Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 181
Filter
1.
Cell Death Differ ; 20(10): 1341-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23787999

ABSTRACT

The activation of the Akt signalling in response to cytokine receptor signalling promotes protein synthesis, cellular growth and proliferation. To determine the role of Akt in interleukin-3 (IL-3) signalling, we generated IL-3-dependent myeloid cell lines from mice lacking Akt1, Akt2 or Akt3. Akt1 deletion resulted in accelerated apoptosis at low concentrations of IL-3. Expression of constitutively active Akt1 was sufficient to delay apoptosis in response to IL-3 withdrawal, but not sufficient to induce proliferation in the absence of IL-3. Akt1 prolonged survival of Bim- or Bad-deficient cells, but not cells lacking Puma, indicating that Akt1-dependent repression of apoptosis was in part dependent on Puma and independent of Bim or Bad. Our data show that a key role of Akt1 during IL-3 signalling is to repress p53-dependent apoptosis pathways, including transcriptional upregulation of Puma. Moreover, our data indicate that regulation of BH3-only proteins by Akt is dispensable for Akt-dependent cell survival.


Subject(s)
Apoptosis/physiology , Cytokines/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Growth Processes/physiology , HEK293 Cells , Humans , Interleukin-3/metabolism , Isoenzymes , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/cytology , Myeloid Cells/enzymology , Receptors, Interleukin-3/metabolism , Signal Transduction
2.
Oncogene ; 32(7): 872-82, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-22469987

ABSTRACT

The infiltration of glioma cells into adjacent tissue is one of the major obstacles in the therapeutic management of malignant brain tumours, in most cases precluding complete surgical resection. Consequently, malignant glioma patients almost invariably experience tumour recurrences. Within the brain, glioma cells migrate rapidly either amoeboidly or mesenchymally to invade surrounding structures, in dependence on the extracellular environment. In addition, radiotherapy, frequently applied as adjuvant therapeutic modality, may enhance tumour cell mobility. Here, we show that the receptor tyrosine kinase Mer (MerTK) is overexpressed in glioblastoma multiforme (GBM) and that this is accompanied with increased invasive potential. MerTK expression is maintained in primary GBM-derived tumour spheres under stem cell culture conditions but diminishes significantly in serum-containing cultures with concomitant downregulation of Nestin and Sox2. Depletion of MerTK disrupts the rounded morphology of glioma cells and decreases their invasive capacity. Furthermore, the expression and phosphorylation of myosin light chain 2 are strongly associated with MerTK activity, indicating that the effect of MerTK on glioma cell invasion is mediated by actomyosin contractility. Finally, DNA damage robustly triggers the upregulation and phosphorylation of MerTK, which protects cells from apoptosis. This effect is strongly impaired upon MerTK depletion or overexpression of an inactive MerTK mutant. Collectively, our data suggests that MerTK is a novel therapeutic target in the treatment of the malignant gliomas.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioblastoma/genetics , Glioblastoma/pathology , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Cell Line, Tumor , Cell Shape/genetics , Cell Survival/genetics , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Neoplasm Invasiveness , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Up-Regulation/genetics , c-Mer Tyrosine Kinase
3.
Cell Death Dis ; 3: e325, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22717580

ABSTRACT

Recent studies identified a highly tumorigenic subpopulation of glioma stem cells (GSCs) within malignant gliomas. GSCs are proposed to originate from transformed neural stem cells (NSCs). Several pathways active in NSCs, including the Notch pathway, were shown to promote proliferation and tumorigenesis in GSCs. Notch2 is highly expressed in glioblastoma multiforme (GBM), a highly malignant astrocytoma. It is therefore conceivable that increased Notch2 signaling in NSCs contributes to the formation of GBM. Here, we demonstrate that mice constitutively expressing the activated intracellular domain of Notch2 in NSCs display a hyperplasia of the neurogenic niche and reduced neuronal lineage entry. Neurospheres derived from these mice show increased proliferation, survival and resistance to apoptosis. Moreover, they preferentially differentiate into astrocytes, which are the characteristic cellular population of astrocytoma. Likewise, we show that Notch2 signaling increases proliferation and resistance to apoptosis in human GBM cell lines. Gene expression profiling of GBM patient tumor samples reveals a positive correlation of Notch2 transcripts with gene transcripts controlling anti-apoptotic processes, stemness and astrocyte fate, and a negative correlation with gene transcripts controlling proapoptotic processes and oligodendrocyte fate. Our data show that Notch2 signaling in NSCs produces features of GSCs and induces astrocytic lineage entry, consistent with a possible role in astrocytoma formation.


