Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 8(12): 3204, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29233971

ABSTRACT

Pancreatic cancer cells show varying sensitivity to the anticancer effects of gemcitabine. However, as a chemotherapeutic agent, gemcitabine can cause intolerably high levels of toxicity and patients often develop resistance to the beneficial effects of this drug. Combination studies show that use of gemcitabine with the pro-apoptotic cytokine TRAIL can enhance the inhibition of survival and induction of apoptosis of pancreatic cancer cells. Additionally, following combination treatment there is a dramatic increase in the level of the hypophosphorylated form of the tumour suppressor protein 4E-BP1. This is associated with inhibition of mTOR activity, resulting from caspase-mediated cleavage of the Raptor and Rictor components of mTOR. Use of the pan-caspase inhibitor Z-VAD-FMK indicates that the increase in level of 4E-BP1 is also caspase-mediated. ShRNA-silencing of 4E-BP1 expression renders cells more resistant to cell death induced by the combination treatment. Since the levels of 4E-BP1 are relatively low in untreated pancreatic cancer cells these results suggest that combined therapy with gemcitabine and TRAIL could improve the responsiveness of tumours to treatment by elevating the expression of 4E-BP1.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Antineoplastic Agents/pharmacology , Deoxycytidine/analogs & derivatives , Gene Expression Regulation, Neoplastic , Phosphoproteins/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/drug effects , Caspases/genetics , Caspases/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Deoxycytidine/pharmacology , Drug Combinations , Drug Synergism , Humans , Pancreas/drug effects , Pancreas/metabolism , Pancreas/pathology , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/metabolism , Phosphorylation , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Regulatory-Associated Protein of mTOR/genetics , Regulatory-Associated Protein of mTOR/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Time-Lapse Imaging , Gemcitabine
2.
J Interferon Cytokine Res ; 34(10): 769-77, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24731196

ABSTRACT

Tumor necrosis factor α-related apoptosis-inducing ligand (TRAIL) is a potent inducer of apoptosis in Jurkat T lymphoma cells. One of the characteristics of the phase preceding overt apoptosis is the marked downregulation of protein synthesis. We have investigated factors that can influence this response and have explored some of the signaling pathways involved. We show that interferon-α (IFNα) pretreatment desensitizes Jurkat cells to TRAIL-induced inhibition of protein synthesis, such that the concentration of TRAIL required for 50% inhibition is increased by 10-fold. The inhibition of translation is characterized by dephosphorylation of the eIF4E-binding protein 4E-BP1 and IFNα desensitizes Jurkat cells to this effect. IFNα also inhibits TRAIL-mediated dephosphorylation of the growth-promoting protein kinase B (Akt). Since Jurkat cells are defective for phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and therefore have constitutive phosphoinositide 3-kinase (PI3K) activity, we investigated the consequences for protein synthesis of inhibiting PI3K using LY294002. Inhibition of PI3K partially inhibits translation, but also enhances the effect of a suboptimal concentration of TRAIL. However, LY294002 does not block the ability of IFNα to protect protein synthesis from TRAIL-induced inhibition. Data are presented suggesting that IFNα impairs the process of activation of caspase-8 within the TRAIL death-inducing signaling complex.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Protein Biosynthesis/drug effects , T-Lymphocytes/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/drug effects , Apoptosis/immunology , Caspase 8/metabolism , Cell Cycle Proteins , Chromones/pharmacology , Humans , Immunomodulation , Interferon-alpha/immunology , Jurkat Cells , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphoproteins/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes/immunology
3.
PLoS One ; 8(8): e71138, 2013.
Article in English | MEDLINE | ID: mdl-23940704

ABSTRACT

The protein kinase mammalian target of rapamycin (mTOR) regulates the phosphorylation and activity of several proteins that have the potential to control translation, including p70S6 kinase and the eIF4E binding proteins 4E-BP1 and 4E-BP2. In spite of this, in exponentially growing cells overall protein synthesis is often resistant to mTOR inhibitors. We report here that sensitivity of wild-type mouse embryonic fibroblasts (MEFs) to mTOR inhibitors can be greatly increased when the cells are subjected to the physiological stress imposed by hypertonic conditions. In contrast, protein synthesis in MEFs with a double knockout of 4E-BP1 and 4E-BP2 remains resistant to mTOR inhibitors under these conditions. Phosphorylation of p70S6 kinase and protein kinase B (Akt) is blocked by the mTOR inhibitor Ku0063794 equally well in both wild-type and 4E-BP knockout cells, under both normal and hypertonic conditions. The response of protein synthesis to hypertonic stress itself does not require the 4E-BPs. These data suggest that under certain stress conditions: (i) translation has a greater requirement for mTOR activity and (ii) there is an absolute requirement for the 4E-BPs for regulation by mTOR. Importantly, dephosphorylation of p70S6 kinase and Akt is not sufficient to affect protein synthesis acutely.


