Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Oncogene ; 27(50): 6473-88, 2008 Oct 27.
Article in English | MEDLINE | ID: mdl-18955974

ABSTRACT

The Akt serine/threonine kinase (also called protein kinase B) has emerged as a critical signaling molecule within eukaryotic cells. Significant progress has been made in clarifying its regulation by upstream kinases and identifying downstream mechanisms that mediate its effects in cells and contribute to signaling specificity. Here, we provide an overview of present advances in the field regarding the function of Akt in physiological and pathological cell function within a more generalized framework of Akt signal transduction. An emphasis is placed on the involvement of Akt in human diseases ranging from cancer to metabolic dysfunction and mental disease.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Humans , Mental Disorders/enzymology , Mental Disorders/metabolism , Metabolic Diseases/enzymology , Metabolic Diseases/metabolism , Models, Animal , Models, Genetic , Neoplasms/enzymology , Neoplasms/metabolism , Oncogenes , Phosphorylation , Signal Transduction
2.
Oncogene ; 27(8): 1114-21, 2008 Feb 14.
Article in English | MEDLINE | ID: mdl-17704808

ABSTRACT

Astrocyte elevated gene-1 (AEG-1) displays oncogenic properties. Its expression is elevated in diverse neoplastic states and it cooperates with Ha-ras to promote cellular transformation. Overexpression of AEG-1 augments invasion and anchorage-independent growth of transformed cells, while AEG-1 siRNA inhibits Ha-ras-mediated colony formation, supporting a potential functional role in tumorigenesis. Additionally, oncogenic Ha-ras induces AEG-1 expression through the phosphatidylinositol 3-kinase (PI3K)-Akt signaling pathway. In the present study, we investigated whether AEG-1 could induce serum-independent cell growth, another property of oncogenes. Overexpression of AEG-1 inhibited serum starvation-induced apoptosis through activation of PI3K-Akt signaling, one of the effector pathways induced by activated Ras. AEG-1 also affected the phosphorylation state of Akt substrates that are implicated in apoptosis suppression, including glycogen synthase kinase 3beta, c-Myc, murine double minute 2, p53, p21/mda-6 and Bad. Additionally, AEG-1 blocked the activity of serum starvation-induced caspases. Taken together, these observations provide evidence that AEG-1 is an oncogene cooperating with Ha-ras as well as functioning as a downstream target gene of Ha-ras and may perform a central role in Ha-ras-mediated carcinogenesis. Activation of survival pathways may be one mechanism by which AEG-1 exerts its oncogenic properties.


Subject(s)
Astrocytes/enzymology , Cell Adhesion Molecules/physiology , Membrane Proteins/physiology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , Animals , Cell Death/physiology , Cell Line, Transformed , Cell Survival/physiology , Gene Targeting , Genes, ras/physiology , Humans , Mice , RNA-Binding Proteins , Rats , ras Proteins/physiology
3.
Blood ; 98(9): 2853-5, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11675360

ABSTRACT

Immunohistochemistry (IHC) was performed on archived bone marrow (BM) with a phosphospecific anti-AKT antibody. IHC on 26 BM biopsies from patients with multiple myeloma (MM) demonstrated phospho-AKT staining of malignant plasma cells in a cell membrane-specific pattern, whereas nonmalignant hematopoietic cells did not stain. Preabsorption of the antibody with phosphorylated AKT peptide, but not nonphosphorylated peptide, abrogated staining. Frequency of plasma cell staining in BMs of patients with stage I or smoldering MM was significantly less than that of stage III MM marrows. Plasma cells in 10 patients with monoclonal gammopathy of undetermined significance were not stained by the antibody. To investigate the significance of AKT activation, 2 cell lines initiated from cultures of primary MM cells were also studied. Both demonstrated constitutive AKT activation. Interruption of AKT activation and activity, achieved by either exposure to wortmannin or by ectopic expression of a dominant negative AKT mutant, resulted in inhibition of MM cell growth in vitro. These results indicate that activation of the AKT kinase is a characteristic of MM cells and suggest that AKT activity is important for MM cell expansion.