Subject(s)
Astrocytes/metabolism , Cell Transformation, Neoplastic/pathology , Neural Stem Cells/metabolism , Receptor, Notch2/metabolism , Signal Transduction , Animals , Astrocytes/pathology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Lineage , Cell Transformation, Neoplastic/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Neural Stem Cells/pathology , Receptor, Notch2/genetics
4.
Oncogene ; 29(24): 3554-65, 2010 Jun 17.
Article in English | MEDLINE | ID: mdl-20400976

ABSTRACT

Protein kinase B (PKB/Akt) is ubiquitously expressed in cells. Phosphorylation of its multiple targets in response to various stimuli, including growth factors or cytokines, promotes cell survival and inhibits apoptosis. PKB is upregulated in many different cancers and a significant amount of the enzyme is present in its activated form. Here we show that PKB phosphorylates one of the anti-apoptotic proteins--transcription factor Twist-1 at Ser42. Cells expressing Twist-1 displayed inefficient p53 upregulation in response to DNA damage induced by gamma-irradiation or the genotoxic drug adriamycin. This influenced the activation of p53 target genes such as p21(Waf1) and Bax and led to aberrant cell-cycle regulation and the inhibition of apoptosis. The impaired induction of these p53 effector molecules is likely to be mediated by PKB-dependent phosphorylation of Twist-1 because, unlike the wild-type mutant, the Twist-1 S42A mutant did not confer cell resistance to DNA damage. Moreover, phosphorylation of Twist-1 at Ser42 was shown in vivo in various human cancer tissues, suggesting that this post-translational modification ensures functional activation of Twist-1 after promotion of survival during carcinogenesis.


Subject(s)
DNA Damage , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Serine , Tumor Suppressor Protein p53/antagonists & inhibitors , Twist-Related Protein 1/chemistry , Twist-Related Protein 1/metabolism , Amino Acid Sequence , Apoptosis/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , Down-Regulation , Doxorubicin/toxicity , Enzyme Activation , Gamma Rays , Humans , Molecular Sequence Data , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation , Protein Processing, Post-Translational , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism
5.
Oncogene ; 28(5): 698-708, 2009 Feb 05.
Article in English | MEDLINE | ID: mdl-19015637

ABSTRACT

The promyelocytic leukemia (PML) tumor suppressor protein, a central regulator of cell proliferation and apoptosis, is frequently fused to the retinoic acid receptor-alpha (RARalpha) in acute PML. Here we show the interaction of PML with another tumor suppressor protein, the serine/threonine kinase homeodomain-interacting protein kinase (HIPK2). In response to DNA damage, HIPK2 phosphorylates PML at serines 8 and 38. Although HIPK2-mediated phosphorylation of PML occurs early during the DNA damage response, the oncogenic PML-RARalpha fusion protein is phosphorylated with significantly delayed kinetics. DNA damage or HIPK2 expression leads to the stabilization of PML and PML-RARalpha proteins. The N-terminal phosphorylation sites contribute to the DNA damage-induced PML SUMOylation and are required for the ability of PML to cooperate with HIPK2 for the induction of cell death.


