Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 5: e1231, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24832603

ABSTRACT

Lymphangioleiomyomatosis (LAM) is a female-predominant interstitial lung disease that can lead to respiratory failure. LAM cells typically have inactivating TSC2 mutations, leading to mTORC1 activation. The gender specificity of LAM suggests that estradiol contributes to disease development, yet the underlying pathogenic mechanisms are not completely understood. Using metabolomic profiling, we identified an estradiol-enhanced pentose phosphate pathway signature in Tsc2-deficient cells. Estradiol increased levels of cellular NADPH, decreased levels of reactive oxygen species, and enhanced cell survival under oxidative stress. Mechanistically, estradiol reactivated Akt in TSC2-deficient cells in vitro and in vivo, induced membrane translocation of glucose transporters (GLUT1 or GLUT4), and increased glucose uptake in an Akt-dependent manner. (18)F-FDG-PET imaging demonstrated enhanced glucose uptake in xenograft tumors of Tsc2-deficient cells from estradiol-treated mice. Expression array study identified estradiol-enhanced transcript levels of glucose-6-phosphate dehydrogenase (G6PD), the rate-limiting enzyme of the pentose phosphate pathway. Consistent with this, G6PD was abundant in xenograft tumors and lung metastatic lesions of Tsc2-deficient cells from estradiol-treated mice. Molecular depletion of G6PD attenuated estradiol-enhanced survival in vitro, and treatment with 6-aminonicotinamide, a competitive inhibitor of G6PD, reduced lung colonization of Tsc2-deficient cells. Collectively, these data indicate that estradiol promotes glucose metabolism in mTORC1 hyperactive cells through the pentose phosphate pathway via Akt reactivation and G6PD upregulation, thereby enhancing cell survival under oxidative stress. Interestingly, a strong correlation between estrogen exposure and G6PD was also found in breast cancer cells. Targeting the pentose phosphate pathway may have therapeutic benefit for LAM and possibly other hormonally dependent neoplasms.


Subject(s)
Breast Neoplasms/enzymology , Estradiol/metabolism , Lymphangioleiomyomatosis/enzymology , Multiprotein Complexes/metabolism , Pentose Phosphate Pathway , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival , Drug Implants , Enzyme Activation , Estradiol/administration & dosage , Female , Gene Expression Profiling , Glucose/metabolism , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 2/metabolism , Glucosephosphate Dehydrogenase/genetics , Glucosephosphate Dehydrogenase/metabolism , Humans , Lymphangioleiomyomatosis/genetics , Lymphangioleiomyomatosis/pathology , Mechanistic Target of Rapamycin Complex 1 , Metabolomics , Mice , Mice, SCID , NADP/metabolism , Oxidative Stress , RNA Interference , Rats , Reactive Oxygen Species/metabolism , Signal Transduction , Time Factors , Transfection , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
2.
Oncogene ; 25(53): 7029-40, 2006 Nov 09.
Article in English | MEDLINE | ID: mdl-16715128

ABSTRACT

Rapamycin, an inhibitor of the mammalian target of rapamycin (mTOR), inhibits tumor cell motility. However, the underlying mechanism is poorly understood. Here, we show that rapamycin inhibited type I insulin-like growth factor (IGF-I)-stimulated motility of a panel of cell lines. Expression of a rapamycin-resistant mutant of mTOR (mTORrr) prevented rapamycin inhibition of cell motility. However, cells expressing a kinase-dead mTORrr remained sensitive to rapamycin. Downregulation of raptor or rictor by RNA interference (RNAi) decreased cell motility. However, only downregulation of raptor mimicked the effect of rapamycin, inhibiting phosphorylation of S6 kinase 1 (S6K1) and 4E-BP1. Cells infected with an adenovirus expressing constitutively active and rapamycin-resistant mutant of p70 S6K1, but not with an adenovirus expressing wild-type S6K1, or a control virus, conferred to resistance to rapamycin. Further, IGF-I failed to stimulate motility of the cells, in which S6K1 was downregulated by RNAi. Moreover, downregulation of eukaryotic initiation factor 4E (eIF4E)-binding protein 1 (4E-BP1) by RNAi-attenuated rapamycin inhibition of cell motility. In contrast, expression of constitutively active 4E-BP1 dramatically inhibited IGF-I-stimulated cell motility. The results indicate that both S6K1 and 4E-BP1 pathways, regulated by TORC1, are required for cell motility. Rapamycin inhibits IGF-I-stimulated cell motility, through suppression of both S6K1 and 4E-BP1/eIF4E-signaling pathways, as a consequence of inhibition of mTOR kinase activity.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Movement/drug effects , Phosphoproteins/metabolism , Protein Kinases/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , Animals , Cattle , Cell Cycle Proteins , Cell Line , Cytoprotection/drug effects , Down-Regulation , Enzyme Activation , Humans , Insulin-Like Growth Factor I/pharmacology , Mice , Phosphorylation , Protein Kinases/genetics , Serum , TOR Serine-Threonine Kinases , Transcription Factors/metabolism
3.
Mol Cell Biol ; 21(21): 7470-80, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11585927