Subject(s)
Multiple Myeloma/enzymology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Androstadienes/pharmacology , Cell Division/drug effects , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Immunohistochemistry , Multiple Myeloma/pathology , Plasma Cells/drug effects , Plasma Cells/enzymology , Plasma Cells/pathology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Transfection , Wortmannin
4.
Mol Cell Biol ; 21(19): 6706-17, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11533257

ABSTRACT

Signals from the extracellular matrix are essential for the survival of many cell types. Dominant-negative mutants of two members of Rho family GTPases, Rac1 and Cdc42, mimic the loss of anchorage in primary mouse fibroblasts and are potent inducers of apoptosis. This pathway of cell death requires the activation of both the p53 tumor suppressor and the extracellular signal-regulated mitogen-activated protein kinases (Erks). Here we characterize the proapoptotic Erk signal and show that it differs from the classically observed survival-promoting one by the intensity of the kinase activation. The disappearance of the GTP-bound forms of Rac1 and Cdc42 gives rise to proapoptotic, moderate activation of the Raf-MEK-Erk cascade via a signaling pathway involving the kinases phosphatidlyinositol 3-kinase and Akt. Moreover, concomitant activation of p53 and inhibition of Akt are both necessary and sufficient to signal anoikis in primary fibroblasts. Our data demonstrate that the GTPases of the Rho family control three major components of cellular signal transduction, namely, p53, Akt, and Erks, which collaborate in the induction of apoptosis due to the loss of anchorage.


Subject(s)
Anoikis , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Proto-Oncogene Proteins/physiology , rho GTP-Binding Proteins/physiology , Animals , Apoptosis , Cell Nucleus/metabolism , Cells, Cultured , Extracellular Matrix/physiology , Fibroblasts/cytology , Fibroblasts/metabolism , MAP Kinase Kinase 1 , MAP Kinase Signaling System , Mice , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Tumor Suppressor Protein p53/metabolism , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/physiology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/physiology , rho GTP-Binding Proteins/genetics
5.
Circulation ; 104(3): 330-5, 2001 Jul 17.
Article in English | MEDLINE | ID: mdl-11457753

ABSTRACT

BACKGROUND: The serine-threonine kinase Akt is activated by several ligand-receptor systems previously shown to be cardioprotective. Akt activation reduces cardiomyocyte apoptosis in models of transient ischemia. Its role in cardiac dysfunction or infarction, however, remains unclear. METHODS AND RESULTS: We examined the effects of a constitutively active Akt mutant (myr-Akt) in a rat model of cardiac ischemia-reperfusion injury. In vivo gene transfer of myr-Akt reduced infarct size by 64% and the number of apoptotic cells by 84% (P<0.005 for each). Ischemia-reperfusion injury decreased regional cardiac wall thickening as well as the maximal rate of left ventricular pressure rise and fall (+dP/dt and -dP/dt). Akt activation restored regional wall thickening and +dP/dt and -dP/dt to levels seen in sham-operated rats. To better understand this benefit, we examined the effects of myr-Akt on hypoxic cardiomyocyte dysfunction in vitro. myr-Akt prevented hypoxia-induced abnormalities in cardiomyocyte calcium transients and shortening. Akt activation also enhanced sarcolemmal expression of Glut-4 in vivo and increased glucose uptake in vitro to the level seen with insulin treatment. CONCLUSIONS: Akt activation exerts a powerful cardioprotective effect after transient ischemia that probably reflects its ability to both inhibit cardiomyocyte death and improve function of surviving cardiomyocytes. Akt may represent an important nodal target for therapy in ischemic and other heart disease.


Subject(s)
Myocardial Ischemia/enzymology , Myocardial Reperfusion Injury/prevention & control , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Adenoviridae/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Count , Cell Survival/drug effects , Cells, Cultured , Coronary Vessels , Disease Models, Animal , Drug Administration Routes , Enzyme Activation/genetics , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Ligation , Male , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Myocardial Ischemia/genetics , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , Proto-Oncogene Proteins/administration & dosage , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley
6.
J Biol Chem ; 276(15): 12190-200, 2001 Apr 13.
Article in English | MEDLINE | ID: mdl-11278518