Subject(s)
Carrier Proteins/metabolism , DNA Damage/physiology , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Cell Death/physiology , Cells, Cultured , Genes, Tumor Suppressor , Humans , Nuclear Proteins/chemistry , Phosphorylation , Promyelocytic Leukemia Protein , Protein Binding , Protein Processing, Post-Translational , Protein Stability , Protein Structure, Tertiary , SUMO-1 Protein/metabolism , Serine/metabolism , Transcription Factors/chemistry , Tumor Suppressor Proteins/chemistry
6.
Biochem Soc Trans ; 35(Pt 5): 1013-7, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17956266

ABSTRACT

The PKB (protein kinase B) and PKC (protein kinase C) families display highly related catalytic domains that require a largely conserved series of phosphorylations for the expression of their optimum activities. However, in cells, the dynamics of these modifications are quite distinct. Based on experimental evidence, it is argued that the underlying mechanisms determining these divergent behaviours relate to the very different manner in which their variant regulatory domains interact with their respective catalytic domains. It is concluded that the distinct behaviours of PKB and PKC proteins are defined by the typical ground states of these proteins.


Subject(s)
Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Catalytic Domain , Enzyme Activation , Phosphorylation
7.
Biochem Soc Trans ; 35(Pt 2): 231-5, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17371246

ABSTRACT

PKB (protein kinase B, also known as Akt) is a serine/threonine protein kinase that is important in various signalling cascades and acts as a major signal transducer downstream of activated phosphoinositide 3-kinase. There are three closely related isoforms of PKB in mammalian cells, PKBalpha (Akt1), PKBbeta (Akt2) and PKBgamma (Akt3), and this review discusses recent advances in our understanding of the functions of these isoforms in the regulation of adipocyte differentiation, glucose homoeostasis and tumour development.


Subject(s)
Adipocytes/cytology , Aging/physiology , Cell Differentiation/physiology , Isoenzymes/physiology , Proto-Oncogene Proteins c-akt/physiology , Adipocytes/physiology , Animals , Enzyme Activation , Glucose/metabolism , Glucose Intolerance/genetics , Homeostasis , Insulin Resistance/genetics , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/deficiency , Proto-Oncogene Proteins c-akt/genetics
8.
Biochem Soc Trans ; 32(Pt 2): 350-4, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15046607

ABSTRACT

The genetic manipulation of mice has become an essential and elegant method for studying the function of proteins in physiology, and for testing the veracity of information obtained from cell culture experiments. During the past few years, a variety of transgenic and knockout mouse models of PKB (protein kinase B)/Akt have been generated and investigated. In this paper, we focus on the phenotypes of these PKB/Akt overexpression and mutant mice that may help to elucidate the functions exerted by PKB/Akt in mammals.


Subject(s)
Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Animals , Brain/pathology , Diabetes Mellitus/pathology , Disease Models, Animal , Female , Humans , Lymphoma/pathology , Male , Mammary Glands, Animal/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Milk/metabolism , Mutation , Myocardium/metabolism , Phenotype , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt , Thymus Neoplasms/pathology
9.
Trends Biochem Sci ; 26(11): 657-64, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11701324

ABSTRACT

It is ten years since the publication of three papers describing the cloning of a new proto-oncogene serine/threonine kinase termed protein kinase B (PKB)/Akt. Key roles for this protein kinase in cellular processes such as glucose metabolism, cell proliferation, apoptosis, transcription and cell migration are now well established. The explosion of publications involving PKB/Akt in the past three years emphasizes the high level of current interest in this signalling molecule. This review focuses on tracing the characterization of this kinase, through the elucidation of its mechanism of regulation, to its role in regulating physiological and pathophysiological processes, to our current understanding of the biology of PKB/Akt, and prospects for the future.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Signal Transduction , Amino Acid Sequence , Animals , Enzyme Activation , Gene Expression Regulation, Enzymologic , Humans , Lipid Metabolism , Models, Biological , Phosphorylation , Protein Structure, Tertiary , Proto-Oncogene Mas , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-akt
10.
Science ; 294(5541): 374-80, 2001 Oct 12.
Article in English | MEDLINE | ID: mdl-11598301