ABSTRACT

RSK is a serine/threonine kinase containing two distinct catalytic domains. Found at the terminus of the Ras/extracellular signal-regulated kinase (ERK)-mitogen-activated protein kinase (MAPK) kinase cascade, mitogen-stimulated ribosomal S6 kinase (RSK) activity requires multiple inputs. These inputs include phosphorylation of the C-terminal kinase domain activation loop by ERK1/2 and phosphorylation of the N-terminal kinase domain activation loop by phosphoinositide-dependent protein kinase-1 (PDK1). Previous work has shown that upon mitogen stimulation, RSK accumulates in the nucleus. Here we show that prior to nuclear translocation, epidermal growth factor-stimulated RSK1 transiently associates with the plasma membrane. Myristylation of wild-type RSK1 results in an activated enzyme in the absence of added growth factors. When RSK is truncated at the C terminus, the characterized ERK docking is removed and RSK phosphotransferase activity is completely abolished. When myristylated, however, this myristylated C-terminal truncated form (myrCTT) is activated at a level equivalent to myristylated wild-type (myrWT) RSK. Both myrWT RSK and myrCTT RSK can signal to the RSK substrate c-Fos in the absence of mitogen activation. Unlike myrWT RSK, myrCTT RSK is not further activated by serum. Only the myristylated RSK proteins are basally phosphorylated on avian RSK1 serine 381, a site critical for RSK activity. The myristylated and unmyristylated RSK constructs interact with PDK1 upon mitogen stimulation, and this interaction is insensitive to the MEK inhibitor UO126. Because a kinase-inactive CTT RSK can be constitutively activated by targeting to the membrane, we propose that ERK may have a dual role in early RSK activation events: preliminary phosphorylation of RSK and escorting RSK to a membrane-associated complex, where additional MEK/ERK-independent activating inputs are encountered.


Subject(s)
Cell Membrane/enzymology , Ribosomal Protein S6 Kinases/chemistry , Ribosomal Protein S6 Kinases/metabolism , Blotting, Western , Butadienes/pharmacology , Cell Line , Cells, Cultured , Enzyme Activation , Enzyme Inhibitors/pharmacology , Glutathione Transferase/metabolism , Humans , Microscopy, Fluorescence , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Models, Genetic , Myristic Acids/metabolism , Nitriles/pharmacology , Phosphorylation , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Transfection
4.
J Biol Chem ; 276(49): 46639-46, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11583996

ABSTRACT

Caspase-8 is believed to play an obligatory role in apoptosis initiation by death receptors, but the role of its structural relative, caspase-10, remains controversial. Although earlier evidence implicated caspase-10 in apoptosis signaling by CD95L and Apo2L/TRAIL, recent studies indicated that these death receptor ligands recruit caspase-8 but not caspase-10 to their death-inducing signaling complex (DISC) even in presence of abundant caspase-10. We characterized a series of caspase-10-specific antibodies and found that certain commercially available antibodies cross-react with HSP60, shedding new light on previous results. The majority of 55 lung and breast carcinoma cell lines expressed mRNA for both caspase-8 and -10; however, immunoblot analysis revealed that caspase-10 protein expression was more frequently absent than that of caspase-8, suggesting a possible selective pressure against caspase-10 production in cancer cells. In nontransfected cells expressing both caspases, CD95L and Apo2L/TRAIL recruited endogenous caspase-10 as well as caspase-8 to their DISC, where both enzymes were proteolytically processed with similar kinetics. Caspase-10 recruitment required the adaptor FADD/Mort1, and caspase-10 cleavage in vitro required DISC assembly, consistent with the processing of an apoptosis initiator. Cells expressing only one of the caspases underwent ligand-induced apoptosis, indicating that each caspase can initiate apoptosis independently of the other. Thus, apoptosis signaling by death receptors involves not only caspase-8 but also caspase-10, and both caspases may have equally important roles in apoptosis initiation.