ABSTRACT

We have identified a novel isoform of rat caspase-9 in which the C terminus of full-length caspase-9 is replaced with an alternative peptide sequence. Casp-9-CTD (where CTD is carboxyl-terminal divergent) is expressed in multiple tissues, with the relative highest expression observed in ovary and heart. Casp-9-CTD was found primarily in the cytoplasm and was not detected in the nucleus. Structural predictions suggest that in contrast to full-length caspase-9, casp-9-CTD will not be processed. Our model is supported by reduced protease activity of casp-9-CTD preparations in vitro and by the lack of detectable processing of casp-9-CTD proenzyme or the induction of cell death following transfection into cells. Both neuronal and non-neuronal cell types transfected with casp-9-CTD were resistant to death evoked by trophic factor deprivation or DNA damage. In addition, cytosolic lysates prepared from cells permanently expressing exogenous casp-9-CTD were resistant to caspase induction by cytochrome c in reconstitution assays. Taken together, our observations indicate that casp-9-CTD acts as a dominant-negative variant. Its expression in various tissues indicates a physiological role in regulating cell death.


Subject(s)
Apoptosis/physiology , Caspases/physiology , Amino Acid Sequence , Animals , Base Sequence , Caspase 3 , Caspase 9 , Caspases/chemistry , Caspases/genetics , Caspases/metabolism , Cell Line , Cloning, Molecular , DNA , DNA Primers , Enzyme Activation , Humans , Molecular Sequence Data , PC12 Cells , Rats , Recombinant Proteins/metabolism , Substrate Specificity
7.
Mol Cell Biol ; 21(3): 893-901, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11154276

ABSTRACT

The Akt family of serine/threonine-directed kinases promotes cellular survival in part by phosphorylating and inhibiting death-inducing proteins. Here we describe a novel functional interaction between Akt and apoptosis signal-regulating kinase 1 (ASK1), a mitogen-activated protein kinase kinase kinase. Akt decreased ASK1 kinase activity stimulated by both oxidative stress and overexpression in 293 cells by phosphorylating a consensus Akt site at serine 83 of ASK1. Activation of the phosphoinositide 3-kinase (PI3-K)/Akt pathway also inhibited the serum deprivation-induced activity of endogenous ASK1 in L929 cells. An association between Akt and ASK1 was detected in cells by coimmunoprecipitation. Phosphorylation by Akt inhibited ASK1-mediated c-Jun N-terminal kinase and activating transcription factor 2 activities in intact cells. Finally, activation of the PI3-K/Akt pathway reduced apoptosis induced by ASK1 in a manner dependent on phosphorylation of serine 83 of ASK1. These results provide the first direct link between Akt and the family of stress-activated kinases.


Subject(s)
MAP Kinase Kinase Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Amino Acid Sequence , Apoptosis , Cell Line , Cell Survival , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase Kinase 5 , MAP Kinase Kinase Kinases/chemistry , MAP Kinase Kinase Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt , Serine/chemistry , Signal Transduction , Substrate Specificity
8.
J Cell Sci ; 114(Pt 1): 81-88, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11112692

ABSTRACT

To investigate the role of Akt in nerve growth factor (NGF)-induced neuronal differentiation, PC12 cells ectopically expressing wild-type or dominant-inhibitory forms of Akt were analyzed. NGF-induced neurite outgrowth was greatly accelerated in cells expressing dominant-inhibitory Akt, compared to parental PC12 cells, but was almost completely blocked in cells expressing wild-type Akt. Since neuronal differentiation requires an arrest of cell growth, several aspects of cell growth of the different cell lines were compared. Cells expressing wild-type Akt were not susceptible to the growth-arresting effect of NGF, whereas parental PC12 cells and notably cells expressing mutant Akt were so affected. Accompanying this, the expressions of CDKs and p21(WAF1) were down- and up-regulated, respectively, in both parental PC12 cells and cells expressing mutant Akt. When treated with some growth arrest-inducing agents such as sodium nitroprusside, forskolin and butyrolactone I, cells expressing wild-type Akt regained their responsiveness to the effects of NGF on differentiation. In summary, our results indicate that Akt overrides the growth-arresting effect of NGF and thereby, negatively regulates neuronal differentiation.