ABSTRACT

The PKB (protein kinase B, also called Akt) family of protein kinases plays a key role in insulin signaling, cellular survival, and transformation. PKB is activated by phosphorylation on residues threonine 308, by the protein kinase PDK1, and Serine 473, by a putative serine 473 kinase. Several protein binding partners for PKB have been identified. Here, we describe a protein partner for PKBalpha termed CTMP, or carboxyl-terminal modulator protein, that binds specifically to the carboxyl-terminal regulatory domain of PKBalpha at the plasma membrane. Binding of CTMP reduces the activity of PKBalpha by inhibiting phosphorylation on serine 473 and threonine 308. Moreover, CTMP expression reverts the phenotype of v-Akt-transformed cells examined under a number of criteria including cell morphology, growth rate, and in vivo tumorigenesis. These findings identify CTMP as a negative regulatory component of the pathway controlling PKB activity.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Cell Membrane/metabolism , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins , Retroviridae Proteins, Oncogenic/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Division , Cell Line , Cell Line, Transformed , Cell Size , Enzyme Activation , Genes, fos , Humans , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mice, Nude , Molecular Sequence Data , Neoplasms, Experimental/etiology , Oncogene Protein v-akt , Palmitoyl-CoA Hydrolase , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-akt , Recombinant Fusion Proteins/metabolism , Retroviridae Proteins, Oncogenic/genetics , Signal Transduction , Thiolester Hydrolases , Transcription, Genetic , Transfection , Tumor Cells, Cultured , Vanadates/pharmacology
11.
Cell Microbiol ; 3(8): 537-50, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11488815

ABSTRACT

The intracellular protozoan parasites Theileria parva and Theileria annulata transform leucocytes by interfering with host cell signal transduction pathways. They differ from tumour cells, however, in that the transformation process can be entirely reversed by elimination of the parasite from the host cell cytoplasm using a specific parasiticidal drug. We investigated the state of activation of Akt/PKB, a downstream target of PI3-K-generated phosphoinositides, in Theileria-transformed leucocytes. Akt/PKB is constitutively activated in a PI3-K- and parasite-dependent manner, as judged by the specific phosphorylation of key residues, in vitro kinase assays and its cellular distribution. In previous work, we demonstrated that the parasite induces constitutive activation of the transcription factor NF-kappaB, providing protection against spontaneous apoptosis that accompanies transformation. In a number of other systems, a link has been established between the PI3-K-Akt/PKB pathway and NF-kappaB activation, resulting in protection against apoptosis. In Theileria-transformed leucocytes, activation of the NF-kappaB and the PI3-K-Akt/PKB pathways are not directly linked. The PI3-K-Akt/PKB pathway does not contribute to the persistent induction of IkappaBalpha phosphorylation, NF-kappaB DNA-binding or transcriptional activity. We show that the two pathways are downregulated with different kinetics when the parasite is eliminated from the host cell cytoplasm and that NF-kappaB-dependent protection against apoptosis is not dependent on a functional PI3-K-Akt/PKB pathway. We also demonstrate that Akt/PKB contributes, at least in part, to the proliferation of Theileria-transformed T cells.


Subject(s)
Leukocytes/parasitology , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Theileria/pathogenicity , Animals , Apoptosis , Cattle , Cell Compartmentation , Lymphocyte Activation , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt , Signal Transduction , Theileria annulata/pathogenicity , Theileria parva/pathogenicity
12.
J Biol Chem ; 276(40): 37459-71, 2001 Oct 05.
Article in English | MEDLINE | ID: mdl-11481331