Subject(s)
Apoptosis , Caspases/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Amino Acid Sequence , Blotting, Western , Caspase 10 , Caspase 8 , Caspase 9 , Caspases/genetics , Caspases/immunology , Electrophoresis, Polyacrylamide Gel , Humans , Isoenzymes/immunology , Isoenzymes/metabolism , Molecular Sequence Data , Protein Biosynthesis , Tumor Cells, Cultured
6.
Proc Natl Acad Sci U S A ; 98(18): 10320-5, 2001 Aug 28.
Article in English | MEDLINE | ID: mdl-11504907

ABSTRACT

PTEN phosphatase acts as a tumor suppressor by negatively regulating the phosphoinositide 3-kinase (PI3K) signaling pathway. It is unclear which downstream components of this pathway are necessary for oncogenic transformation. In this report we show that transformed cells of PTEN(+/-) mice have elevated levels of phosphorylated Akt and activated p70/S6 kinase associated with an increase in proliferation. Pharmacological inactivation of mTOR/RAFT/FRAP reduced neoplastic proliferation, tumor size, and p70/S6 kinase activity, but did not affect the status of Akt. These data suggest that p70/S6K and possibly other targets of mTOR contribute significantly to tumor development and that inhibition of these proteins may be therapeutic for cancer patients with deranged PI3K signaling.


Subject(s)
Phosphoric Monoester Hydrolases/deficiency , Protein Kinase Inhibitors , Protein Kinases , Ribosomal Protein S6 Kinases/metabolism , Tumor Suppressor Proteins , Alleles , Animals , Base Sequence , Cell Transformation, Neoplastic/drug effects , DNA Primers/genetics , Female , Humans , Mice , Mice, Knockout , PTEN Phosphohydrolase , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/genetics , Signal Transduction , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Uterine Neoplasms/drug therapy , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology
7.
Cell Growth Differ ; 12(6): 297-306, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11432804

ABSTRACT

Oncogenic Ras induces cells to undergo apoptosis after inhibition of protein kinase C (PKC) activity. The integration of differential signaling pathways is required for full execution of apoptosis. In this study, we used Jurkat as well as Fas/FADD-defective cell lines expressing v-ras to determine the upstream elements required for activation of the caspase cascade in PKC/Ras-mediated apoptosis. During this Ras-induced apoptotic process, caspase-8 was activated, possibly through its binding to Fas-associated death domain (FADD), in Jurkat/ras and Jurkat/Fas(m)/ras cells but not in Jurkat/FADD(m)/ras cells. c-Jun NH(2)-terminal kinase (JNK) was activated in all three cell lines expressing ras in response to apoptotic stimulation. Suppression of JNK by dn-JNK1 blocked the interaction of FADD and caspase-8 and partially protected Jurkat/ras and Jurkat/Fas(m)/ras cells from apoptosis. However, dn-JNK1 had no effect on PKC/Ras-induced apoptosis in Jurkat/FADD(m)/ras cells. The results indicate that FADD/caspase-8 signaling is involved in PKC/Ras-mediated apoptosis, and JNK may be an upstream effector of caspase activation.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis , Carrier Proteins/metabolism , Caspases/metabolism , fas Receptor/metabolism , ras Proteins/metabolism , BH3 Interacting Domain Death Agonist Protein , Caspase 8 , Caspase 9 , Cytochrome c Group/metabolism , Enzyme Activation , Fas-Associated Death Domain Protein , Humans , JNK Mitogen-Activated Protein Kinases , Jurkat Cells , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction
8.
J Biol Chem ; 276(35): 32585-90, 2001 Aug 31.
Article in English | MEDLINE | ID: mdl-11384965