Subject(s)
4-Butyrolactone/analogs & derivatives , Cell Differentiation/physiology , Neurites/physiology , Neurons/cytology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/physiology , 4-Butyrolactone/pharmacology , Animals , CDC2 Protein Kinase/metabolism , Cell Division , Colforsin/pharmacology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression , Nerve Growth Factor/physiology , Neurites/drug effects , Neurons/drug effects , Nitroprusside/pharmacology , PC12 Cells , Protein Kinase Inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Rats
9.
Endocrinology ; 141(12): 4503-11, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11108261

ABSTRACT

This study examines whether the serine/threonine protein kinase, Akt, is involved in the cross-talk between epidermal growth factor (EGF) and insulin-related growth factor I (IGF-I) receptors and ER-alpha. Treatment of MCF-7 cells with either EGF or IGF-I resulted in a rapid phosphorylation of Akt and a 14- to 16-fold increase in Akt activity, respectively. Akt activation was blocked by inhibitors of phosphatidylinositol 3-kinase, but not by an inhibitor of the ribosomal protein kinase p70S6K. Stable transfection of cells with a dominant negative Akt mutant blocked the effects of EGF and IGF-I on ER-alpha expression and activity, whereas stable transfection of cells with a constitutively active Akt mutant mimicked the effects of EGF and IGF-I. In the latter cells, there was a decrease in the amount of ER-alpha protein and messenger RNA (70-80%) and an increase in the amount of progesterone receptor protein, messenger RNA (4- to 9- and by 3.5- to 7-fold, respectively) and pS2 (3- to 5-fold). Coexpression of wild-type ER-alpha and the dominant negative Akt mutant in COS-1 cells also blocked the growth factor-stimulated activation of ER-alpha, but coexpression of the wild-type receptor with the constitutively active Akt mutant increased ER-alpha activity. Receptor activation was blocked by an antiestrogen. Studies using mutants of ER-alpha demonstrated that Akt increased estrogen receptor activity through the amino-terminal activation function-1 (AF-1). Serines S104 S106, S118, and S167 appear to play a role in the activation of ER-alpha by Akt.


Subject(s)
Epidermal Growth Factor/physiology , Estrogens/physiology , Insulin-Like Growth Factor I/physiology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/physiology , Receptors, Estrogen/physiology , Animals , COS Cells , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , Estrogen Receptor alpha , Gene Expression , Humans , Insulin-Like Growth Factor I/pharmacology , Mutation , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , RNA, Messenger/analysis , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Transfection , Tumor Cells, Cultured
10.
Diabetes ; 49(5): 847-56, 2000 May.
Article in English | MEDLINE | ID: mdl-10905496

ABSTRACT

To determine whether impaired Akt (protein kinase B or rac) activation contributes to insulin resistance in vivo, we examined the expression, phosphorylation, and kinase activities of Akt1 and Akt2 isoforms in insulin target tissues of insulin-resistant obese Zucker rats. In lean rats, insulin (10 U/kg i.v. x 2.5 min) stimulated Akt1 activity 6.2-, 8.8-, and 4.4-fold and Akt2 activity 5.4-, 9.3-, and 1.8-fold in muscle, liver, and adipose tissue, respectively. In obese rats, insulin-stimulated Akt1 activity decreased 30% in muscle and 21% in adipose tissue but increased 37% in liver compared with lean littermates. Insulin-stimulated Akt2 activity decreased 29% in muscle and 37% in liver but increased 24% in adipose tissue. Akt2 protein levels were reduced 56% in muscle and 35% in liver of obese rats, but Akt1 expression was unaltered. Phosphoinositide 3-kinase (PI3K) activity associated with insulin receptor substrate (IRS)-1 or phosphotyrosine was reduced 67-86% in tissues of obese rats because of lower IRS-1 protein levels and reduced insulin receptor and IRS-1 phosphorylation. In adipose tissue of obese rats, in spite of an 86% reduction in insulin-stimulated PI3K activity, activation of Akt2 was increased. Maximal insulin-stimulated (100 nmol/l) glucose transport was reduced 70% in isolated adipocytes, with a rightward shift in the insulin dose response for transport and for Akt1 stimulation but normal sensitivity for Akt2. These findings suggest that PI3K-dependent effects on glucose transport in adipocytes are not mediated primarily by Akt2. Akt1 and Akt2 activations by insulin have a similar time course and are maximal by 2.5 min in adipocytes of both lean and obese rats. We conclude that 1) activation of Akt1 and Akt2 in vivo is much less impaired than activation of PI3K in this insulin-resistant state, and 2) the mechanisms for divergent alterations in insulin action on Akt1 and Akt2 activities in tissues of insulin-resistant obese rats involve tissue- and isoform-specific changes in both expression and activation.