ABSTRACT

3-Phosphoinositide-dependent protein kinase-1 (PDK1) plays a central role in signal transduction pathways that activate phosphoinositide 3-kinase. Despite its key role as an upstream activator of enzymes such as protein kinase B and p70 ribosomal protein S6 kinase, the regulatory mechanisms controlling PDK1 activity are poorly understood. PDK1 has been reported to be constitutively active in resting cells and not further activated by growth factor stimulation (Casamayor, A., Morrice, N. A., and Alessi, D. R. (1999) Biochem. J. 342, 287-292). Here, we report that PDK1 becomes tyrosine-phosphorylated and translocates to the plasma membrane in response to pervanadate and insulin. Following pervanadate treatment, PDK1 kinase activity increased 1.5- to 3-fold whereas the activity of PDK1 associated with the plasma membrane increased approximately 6-fold. The activity of PDK1 localized to the plasma membrane was also increased by insulin treatment. Three tyrosine phosphorylation sites of PDK1 (Tyr-9 and Tyr-373/376) were identified using in vivo labeling and mass spectrometry. Using site-directed mutants, we show that, although phosphorylation on Tyr-373/376 is important for PDK1 activity, phosphorylation on Tyr-9 has no effect on the activity of the kinase. Both of these residues can be phosphorylated by v-Src tyrosine kinase in vitro, and co-expression of v-Src leads to tyrosine phosphorylation and activation of PDK1. Thus, these data suggest that PDK1 activity is regulated by reversible phosphorylation, possibly by a member of the Src kinase family.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Tyrosine/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Amino Acid Sequence , Biological Transport , Cell Membrane/drug effects , Cell Membrane/enzymology , Cell Membrane/metabolism , Cells, Cultured , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Molecular Sequence Data , Mutation , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Sequence Homology, Amino Acid , Tyrosine/genetics , Vanadates/pharmacology , src-Family Kinases/metabolism
13.
J Biol Chem ; 276(41): 38193-200, 2001 Oct 12.
Article in English | MEDLINE | ID: mdl-11473109

ABSTRACT

Hyperphosphorylated isoforms of the microtubule-associated protein tau are the major components of neurofibrillary lesions in Alzheimer's disease (AD). Protein phosphatase (PP) 2A is a major phosphatase implicated in tau dephosphorylation in vitro. Dephosphorylation of tau can be blocked in vivo by okadaic acid, a potent inhibitor of PP2A. Moreover, activity of PP2A is reduced in AD brains. To elucidate the role of PP2A in tau phosphorylation and pathogenesis, we expressed a dominant negative mutant form of the catalytic subunit Calpha of PP2A, L199P, in mice by using a neuron-specific promoter. We obtained mice with high expression levels of Calpha L199P in cortical, hippocampal, and cerebellar neurons. PP2A activity in brain homogenates of transgenic mice was reduced to 66%. Endogenous tau protein was hyperphosphorylated at distinct sites including the AT8 epitope Ser-202/Thr-205, a major AD-associated tau phosphoepitope. AT8-positive tau aggregates accumulated in the soma and dendrites of cortical pyramidal cells and cerebellar Purkinje cells and co-localized with ubiquitin. Our data establish that PP2A plays a crucial role in tau phosphorylation. Our results also show that reduced PP2A activity is associated with altered compartmentalization and ubiquitination of tau, resembling a key pathological finding in AD.


Subject(s)
Cell Compartmentation , Phosphoprotein Phosphatases/metabolism , tau Proteins/metabolism , Animals , Base Sequence , DNA Primers , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Neurons/enzymology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/genetics , Phosphorylation , Protein Phosphatase 2
14.
J Biol Chem ; 276(31): 29550-8, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11387345

ABSTRACT

We have reported previously the cloning and characterization of human and mouse protein kinase B gamma (PKB gamma), the third member of the PKB family of second messenger-regulated serine/threonine kinases (Brodbeck, D., Cron, P., and Hemmings, B. A. (1999) J. Biol. Chem. 274, 9133--9136). Here we report the isolation of human and mouse PKB gamma 1, a splice variant lacking the second regulatory phosphorylation site Ser-472 in the hydrophobic C-terminal domain. Expression of PKB gamma 1 is low compared with PKB gamma, and it is regulated in different human tissues. We show that PKB gamma and PKB gamma 1 differ in their response to stimulation by insulin, pervanadate, peroxide, or okadaic acid. Activation of PKB gamma 1 requires phosphorylation at a single regulatory site Thr-305. Interestingly, this site is phosphorylated to a higher extent in PKB gamma compared with PKB gamma 1 upon maximal stimulation by pervanadate, and this is reflected in the respective specific kinase activities. Furthermore, upon insulin stimulation of transfected cells, PKB gamma 1 translocates to the plasma membrane to a lesser extent than PKB gamma. Taken together, these results suggest that phosphorylation of the hydrophobic motif at the extreme C terminus of PKB gamma may facilitate translocation of the kinase to the membrane and/or its phosphorylation on the activation loop site by phosphoinositide-dependent protein kinase-1.