ABSTRACT

Fas, a death domain-containing member of the tumor necrosis factor receptor family and its ligand FasL have been predominantly studied with respect to their capability to induce cell death. However, a few studies indicate a proliferation-inducing signaling activity of these molecules too. We describe here a novel signaling pathway of FasL and the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) that triggers transcriptional activation of the proto-oncogene c-fos, a typical target gene of mitogenic pathways. FasL- and TRAIL-mediated up-regulation of c-Fos was completely dependent on the presence of Fas-associated death domain protein (FADD) and caspase-8, but caspase activity seemed to be dispensable as a pan inhibitor of caspases had no inhibitory effect. Upon overexpression of the long splice form of cellular FADD-like interleukin-1-converting enzyme (FLICE) inhibitory protein (cFLIP) in Jurkat cells, FasL- and TRAIL-induced up-regulation of c-Fos was almost completely blocked. The short splice form of FLIP, however, showed a rather stimulatory effect on c-Fos induction. Together these data demonstrate the existence of a death receptor-induced, FADD- and caspase-8-dependent pathway leading to c-Fos induction that is inhibited by the long splice form FLIP-L.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Caspases/metabolism , Gene Expression Regulation , Genes, fos , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/physiology , Tumor Necrosis Factor-alpha/physiology , fas Receptor/physiology , Alternative Splicing , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis Regulatory Proteins , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/genetics , Caspase 8 , Caspase 9 , Fas Ligand Protein , Fas-Associated Death Domain Protein , Humans , Jurkat Cells , Models, Biological , Proto-Oncogene Mas , Proto-Oncogene Proteins c-fos/genetics , Recombinant Proteins/metabolism , TNF-Related Apoptosis-Inducing Ligand , Transfection
9.
J Biol Chem ; 276(15): 12466-75, 2001 Apr 13.
Article in English | MEDLINE | ID: mdl-11278584

ABSTRACT

The anti-tumorigenic and anti-proliferative effects of N-alpha-tosyl-l-phenylalanyl chloromethyl ketone (TPCK) have been known for more than three decades. Yet little is known about the discrete cellular targets of TPCK controlling these effects. Previous work from our laboratory showed TPCK, like the immunosuppressant rapamycin, to be a potent inhibitor of the 70-kilodalton ribosomal S6 kinase 1 (S6K1), which mediates events involved in cell growth and proliferation. We show here that rapamycin and TPCK display distinct inhibitory mechanisms on S6K1 as a rapamycin-resistant form of S6K1 was TPCK-sensitive. Additionally, we show that TPCK inhibited the activation of the related kinase and proto-oncogene Akt. Upstream regulators of S6K1 and Akt include phosphoinositide 3-kinase (PI 3-K) and 3-phosphoinositide-dependent kinase 1 (PDK1). Whereas TPCK had no effect on either mitogen-regulated PI 3-K activity or total cellular PDK1 activity, TPCK prevented phosphorylation of the PDK1 regulatory sites in S6K1 and Akt. Furthermore, whereas both PDK1 and the mitogen-activated protein kinase (MAPK) are required for full activation of the 90-kilodalton ribosomal S6 kinase (RSK), TPCK inhibited RSK activation without inhibiting MAPK activation. Consistent with the capacity of RSK and Akt to mediate a cell survival signal, in part through phosphorylation of the pro-apoptotic protein BAD, TPCK reduced BAD phosphorylation and led to cell death in interleukin-3-dependent 32D cells. Finally, in agreement with results seen in embryonic stem cells lacking PDK1, protein kinase A activation was not inhibited by TPCK showing TPCK specificity for mitogen-regulated PDK1 signaling. TPCK inhibition of PDK1 signaling thus disables central kinase cascades governing diverse cellular processes including proliferation and survival and provides an explanation for its striking biological effects.


Subject(s)
Anticarcinogenic Agents/pharmacology , Cell Division/drug effects , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Tosylphenylalanyl Chloromethyl Ketone/pharmacology , 3-Phosphoinositide-Dependent Protein Kinases , 3T3 Cells , Animals , Cell Line, Transformed , Enzyme Activation , Humans , Mice , Phosphorylation , Proto-Oncogene Mas , Ribosomal Protein S6 Kinases/antagonists & inhibitors , Ribosomal Protein S6 Kinases/metabolism
10.
J Cell Biol ; 152(5): 959-70, 2001 Mar 05.
Article in English | MEDLINE | ID: mdl-11238452