Subject(s)
Insulin/physiology , Isoenzymes/metabolism , Obesity/enzymology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Adipose Tissue/metabolism , Animals , Biological Transport , Female , Glucose/metabolism , Insulin/pharmacology , Insulin Receptor Substrate Proteins , Peptide Fragments/metabolism , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt , Rats , Rats, Zucker , Reference Values , Tissue Distribution , Tyrosine/metabolism
11.
Methods Enzymol ; 322: 400-10, 2000.
Article in English | MEDLINE | ID: mdl-10914036

ABSTRACT

An increasing number of publications have underscored the importance of the serine/threonine kinase Akt in the regulation of cell survival, proliferation, and insulin-dependent metabolic cell responses. Critical to the understanding of Akt signaling in cells are experimental methods that assess its activation and phosphorylation state. In this chapter, we evaluate the most commonly used techniques to examine Akt activity. Immunocomplex kinase assays that utilize Akt-specific substrates are described, as is the use of phosphospecific antibodies directed against Akt phosphorylation sites. Furthermore, we introduce coupled enzyme assays that indirectly measure the activity of Akt by examining the activity of Akt substrates.


Subject(s)
Proto-Oncogene Proteins/analysis , Animals , Cell Line , Isoenzymes/analysis , Isoenzymes/metabolism , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Recombinant Fusion Proteins/analysis , Recombinant Proteins/analysis , Recombinant Proteins/metabolism , Spodoptera , Transfection/methods
12.
J Biol Chem ; 275(19): 14624-31, 2000 May 12.
Article in English | MEDLINE | ID: mdl-10799549

ABSTRACT

The serine/threonine kinase Akt (also known as protein kinase B) is activated in response to various stimuli by a mechanism involving phosphoinositide 3-kinase (PI3-K). Akt provides a survival signal that protects cells from apoptosis induced by growth factor withdrawal, but its function in other forms of stress is less clear. Here we investigated the role of PI3-K/Akt during the cellular response to oxidant injury. H(2)O(2) treatment elevated Akt activity in multiple cell types in a time- (5-30 min) and dose (400 microM-2 mm)-dependent manner. Expression of a dominant negative mutant of p85 (regulatory component of PI3-K) and treatment with inhibitors of PI3-K (wortmannin and LY294002) prevented H(2)O(2)-induced Akt activation. Akt activation by H(2)O(2) also depended on epidermal growth factor receptor (EGFR) signaling; H(2)O(2) treatment led to EGFR phosphorylation, and inhibition of EGFR activation prevented Akt activation by H(2)O(2). As H(2)O(2) causes apoptosis of HeLa cells, we investigated whether alterations of PI3-K/Akt signaling would affect this response. Wortmannin and LY294002 treatment significantly enhanced H(2)O(2)-induced apoptosis, whereas expression of exogenous myristoylated Akt (an activated form) inhibited cell death. Constitutive expression of v-Akt likewise enhanced survival of H(2)O(2)-treated NIH3T3 cells. These results suggest that H(2)O(2) activates Akt via an EGFR/PI3-K-dependent pathway and that elevated Akt activity confers protection against oxidative stress-induced apoptosis.


Subject(s)
Cell Survival , ErbB Receptors/metabolism , Oxidative Stress , Retroviridae Proteins, Oncogenic/metabolism , Animals , Apoptosis/drug effects , Enzyme Activation , Humans , Hydrogen Peroxide/pharmacology , Oncogene Protein v-akt , Phosphatidylinositol 3-Kinases/metabolism
13.
Mol Cell Endocrinol ; 170(1-2): 211-5, 2000 Dec 22.
Article in English | MEDLINE | ID: mdl-11162904