Subject(s)
Alternative Splicing , Brain/enzymology , Genetic Variation , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Serine , Transcription, Genetic , Amino Acid Sequence , Animals , Cloning, Molecular , Conserved Sequence , Embryo, Mammalian , Enzyme Activation , Exons , Humans , Kinetics , Male , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligopeptides/chemistry , Oligopeptides/metabolism , Oncogene Proteins/chemistry , Organ Specificity , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Proto-Oncogene Proteins c-akt , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity
15.
Biochem J ; 357(Pt 1): 225-32, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11415453

ABSTRACT

Members of the phosphoprotein phosphatase (PPP) family of protein serine/threonine phosphatases, including protein phosphatase (PP)1, PP2A and PP2B, share invariant active-site residues that are critical for catalytic function [Zhuo, Clemens, Stone and Dixon (1994) J. Biol. Chem. 269, 26234-26238]. Mutation of the active-site residues Asp(88) or His(118) within the human PP2A catalytic subunit (PP2Ac)alpha impaired catalytic activity in vitro; the D88N and H118N substitutions caused a 9- and 23-fold reduction in specific activity respectively, when compared with wild-type recombinant PP2Ac, indicating an important role for these residues in catalysis. Consistent with this, the D88N and H118N substituted forms failed to provide PP2A function in vivo, because, unlike wild-type human PP2Acalpha, neither substituted for the endogenous PP2Ac enzyme of budding yeast. Relative to wild-type PP2Ac, the active-site mutants were dramatically overexpressed in High Five insect cells using the baculovirus system. Milligram quantities of PP2Ac were purified from 1x10(9) High Five cells and the kinetic constants for dephosphorylation of the peptide RRA(pT)VA (single-letter amino-acid notation) by PP2Ac (K(m)=337.5 microM; k(cat)=170 s(-1)) and D88N (K(m)=58.4 microM; k(cat)=2 s(-1)) were determined. The results show that the substitution impairs catalysis severely without a significant effect on substrate binding, consistent with the PPP catalytic mechanism. Combination of the baculovirus and yeast systems provides a strategy whereby the structure-function of PP2Ac may be fully explored, a goal which has previously proven difficult, owing to the stringent auto-regulatory control of PP2Ac protein levels in vivo.


Subject(s)
Phosphoprotein Phosphatases/chemistry , Phosphoprotein Phosphatases/metabolism , Amino Acid Substitution , Animals , Aspartic Acid , Baculoviridae , Binding Sites , Catalysis , Cell Line , Histidine , Humans , Kinetics , Mutagenesis, Site-Directed , Phosphoprotein Phosphatases/genetics , Protein Phosphatase 2 , Protein Subunits , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spodoptera , Transfection
16.
J Biol Chem ; 276(28): 25643-6, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11373274

ABSTRACT

Full activation of protein kinase B (PKB, also called Akt) requires phosphorylation on two regulatory sites, Thr-308 in the activation loop and Ser-473 in the hydrophobic C-terminal regulatory domain (numbering for PKB alpha/Akt-1). Although 3'-phosphoinositide-dependent protein kinase 1 (PDK1) has now been identified as the Thr-308 kinase, the mechanism of the Ser-473 phosphorylation remains controversial. As a step to further characterize the Ser-473 kinase, we examined the effects of a range of protein kinase inhibitors on the activation and phosphorylation of PKB. We found that staurosporine, a broad-specificity kinase inhibitor and inducer of cell apoptosis, attenuated PKB activation exclusively through the inhibition of Thr-308 phosphorylation, with Ser-473 phosphorylation unaffected. The increase in Thr-308 phosphorylation because of overexpression of PDK1 was also inhibited by staurosporine. We further show that staurosporine (CGP 39360) potently inhibited PDK1 activity in vitro with an IC(50) of approximately 0.22 microm. These data indicate that agonist-induced phosphorylation of Ser-473 of PKB is independent of PDK1 or PKB activity and occurs through a distinct Ser-473 kinase that is not inhibited by staurosporine. Moreover, our results suggest that inhibition of PKB signaling is involved in the proapoptotic action of staurosporine.