ABSTRACT

The cargo that the molecular motor kinesin moves along microtubules has been elusive. We searched for binding partners of the COOH terminus of kinesin light chain, which contains tetratricopeptide repeat (TPR) motifs. Three proteins were found, the c-jun NH(2)-terminal kinase (JNK)-interacting proteins (JIPs) JIP-1, JIP-2, and JIP-3, which are scaffolding proteins for the JNK signaling pathway. Concentration of JIPs in nerve terminals requires kinesin, as evident from the analysis of JIP COOH-terminal mutants and dominant negative kinesin constructs. Coprecipitation experiments suggest that kinesin carries the JIP scaffolds preloaded with cytoplasmic (dual leucine zipper-bearing kinase) and transmembrane signaling molecules (the Reelin receptor, ApoER2). These results demonstrate a direct interaction between conventional kinesin and a cargo, indicate that motor proteins are linked to their membranous cargo via scaffolding proteins, and support a role for motor proteins in spatial regulation of signal transduction pathways.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Kinesins/metabolism , Signal Transduction , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , Kinesins/chemistry , Kinesins/genetics , LDL-Receptor Related Proteins , MAP Kinase Kinase Kinases/metabolism , Mice , Models, Biological , Molecular Motor Proteins/chemistry , Molecular Motor Proteins/genetics , Molecular Motor Proteins/metabolism , Molecular Sequence Data , Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Precipitin Tests , Protein Binding , Rats , Receptors, Lipoprotein/metabolism , Reelin Protein , Sequence Alignment , Two-Hybrid System Techniques
11.
J Biol Chem ; 276(11): 7884-91, 2001 Mar 16.
Article in English | MEDLINE | ID: mdl-11108711

ABSTRACT

Ribosomal S6 kinase (S6K1), through phosphorylation of the 40 S ribosomal protein S6 and regulation of 5'-terminal oligopyrimidine tract mRNAs, is an important regulator of cellular translational capacity. S6K1 has also been implicated in regulation of cell size. We have recently identified S6K2, a homolog of S6K1, which phosphorylates S6 in vitro and is regulated by the phosphatidylinositide 3-kinase (PI3-K) and mammalian target of rapamycin pathways in vivo. Here, we characterize S6K2 regulation by PI3-K signaling intermediates and compare its regulation to that of S6K1. We report that S6K2 is activated similarly to S6K1 by the PI3-K effectors phosphoinositide-dependent kinase 1, Cdc42, Rac, and protein kinase Czeta but that S6K2 is more sensitive to basal activation by myristoylated protein kinase Czeta than is S6K1. The C-terminal sequence of S6K2 is divergent from that of S6K1. We find that the S6K2 C terminus plays a greater role in S6K2 regulation than does the S6K1 C terminus by functioning as a potent inhibitor of activation by various agonists. Removal of the S6K2 C terminus results in an enzyme that is hypersensitive to agonist-dependent activation. These data suggest that S6K1 and S6K2 are similarly activated by PI3-K effectors but that sequences unique to S6K2 contribute to stronger inhibition of its kinase activity. Understanding the regulation of the two S6K homologs may provide insight into the physiological roles of these kinases.


Subject(s)
Isoenzymes/metabolism , Phosphatidylinositol 3-Kinases/physiology , Ribosomal Protein S6 Kinases/metabolism , Cells, Cultured , Humans , Protein Kinase C/physiology , cdc42 GTP-Binding Protein/physiology
12.
J Biol Chem ; 276(11): 7892-8, 2001 Mar 16.
Article in English | MEDLINE | ID: mdl-11108720

ABSTRACT

Ribosomal S6 kinase 2 (S6K2) is a recently identified serine/threonine protein kinase that phosphorylates the 40 S ribosomal protein S6 in vitro. S6K2 is highly homologous to S6K1 in the core kinase and linker regulatory domains but differs from S6K1 in the N- and C-terminal regions and is differently localized primarily to the nucleus because of a C-terminal nuclear localization signal unique to S6K2. We have recently demonstrated that S6K2 is regulated similarly to S6K1 by the mammalian target of rapamycin pathway and by multiple PI3-K pathway effectors in vivo. However, deletion of the C-terminal domain of S6K2 enhances kinase activity, whereas analogous deletion of S6K1 is inhibitory. Here, we characterize the S6K2 C-terminal motifs that confer this differential regulation. We demonstrate that the inhibitory effects of the S6K2 C-terminal domain are only partly attributable to the nuclear localization signal but that three C-terminal proline-directed potential mitogen-activated protein kinase phosphorylation sites are critical mediators of this inhibitory effect. Site-specific mutation of these sites to alanine completely desensitizes S6K2 to activating inputs, whereas mutation to aspartic acid to mimic phosphorylation results in an activated enzyme which is hypersensitive to activating inputs. Pretreatment of cells with the mitogen-activated protein-extracellular signal-regulated kinase kinase (MEK) inhibitor U0126 inhibited S6K2 activation to a greater extent than S6K1. Furthermore, S6K2 mutants with C-terminal deletion or acidic phosphorylation site mutations displayed greatly reduced U0126 sensitivity. Thus, MEK-dependent inputs to C-terminal phosphorylation sites appear to be essential for relief of S6K2 inhibition but less critical for activation of S6K1. These data suggest a mechanism by which weak PI3-K agonists can regulate S6 phosphorylation and selective translation in the presence of mitogen-activated protein kinase signaling.