ABSTRACT

We previously reported insulin-like growth factor-I (IGF-I) promotes Schwann cell (SC) motility and rescues SC from apoptosis induced by serum deprivation. This effect is mediated by phosphatidylinositol-3 (PI-3) kinase. In the current study, we examined the role of Akt, a downstream kinase of PI-3K, in SC motility and IGF-I mediated protection from apoptosis. IGF-I induces Akt phosphorylation at Ser473, an event which may be blocked by pretreatment with a PI-3K inhibitor, LY294002. In dominant negative K179M Akt (K179M) transfected SC, however, Akt is not activated in response to IGF-I. In addition, IGF-I is unable to promote SC motility and survival in K179M SC. These results suggest a critical role for Akt in IGF-I mediated motility and survival in SC.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/pharmacology , Schwann Cells/cytology , Animals , Cell Movement/drug effects , Cell Survival/drug effects , Cells, Cultured , Humans , Mutation, Missense , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , Schwann Cells/drug effects , Transfection
14.
Oncogene ; 18(47): 6564-72, 1999 Nov 11.
Article in English | MEDLINE | ID: mdl-10597260

ABSTRACT

To investigate the role of protein kinase C (PKC) in apoptotic signaling induced by cytokine withdrawal, we expressed PKC-alpha, -delta and -epsilon individually in the 32D myeloid progenitor cells. The parental and PKC-delta- and PKC-epsilon-transfected 32D cells underwent apoptosis within 24 h in the absence of interleukin 3. In contrast, expression of PKC-alpha inhibited the onset of apoptosis as determined by genomic DNA fragmentation and flow cytometric analysis. Correlating with the inhibition of apoptosis, PKC-alpha transfectants exhibited increased activity of the endogenous Akt serine/threonine kinase. Furthermore, PKC-alpha, but not PKC-delta or -epsilon, specifically activated overexpressed Akt. PKC-alpha-induced Akt activity was partially dependent on phosphoinositol 3' kinase (PI 3'K) since a PI 3'K inhibitor was able to suppress PKC-alpha-induced Akt activation. Both basal and interleukin 3-stimulated phosphorylation of Akt on serine 473 was enhanced in the PKC-alpha and Akt contransfectants. Coexpression of wild type Akt and PKC-alpha resulted in greater suppression of apoptosis than PKC-alpha expression alone. Together, our results demonstrate that suppression of apoptosis by PKC-alpha correlates with its ability of activating endogenous Akt. Furthermore, activation of overexpressed Akt by PKC-alpha is consistent with their synergistic effect on suppressing apoptosis, providing the strong evidence of cross talk between Akt and PKC-alpha.


Subject(s)
Apoptosis , Interleukin-3/pharmacology , Isoenzymes/metabolism , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Androstadienes/pharmacology , Apoptosis/drug effects , Cell Line , DNA Replication , Enzyme Inhibitors/pharmacology , Interleukin-3/administration & dosage , Isoenzymes/genetics , Phosphorylation , Protein Kinase C/genetics , Protein Kinase C-alpha , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Serine/metabolism , Transfection , Wortmannin
15.
Circulation ; 100(23): 2373-9, 1999 Dec 07.
Article in English | MEDLINE | ID: mdl-10587343

ABSTRACT

BACKGROUND: The intracellular signaling pathways that control cardiomyocyte apoptosis have not been fully defined. Because insulin-like growth factor-1 (IGF-1) prevents cardiomyocyte apoptosis, we examined the role of its downstream signaling molecules in an in vitro model of hypoxia-induced cardiomyocyte apoptosis. METHODS AND RESULTS: Treatment of rat neonatal cardiomyocytes with IGF-1 increased activity of both phosphatidylinositol 3' (PI 3)-kinase and its downstream target, Akt (also known as protein kinase B or PKB). Cardiomyocytes were subjected to hypoxia for 24 hours, and apoptosis was assessed by DNA laddering, TUNEL staining, and ELISA for histone-associated DNA fragments. IGF-1 treatment (100 nmol/L) reduced cardiomyocyte apoptosis, and this effect was inhibited by simultaneous treatment with a PI 3-kinase inhibitor. Cardiomyocytes were infected with either a control adenovirus (Ad.EGFP) or adenoviruses carrying constitutively active forms of PI 3-kinase (Ad.BD110) or Akt (Ad. myr-Akt-HA). Ad.BD110 significantly inhibited apoptosis of hypoxic cardiomyocytes compared with Ad.EGFP (61.0+/-4.6% less DNA fragmentation than in Ad.EGFP-infected cells, P<0.0001). Ad. myr-Akt-HA even more dramatically inhibited apoptosis of hypoxic cardiomyocytes (90.9+/-1.4% less DNA fragmentation than in controls, P<0.0001). CONCLUSIONS: IGF-1 activates PI 3-kinase and Akt in cardiomyocytes. Activated PI 3-kinase and Akt are each sufficient to protect hypoxic cardiomyocytes against apoptosis in vitro. Adenoviral gene transfer provides a useful tool for investigating the role of these signaling pathways in cardiomyocyte apoptosis.