Subject(s)
Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Animals , Cell Line , Enzyme Activation , Insulin/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt , Serine/metabolism , Signal Transduction , Staurosporine/metabolism
17.
Diabetes ; 50(3): 690-3, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11246893

ABSTRACT

In humans, the Met326Ile missense variant of the p85alpha regulatory subunit of the phosphoinositide 3-kinase (PI3K) has been associated with either significant reductions in glucose effectiveness and intravenous glucose tolerance in Caucasians or a significantly higher insulin secretory response in Pima Indians. In the present study, we genotyped 1,190 Caucasian males to evaluate the impact in vivo of the Met326Ile variant of the p85alpha subunit of PI3K on the acute insulin response, intravenous glucose tolerance, insulin-mediated glucose uptake, and the prevalence of type 2 diabetes after 20 years of follow-up. We also expressed the variant in vitro to evaluate the impact on insulin-stimulated activation of protein kinase B (PKB). The Met326Ile variant of p85alpha was not associated with type 2 diabetes or with alterations in insulin secretion, insulin sensitivity, or intravenous glucose tolerance in vivo. Expressed in vitro, the Ile326 and the Met326 variant acted equally as a dominant-negative and prevented (60-70% inhibition) insulin-mediated activation of PKB by inhibiting the phosphorylation of PKB at Thr308. We conclude that the Met326Ile variant of the p85alpha regulatory subunit of PI3K is likely to be as functionally normal in vivo as in vitro.


Subject(s)
Diabetes Mellitus, Type 2/enzymology , Genetic Variation , Glucose/metabolism , Insulin Resistance , Insulin/metabolism , Phosphatidylinositol 3-Kinases/genetics , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/antagonists & inhibitors , Aged , Cell Line , Cross-Sectional Studies , Enzyme Activation/drug effects , Enzyme Activation/physiology , Glucose Tolerance Test , Humans , Insulin/physiology , Insulin Secretion , Male , Middle Aged , Mutation, Missense/physiology , Phosphatidylinositol 3-Kinases/pharmacology , Phosphorylation/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt
18.
Mol Cell Biol ; 21(3): 952-65, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11154281

ABSTRACT

Serum- and glucocorticoid-inducible kinases (SGKs) form a novel family of serine/threonine kinases that are activated in response to a variety of extracellular stimuli. SGKs are related to Akt (also called PKB), a serine/threonine kinase that plays a crucial role in promoting cell survival. Like Akt, SGKs are activated by the phosphoinositide-3 kinase (PI3K) and translocate to the nucleus upon growth factor stimulation. However the physiological substrates and cellular functions of SGKs remained to be identified. We hypothesized that SGKs regulate cellular functions in concert with Akt by phosphorylating common targets within the nucleus. The best-characterized nuclear substrates of Akt are transcription factors of the Forkhead family. Akt phosphorylates Forkhead transcription factors such as FKHRL1, leading to FKHRL1's exit from the nucleus and the consequent shutoff of FKHRL1 target genes. We show here that SGK1, like Akt, promotes cell survival and that it does so in part by phosphorylating and inactivating FKHRL1. However, SGK and Akt display differences with respect to the efficacy with which they phosphorylate the three regulatory sites on FKHRL1. While both kinases can phosphorylate Thr-32, SGK displays a marked preference for Ser-315 whereas Akt favors Ser-253. These findings suggest that SGK and Akt may coordinately regulate the function of FKHRL1 by phosphorylating this transcription factor at distinct sites. The efficient phosphorylation of these three sites on FKHRL1 by SGK and Akt appears to be critical to the ability of growth factors to suppress FKHRL1-dependent transcription, thereby preventing FKHRL1 from inducing cell cycle arrest and apoptosis. These findings indicate that SGK acts in concert with Akt to propagate the effects of PI3K activation within the nucleus and to mediate the biological outputs of PI3K signaling, including cell survival and cell cycle progression.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Animals , Apoptosis , Base Sequence , Binding Sites , Cell Cycle , Cell Line , Cell Survival , Cricetinae , DNA Primers/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors , Humans , Immediate-Early Proteins , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine/chemistry , Signal Transduction , Transcription Factors/chemistry , Transcription Factors/genetics
19.
J Biol Chem ; 276(11): 8173-9, 2001 Mar 16.
Article in English | MEDLINE | ID: mdl-11087731