Subject(s)
Isoenzymes/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/physiology , Ribosomal Protein S6 Kinases/antagonists & inhibitors , Butadienes/pharmacology , Cells, Cultured , Enzyme Activation , Epidermal Growth Factor/pharmacology , Humans , Isoenzymes/metabolism , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/physiology , Nitriles/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Ribosomal Protein S6 Kinases/metabolism
13.
J Biol Chem ; 275(49): 38905-11, 2000 Dec 08.
Article in English | MEDLINE | ID: mdl-10988287

ABSTRACT

p53's dual regulation of arrest versus apoptosis may underlie tumor-selective effects of anti-cancer therapy. p53's apoptotic effect has been suggested to involve both transcription-dependent and -independent mechanisms. It is shown here that caspase-8 is activated early in cells undergoing p53-mediated apoptosis and in S100 cell-free extracts that recapitulate transcription-independent apoptosis. Depletion or inactivation of caspase-8 either in cells or cell-free extracts completely prevents this transcription-independent apoptosis and significantly attenuates overall death induced by wild-type p53. Importantly, caspase-8 activation appears to be independent of FADD, and caspase-8 is found in a novel 600-kDa complex following p53 activation. These findings highlight the roles of both transcription-dependent and -independent apoptosis by p53 and identify an essential role for caspase-8 in the transcription-independent pathway.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis/physiology , Caspases/metabolism , Transcription, Genetic/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Carrier Proteins/metabolism , Caspase 8 , Caspase 9 , Cell-Free System , Cells, Cultured , Fas-Associated Death Domain Protein , Fibroblasts/cytology , Fibroblasts/physiology , Genes, p53 , Mice , Tumor Suppressor Protein p53/genetics
14.
Immunity ; 12(6): 599-609, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10894160

ABSTRACT

Apoptosis induced by tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL/APO-2L) has been shown to exert important functions during various immunological processes. The involvement of the death adaptor proteins FADD/MORT1, TRADD, and RIP and the apoptosis-initiating caspases-8 and -10 in death signaling by the two death-inducing TRAIL receptors 1 and 2 (TRAIL-R1 and TRAIL-R2) are controversial. Analysis of the native TRAIL death-inducing signaling complex (DISC) revealed ligand-dependent recruitment of FADD/MORT1 and caspase-8. Differential precipitation of ligand-stimulated TRAIL receptors demonstrated that FADD/MORT1 and caspase-8 were recruited to TRAIL-R1 and TRAIL-R2 independently of each other. FADD/MORT1- and caspase-8-deficient Jurkat cells expressing only TRAIL-R2 were resistant to TRAIL-induced apoptosis. Thus, FADD/MORT1 and caspase-8 are essential for apoptosis induction via TRAIL-R2.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis/immunology , Carrier Proteins/physiology , Caspases/physiology , Receptors, Tumor Necrosis Factor/physiology , fas Receptor/physiology , B-Lymphocytes/cytology , B-Lymphocytes/enzymology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Carrier Proteins/metabolism , Caspase 8 , Caspase 9 , Caspases/metabolism , Cell Line , Fas-Associated Death Domain Protein , Humans , Jurkat Cells , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/biosynthesis , Signal Transduction/immunology , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Cells, Cultured
16.
Curr Biol ; 10(3): 127-35, 2000 Feb 10.
Article in English | MEDLINE | ID: mdl-10679322