Subject(s)
Adenoviridae , Apoptosis/physiology , Gene Transfer Techniques , Phosphatidylinositol 3-Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins , Animals , Apoptosis/drug effects , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cells, Cultured , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Viral , In Situ Nick-End Labeling , In Vitro Techniques , Insulin-Like Growth Factor I/pharmacology , Morpholines/pharmacology , Muscle Fibers, Skeletal/cytology , Myocardial Ischemia/enzymology , Myocardium/cytology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt , Rats , Signal Transduction/physiology
16.
J Biol Chem ; 274(37): 26393-8, 1999 Sep 10.
Article in English | MEDLINE | ID: mdl-10473597

ABSTRACT

The signaling pathways activated by the macrophage colony-stimulating factor (M-CSF) to promote survival of monocyte and macrophage lineage cells are not well established. In an effort to elucidate these pathways, we have used two cell types responsive to M-CSF: NIH 3T3 fibroblasts genetically engineered to express human M-CSF receptors (3T3-FMS cells) and human monocytes. M-CSF treatment induced M-CSF receptor tyrosine phosphorylation and recruitment of the p85 subunit of phosphatidylinositol 3-kinase (PI3K) to these receptors. These M-CSF receptor events correlated with activation of the serine/threonine kinase Akt. To clarify that PI3K products activate Akt in response to M-CSF, NIH 3T3 fibroblasts expressing mutant human M-CSF receptors (3T3-FMS(Y809F)) that fail to activate Ras in response to M-CSF also exhibit increased Akt kinase activity in response to M-CSF challenge. Furthermore, Akt appears to be the primary regulator of survival in 3T3-FMS cells, as transfection of genes encoding dominant-negative Akt isoforms into these fibroblasts blocked M-CSF-induced survival. In normal human monocytes, M-CSF increased the levels of tyrosine-phosphorylated proteins and induced Akt activation in a PI3K-dependent manner. The PI3K inhibitor LY294002 blocked M-CSF-mediated monocyte survival, an effect that was partially restored by caspase-9 inhibitors. These data suggest that M-CSF may induce cell survival through Akt-induced suppression of caspase-9 activation.


Subject(s)
Cell Survival/physiology , Macrophage Colony-Stimulating Factor/physiology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , 3T3 Cells , Animals , Caspase 9 , Caspases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Humans , Mice , Monocytes/drug effects , Monocytes/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt
17.
Nature ; 399(6736): 597-601, 1999 Jun 10.
Article in English | MEDLINE | ID: mdl-10376602

ABSTRACT

Endothelial nitric oxide synthase (eNOS) is the nitric oxide synthase isoform responsible for maintaining systemic blood pressure, vascular remodelling and angiogenesis. eNOS is phosphorylated in response to various forms of cellular stimulation, but the role of phosphorylation in the regulation of nitric oxide (NO) production and the kinase(s) responsible are not known. Here we show that the serine/threonine protein kinase Akt (protein kinase B) can directly phosphorylate eNOS on serine 1179 and activate the enzyme, leading to NO production, whereas mutant eNOS (S1179A) is resistant to phosphorylation and activation by Akt. Moreover, using adenovirus-mediated gene transfer, activated Akt increases basal NO release from endothelial cells, and activation-deficient Akt attenuates NO production stimulated by vascular endothelial growth factor. Thus, eNOS is a newly described Akt substrate linking signal transduction by Akt to the release of the gaseous second messenger NO.