ABSTRACT

Insulin-like growth factors positively regulate muscle differentiation through activation of the phosphatidylinositol 3-kinase/protein kinase B (PKB/Akt) signaling pathway. Here, we compare the role of the two closely related alpha (Akt1) and beta (Akt2) isoforms of PKB in muscle differentiation. During differentiation of C2.7 or L6D2 myoblasts, PKBbeta was up-regulated whereas expression of PKBalpha was unaltered. Although the two isoforms were found active in both myoblasts and myotubes, cell fractionation experiments indicated that they displayed distinct subcellular localizations in differentiated cells with only PKBbeta localized in the nuclei. In a transactivation assay, PKBbeta (either wild-type or constitutively active) was more efficient than PKBalpha in activating muscle-specific gene expression. Moreover, microinjection of specific antibodies to PKBbeta inhibited differentiation of muscle cells, whereas control or anti-PKBalpha antibodies did not. On the other hand, microinjection of the anti-PKBalpha antibodies caused a block in cell cycle progression in both non muscle and muscle cells, whereas anti-PKBbeta antibodies had no effect. Taken together, these results show that PKBbeta plays a crucial role in the commitment of myoblasts to differentiation that cannot be substituted by PKBalpha.


Subject(s)
Cell Differentiation , Muscles/cytology , Muscles/enzymology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/physiology , Gene Expression Regulation, Enzymologic , Humans , Microinjections , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Transcriptional Activation
20.
Mol Gen Genet ; 264(4): 425-32, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11129046

ABSTRACT

Protein phosphatase 2A is ubiquitous among eukaryotes and exists as a family of holoenzymes in which the catalytic subunit. PP2Ac, binds a variety of regulatory subunits. Using the yeast Saccharomyces cerevisia, we have investigated the role of the phylogenetically invariant C-terminal leucine residue of PP2Ac, which, in mammalian cells, undergoes reversible methylation and modulates binding of the PR55/B subunit. In S. cerevisiae, the C-terminal Leu-377 residue of Pph22p (equivalent to human PP2Ac Leu-309) was dispensable for cell growth under optimum conditions and its removal, or substitution by alanine, did not inhibit PP2A activity in vitro. However, Leu-377 is required for binding of the yeast PR55/B subunit, Cdc55p, by Pph22p, though apparently not for the binding of Rts1p, the yeast PR61/B' subunit. Furthermore, mutation of this leucine enhanced the sensitivity of cells to microtubule destabilization, a defect characteristic of cdc55delta mutant cells, which are impaired for spindle checkpoint function. These results demonstrate that the regulation of PP2A, mediated by PR55/B binding to the highly conserved PP2Ac C-terminus, is critical for cell viability under conditions of microtubule damage and support a role for PP2A in exit from mitosis.


Subject(s)
Mutation , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Benomyl/toxicity , Cell Cycle Proteins/metabolism , Cell Division , Humans , In Vitro Techniques , Leucine/chemistry , Microtubules/drug effects , Microtubules/metabolism , Nocodazole/toxicity , Phosphoprotein Phosphatases/chemistry , Protein Phosphatase 2 , Protein Subunits , Saccharomyces cerevisiae/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...