ABSTRACT

BACKGROUND: Growth factors activate an array of cell survival signaling pathways. Mitogen-activated protein (MAP) kinases transduce signals emanating from their upstream activators MAP kinase kinases (MEKs). The MEK-MAP kinase signaling cassette is a key regulatory pathway promoting cell survival. The downstream effectors of the mammalian MEK-MAP kinase cell survival signal have not been previously described. RESULTS: We identify here a pro-survival role for the serine/threonine kinase Rsk1, a downstream target of the MEK-MAP kinase signaling pathway. In cells that are dependent on interleukin-3 (IL-3) for survival, pharmacological inhibition of MEKs antagonized the IL-3 survival signal. In the absence of IL-3, a kinase-dead Rsk1 mutant eliminated the survival effect afforded by activated MEK. Conversely, a novel constitutively active Rsk1 allele restored the MEK-MAP kinase survival signal. Experiments in vitro and in vivo demonstrated that Rsk1 directly phosphorylated the pro-apoptotic protein Bad at the serine residues that, when phosphorylated, abrogate Bad's pro-apoptotic function. Constitutively active Rsk1 caused constitutive Bad phosphorylation and protection from Bad-modulated cell death. Kinase-inactive Rsk1 mutants antagonize Bad phosphorylation. Bad mutations that prevented phosphorylation by Rsk1 also inhibited Rsk1-mediated cell survival. CONCLUSIONS: These data support a model in which Rsk1 transduces the mammalian MEK-MAP kinase signal in part by phosphorylating Bad.


Subject(s)
Apoptosis , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Ribosomal Protein S6 Kinases, 90-kDa , Ribosomal Protein S6 Kinases/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cell Survival , Humans , Interleukin-3/pharmacology , Interleukin-3/physiology , Kidney , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/genetics , Phosphorylation , Plasmids , Ribosomal Protein S6 Kinases/genetics , Signal Transduction , Transfection , bcl-Associated Death Protein
17.
Blood ; 95(8): 2552-8, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10753834

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF) activates several kinases and transcription factors through interaction with a heterodimeric receptor complex. We previously demonstrated that phosphorylation of the cyclic adenosine monophosphate (cAMP) response element-binding protein, CREB, occurs through a protein kinase A-independent pathway and is required for GM-CSF-induced transcriptional activation of the immediate early gene, early growth response-1 (egr-1). Recent reports indicate that receptor tyrosine kinases can induce CREB phosphorylation through activation of pp90RSK. We performed immune complex kinase assays in the human myeloid leukemic cell line, TF-1, which revealed that GM-CSF induced pp90RSK activation and phosphorylation of CREB within 5 minutes of stimulation. Transfection with the kinase-defective pp90RSK expression plasmid demonstrated a statistically significant decrease in transcriptional activation of a -116 CAT/egr-1 promoter construct in response to GM-CSF. Furthermore, activation of pp90RSK, CREB and egr-1 in GM-CSF-treated cells was inhibited by the presence of the inhibitor, PD98059. In this study, we report that GM-CSF induces CREB phosphorylation and egr-1 transcription by activating pp90RSK through an MEK-dependent signaling pathway. (Blood. 2000;95:2552-2558)


Subject(s)
Cyclic AMP Response Element-Binding Protein/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Ribosomal Protein S6 Kinases/physiology , Signal Transduction/drug effects , Enzyme Activation , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Humans , Phosphorylation , Transfection , Tumor Cells, Cultured
18.
Curr Biol ; 9(15): 810-20, 1999.
Article in English | MEDLINE | ID: mdl-10469565

ABSTRACT

BACKGROUND: The rsk1 gene encodes the 90 kDa ribosomal S6 kinase 1 (RSK1) protein, which contains two kinase domains. RSK1, which is involved in regulating cell survival and proliferation, lies at the end of the signaling cascade mediated by the extracellular signal-regulated kinase (ERK) subfamily of mitogen-activated protein (MAP) kinases. ERK activation and subsequent phosphorylation of the RSK1 carboxy-terminal catalytic loop stimulates phosphotransferase activity in the RSK1 amino-terminal kinase domain. When activated, RSK1 phosphorylates both nuclear and cytoplasmic substrates through this amino-terminal catalytic domain. It is thought that stimulation of the ERK/MAP kinase pathway is sufficient for RSK1 activation, but how ERK phosphorylation activates the RSK1 amino-terminal kinase domain is not known. RESULTS: The individual isolated RSK1 kinase domains were found to be under regulatory control. In vitro kinase assays established that ERK phosphorylates RSK1 within the carboxy-terminal kinase domain, and the phosphoinositide-dependent kinase 1 (PDK1) phosphorylates RSK1 within the amino-terminal kinase domain. In transiently transfected HEK 293E cells, PDK1 alone stimulated phosphotransferase activity of an isolated RSK1 amino-terminal kinase domain. Nevertheless, activation of full-length RSK1 in the absence of serum required activation by both PDK1 and ERK. CONCLUSIONS: RSK1 is phosphorylated by PDK1 in the amino-terminal kinase-activation loop, and by ERK in the carboxy-terminal kinase-activation loop. Activation of phosphotransferase activity of full-length RSK1 in vivo requires both PDK1 and ERK. RSK1 activation is therefore regulated by both the mitogen-stimulated ERK/MAP kinase pathway and a PDK1-dependent pathway.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Ribosomal Protein S6 Kinases/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Amino Acid Sequence , Binding Sites , Cell Line , Enzyme Activation , Humans , In Vitro Techniques , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ribosomal Protein S6 Kinases/chemistry , Ribosomal Protein S6 Kinases/genetics , Signal Transduction
19.
Oncogene ; 18(36): 5108-14, 1999 Sep 09.
Article in English | MEDLINE | ID: mdl-10490847