Subject(s)
Endothelium, Vascular/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/biosynthesis , Retroviridae Proteins, Oncogenic/metabolism , Animals , COS Cells , Cattle , Humans , Mutation , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type III , Oncogene Protein v-akt , Phosphorylation , Rats , Serine/metabolism , Signal Transduction , Transfection
18.
Science ; 284(5412): 339-43, 1999 Apr 09.
Article in English | MEDLINE | ID: mdl-10195903

ABSTRACT

The Ca2+-activated protein phosphatase calcineurin induces apoptosis, but the mechanism is unknown. Calcineurin was found to dephosphorylate BAD, a pro-apoptotic member of the Bcl-2 family, thus enhancing BAD heterodimerization with Bcl-xL and promoting apoptosis. The Ca2+-induced dephosphorylation of BAD correlated with its dissociation from 14-3-3 in the cytosol and translocation to mitochondria where Bcl-xL resides. In hippocampal neurons, L-glutamate, an inducer of Ca2+ influx and calcineurin activation, triggered mitochondrial targeting of BAD and apoptosis, which were both suppressible by coexpression of a dominant-inhibitory mutant of calcineurin or pharmacological inhibitors of this phosphatase. Thus, a Ca2+-inducible mechanism for apoptosis induction operates by regulating BAD phosphorylation and localization in cells.


Subject(s)
Apoptosis , Calcineurin/metabolism , Calcium/metabolism , Carrier Proteins/metabolism , Tyrosine 3-Monooxygenase , 14-3-3 Proteins , Animals , Calcineurin/genetics , Calcineurin Inhibitors , Calcium/pharmacology , Carrier Proteins/chemistry , Cell Line , Cells, Cultured , Dimerization , Enzyme Inhibitors/pharmacology , Glutamic Acid/pharmacology , Hippocampus/cytology , Humans , Mitochondria/metabolism , Neurons/cytology , Neurons/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Recombinant Fusion Proteins/metabolism , Transfection , bcl-Associated Death Protein , bcl-X Protein
19.
Science ; 282(5392): 1318-21, 1998 Nov 13.
Article in English | MEDLINE | ID: mdl-9812896

ABSTRACT

Caspases are intracellular proteases that function as initiators and effectors of apoptosis. The kinase Akt and p21-Ras, an Akt activator, induced phosphorylation of pro-caspase-9 (pro-Casp9) in cells. Cytochrome c-induced proteolytic processing of pro-Casp9 was defective in cytosolic extracts from cells expressing either active Ras or Akt. Akt phosphorylated recombinant Casp9 in vitro on serine-196 and inhibited its protease activity. Mutant pro-Casp9(Ser196Ala) was resistant to Akt-mediated phosphorylation and inhibition in vitro and in cells, resulting in Akt-resistant induction of apoptosis. Thus, caspases can be directly regulated by protein phosphorylation.


Subject(s)
Apoptosis , Caspases/metabolism , Caspase 9 , Caspase Inhibitors , Cell Line , Cytochrome c Group/pharmacology , Enzyme Precursors/metabolism , Humans , Mass Spectrometry , Mutation , Peptide Fragments/metabolism , Phosphorylation , Phosphoserine/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins p21(ras)/metabolism , Recombinant Fusion Proteins/metabolism , Transfection
20.
Mol Biol Cell ; 9(11): 3107-18, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9802900

ABSTRACT

We have shown previously that interleukin-4 (IL-4) protects TS1alphabeta cells from apoptosis, but very little is known about the mechanism by which IL-4 exerts this effect. We found that Akt activity, which is dependent on phosphatidylinositol 3 kinase, is reduced in IL-4-deprived TS1alphabeta cells. Overexpression of wild-type Akt or a constitutively active Akt mutant protects cells from IL-4 deprivation-induced apoptosis. Readdition of IL-4 before the commitment point is able to restore Akt activity. We also show expression and c-Jun N-terminal kinase 2 activation after IL-4 deprivation. Overexpression of the constitutively activated Akt mutant in IL-4-deprived cells correlates with inhibition of c-Jun N-terminal kinase 2 activity. Finally, TS1alphabeta survival is independent of Bcl-2, Bcl-x, or Bax.


Subject(s)
Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Interleukin-4/metabolism , Mitogen-Activated Protein Kinases , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Animals , Cell Line , Enzyme Activation , JNK Mitogen-Activated Protein Kinases , Mice , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...