymologic/genetics , Repressor Proteins/metabolism , Ribosomal Protein S6 Kinases/metabolism , Amino Acid Sequence , Androstadienes/pharmacology , Animals , Cell Line , Chemokine CCL26 , Chemokines, CC/genetics , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Insulin/pharmacology , Mice , Peptide Fragments , Phosphorylation , Rabbits , Rats , Repressor Proteins/genetics , Ribosomal Protein S6 Kinases/antagonists & inhibitors , Ribosomal Protein S6 Kinases/genetics , Sirolimus/pharmacology , Transfection , Tuberous Sclerosis/genetics , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins , Wortmannin
8.
J Biol Chem ; 278(18): 16433-42, 2003 May 02.
Article in English | MEDLINE | ID: mdl-12586835

ABSTRACT

During mitosis, the cyclin-dependent kinase, Cdc2, signals the inactivation of major anabolic processes such as transcription, mRNA processing, translation, and ribosome biogenesis, thereby providing energy needed for the radical and energetically costly structural reorganization of the cell. This is accomplished by phosphorylation and inactivation of several key anabolic elements, including TFIIIB, TFIID, RNA polymerase II, poly(A) polymerase, and translation elongation factor 1gamma. We report here that ribosomal S6 kinase 1 (S6K1), a protein kinase linked to the translation of ribosomal protein mRNAs, is also subject to regulation by Cdc2 in mitosis. In mitotic HeLa cells, when the activity of Cdc2 is high, S6K1 is phosphorylated at multiple Ser/Thr, Pro (S/TP) sites, including Ser(371), Ser(411), Thr(421), and Ser(424). Concomitant with this, the phosphorylation of the hydrophobic motif site, Thr(389), is reduced resulting in a decrease in the specific activity of S6K1. The mitotic S/TP phosphorylation sites are readily phosphorylated by Cdc2.cyclin B in vitro. These proline-directed phosphorylations are sensitive to chemical inhibitors of Cdc2 but not to inhibitors of mammalian target of rapamycin, phosphatidylinositol 3-kinase, MEK1/2, or p38. In murine FT210 cells arrested in mitosis, conditional inactivation of Cdc2 reduces phosphorylation of S6K1 at S/TP sites while simultaneously increasing phosphorylation of Thr(389) and of the S6K1 substrate, RPS6. A physical interaction exists between Cdc2 and S6K1, and this interaction is enhanced in mitotic cells. These results suggest that Cdc2 provides a signal that triggers inactivation of S6K1 in mitosis, presumably serving to spare energy for costly mitotic processes at the expense of ribosomal protein synthesis.


Subject(s)
CDC2 Protein Kinase/physiology , Mitosis , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , G2 Phase , HeLa Cells , Humans , Phosphorylation , Proline , Ribosomal Protein S6 Kinases, 70-kDa/chemistry , Serine , Threonine
9.
Biochem J ; 366(Pt 1): 57-62, 2002 Aug 15.
Article in English | MEDLINE | ID: mdl-12014987

ABSTRACT

Considerable biochemical and pharmacological evidence suggests that the activation of ribosomal protein S6 kinases (S6Ks) by activated receptor tyrosine kinases involves multiple co-ordinated input signals. However, the identities of many of these inputs remain poorly described, and their precise involvement in S6K activation has been the subject of great investigative effort. In the present study, we have shown that 4-amino-5-(4-methylphenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP1), a selective inhibitor of the Src family of non-receptor tyrosine kinases, interferes with the activation of 70 and 85 kDa S6K gene products (p70S6K1 and p85S6K1) by insulin, insulin-like growth factor 1, sodium orthovanadate and activated alleles of phosphoinositide 3-kinase and H-Ras. PP1 also impedes the activation of AKT/protein kinase B and the extracellular signal-regulated protein kinases 1 and 2 by these various stimuli. Insulin-like growth factor 1 was observed to induce a sustained increase in c-Src autophosphorylation as revealed using anti-phospho-Y416 antisera, but this effect was absent from the cells treated with PP1. To conclude, an activated allele of p70S6K1 is compared with the wild-type allele, resistant to inhibition by PP1 when co-expressed with phosphoinositide-dependent kinase 1 (PDK1), suggesting that PP1 affects p70S6K1 via a PDK1-independent pathway. Thus activation of Src may supply a necessary signal for the activation of p70S6K1 and possibly other S6Ks.


Subject(s)
Pyrazoles/pharmacology , Pyrimidines/pharmacology , Ribosomal Protein S6 Kinases/metabolism , Alleles , Animals , Blotting, Western , CHO Cells , Cell Line , Cricetinae , Enzyme Activation , Enzyme Inhibitors/pharmacology , Fibroblasts/metabolism , Genes, Dominant , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Mice , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Signal Transduction , Time Factors , Vanadates/pharmacology , ras Proteins/metabolism
10.
J Biol Chem ; 277(4): 2525-33, 2002 Jan 25.
Article in English | MEDLINE | ID: mdl-11705993

ABSTRACT

Protein metabolism in eukaryotic organisms is defined by a synthesis-degradation equilibrium that is subject to regulation by hormonal and nutritional signals. In mammalian tissues such as skeletal muscle, glucocorticoid hormones specify a catabolic response that influences both protein synthetic and protein degradative pathways. With regard to the former, glucocorticoids attenuate mRNA translation at two levels: translational efficiency, i.e. translation initiation, and translational capacity, i.e. ribosome biogenesis. Glucocorticoids may impair translational capacity through the ribosomal S6 protein kinase (p70 S6K), a recognized glucocorticoid target and an effector of ribosomal protein synthesis. We demonstrate here that the reduction in growth factor-activated p70 S6K activity by glucocorticoids depends upon a functional glucocorticoid receptor (GR) and that the GR is both necessary and sufficient to render p70 S6K subject to glucocorticoid regulation. Furthermore, the DNA binding and transcriptional activation but not repression properties of the GR are indispensable for p70 S6K regulation. Finally, a mutational analysis of the p70 S6K carboxyl terminus indicates that this region confers glucocorticoid sensitivity, and thus glucocorticoids may facilitate autoinhibition of the enzyme ultimately reducing the efficiency with which T389 is phosphorylated.


Subject(s)
Receptors, Glucocorticoid/metabolism , Ribosomal Protein S6 Kinases/metabolism , Animals , Blotting, Western , CHO Cells , COS Cells , Cell Line , Cricetinae , DNA/metabolism , DNA Mutational Analysis , DNA, Complementary/metabolism , Dexamethasone/pharmacology , Epitopes , Glucocorticoids/metabolism , Humans , Mifepristone/pharmacology , Models, Biological , Phosphorylation , Protein Binding , Protein Biosynthesis , Protein Structure, Tertiary , RNA, Messenger/metabolism , Rats , Serine/chemistry , Threonine/chemistry , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...