ABSTRACT

Rapamycin is an immunosuppressant which antagonizes cellular proliferation by inhibiting the function of mTOR. The mTOR:FKBP12: rapamycin complex blocks G1/S transition by inhibiting downstream targets essential for cell cycle progression. One such target is p70S6k1 (S6K1), a serine/threonine kinase which is inactivated by the mTOR : FKBP12 : rapamycin complex, and which has been linked to translational control by virtue of its ability to phosphorylate the ribosomal protein S6. In the current work, we describe cloning and characterization of a novel S6K1 homolog, p54 S6 kinase 2 (p54S6k2/S6K2). Similar to S6K1, S6K2 is activated by mitogens and by constitutively active PI3K, and is inhibited by rapamycin as well as wortmannin. Differences between activation of S6K1 and S6K2 by PDK1 were observed, suggesting potential differences in the regulation of these homologs. Strikingly, S6K2 activity and S6 phosphorylation were both intact in S6K1-/-ES cell, indicating a possible role for S6K2 in in vivo S6 phosphorylation. Interestingly, we found two isoforms of S6K2 which are localized to distinct cellular compartments; the smaller form resides in the detergent-soluble fraction, whereas the larger form is found in the particulate fraction. Our findings demonstrate the existence of a family of rapamycin-sensitive protein kinases potentially involved in S6 phosphorylation, translational control, and transduction of mTOR signals.


Subject(s)
Isoenzymes/genetics , Ribosomal Protein S6 Kinases/genetics , Sequence Homology, Nucleic Acid , Cell Line , Expressed Sequence Tags , Isoenzymes/metabolism , Molecular Sequence Data , Phosphorylation , Ribosomal Protein S6 Kinases/metabolism , Sirolimus/pharmacology
20.
Neuron ; 22(3): 623-33, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10197541

ABSTRACT

We show here that caspase-8 is required for the death of primary rat neurons induced by an expanded polyglutamine repeat (Q79). Expression of Q79 recruited and activated caspase-8. Inhibition of caspase-8 blocked polyglutamine-induced cell death. Coexpression of Q79 with the caspase inhibitor CrmA, a dominant-negative mutant of FADD (FADD DN), Bcl-2, or Bcl-xL, but not an N-terminally tagged Bcl-xL, prevented the recruitment of caspase-8 and inhibited polyglutamine-induced cell death. Furthermore, Western blot analysis revealed the presence of activated caspase-8 in the insoluble fraction of affected brain regions from Huntington's disease (HD) patients but not in those from neurologically unremarkable controls, suggesting the relocation and activation of caspase-8 during the pathogenesis of HD. These results suggest an essential role of caspase-8 in HD-related neural degenerative diseases.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis/physiology , Caspases/physiology , Neurons/physiology , Peptides/physiology , Viral Proteins , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/metabolism , Caspase 8 , Caspase 9 , Caspase Inhibitors , Caspases/genetics , Cell Line , Cysteine Proteinase Inhibitors/biosynthesis , Cysteine Proteinase Inhibitors/metabolism , Enzyme Activation , Fas-Associated Death Domain Protein , Fluorescent Antibody Technique, Direct , Humans , Immunoblotting , Microscopy, Confocal , Mutation , Neurons/enzymology , Plasmids , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Repetitive Sequences, Amino Acid , Serpins/biosynthesis , Serpins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...