Subject(s)
Eukaryotic Initiation Factor-4E/metabolism , Morpholines/pharmacology , Protein Biosynthesis/drug effects , Pyrimidines/pharmacology , Saline Solution, Hypertonic/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/physiology , Cell Cycle Proteins , Cells, Cultured , Down-Regulation/drug effects , Eukaryotic Initiation Factors/physiology , Furans/pharmacology , Mice , Phosphoproteins/physiology , Protein Biosynthesis/genetics , Pyridines/pharmacology
4.
Biol Cell ; 105(9): 414-29, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23734772

ABSTRACT

BACKGROUND INFORMATION: Tumour cells can be induced to undergo apoptosis after treatment with the tumour necrosis factor α-related death-inducing ligand (TRAIL). Although human pancreatic cancer cells show varying degrees of response they can be sensitised to the pro-apoptotic effects of TRAIL in the presence of celastrol, a natural compound extracted from the plant Tripterygium wilfordii Hook F. One important aspect of the cellular response to TRAIL is the control of protein synthesis, a key regulator of which is the eukaryotic initiation factor 4E-binding protein, 4E-BP1. RESULTS: We examined the effects of celastrol and TRAIL in several pancreatic cancer cell lines. In cells that are normally resistant to TRAIL, synergistic effects of TRAIL plus celastrol on commitment to apoptosis and inhibition of protein synthesis were observed. These were associated with a strong up-regulation and dephosphorylation of 4E-BP1. The enhancement of 4E-BP1 expression, which correlated with a threefold increase in the level of the 4E-BP1 transcript, was blocked by inhibitors of reactive oxygen species and the JNK protein kinase. When the expression of 4E-BP1 was reduced by an inducible micro-RNA, TRAIL-mediated apoptosis was inhibited. CONCLUSION: These results suggest that 4E-BP1 plays a critical role in the mechanism by which TRAIL and celastrol together cause apoptotic cell death in human pancreatic tumour cells.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Phosphoproteins/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Triterpenes/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/drug effects , Cell Cycle Proteins , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pentacyclic Triterpenes , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Protein Biosynthesis , Recombinant Proteins/pharmacology , Signal Transduction , Pancreatic Neoplasms
5.
Proc Natl Acad Sci U S A ; 107(12): 5375-80, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20212154

ABSTRACT

The DNA damage response activates several pathways that stall the cell cycle and allow DNA repair. These consist of the well-characterized ATR (Ataxia telangiectasia and Rad-3 related)/CHK1 and ATM (Ataxia telangiectasia mutated)/CHK2 pathways in addition to a newly identified ATM/ATR/p38MAPK/MK2 checkpoint. Crucial to maintaining the integrity of the genome is the S-phase checkpoint that functions to prevent DNA replication until damaged DNA is repaired. Inappropriate expression of the proto-oncogene c-Myc is known to cause DNA damage. One mechanism by which c-Myc induces DNA damage is through binding directly to components of the prereplicative complex thereby promoting DNA synthesis, resulting in replication-associated DNA damage and checkpoint activation due to inappropriate origin firing. Here we show that following etoposide-induced DNA damage translation of c-Myc is repressed by miR-34c via a highly conserved target-site within the 3(') UTR. While miR-34c is induced by p53 following DNA damage, we show that in cells lacking p53 this is achieved by an alternative pathway which involves p38 MAPK signalling to MK2. The data presented here suggest that a major physiological target of miR-34c is c-Myc. Inhibition of miR-34c activity prevents S-phase arrest in response to DNA damage leading to increased DNA synthesis, DNA damage, and checkpoint activation in addition to that induced by etoposide alone, which are all reversed by subsequent c-Myc depletion. These data demonstrate that miR-34c is a critical regulator of the c-Myc expression following DNA damage acting downstream of p38 MAPK/MK2 and suggest that miR-34c serves to remove c-Myc to prevent inappropriate replication which may otherwise lead to genomic instability.


Subject(s)
DNA Damage , DNA Replication/physiology , Intracellular Signaling Peptides and Proteins/metabolism , MicroRNAs/biosynthesis , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , 3' Untranslated Regions , Animals , Cell Line , DNA Replication/genetics , HeLa Cells , Humans , MAP Kinase Signaling System , Mice , MicroRNAs/genetics , Proto-Oncogene Mas , S Phase/genetics , S Phase/physiology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
J Biol Chem ; 285(4): 2610-21, 2010 Jan 22.
Article in English | MEDLINE | ID: mdl-19940154

ABSTRACT

3'-Deoxyadenosine, also known as cordycepin, is a known polyadenylation inhibitor with a large spectrum of biological activities, including anti-proliferative, pro-apoptotic and anti-inflammatory effects. In this study we confirm that cordycepin reduces the length of poly(A) tails, with some mRNAs being much more sensitive than others. The low doses of cordycepin that cause poly(A) changes also reduce the proliferation of NIH3T3 fibroblasts. At higher doses of the drug we observed inhibition of cell attachment and a reduction of focal adhesions. Furthermore, we observed a strong inhibition of total protein synthesis that correlates with an inhibition of mammalian target of rapamycin (mTOR) signaling, as observed by reductions in Akt kinase and 4E-binding protein (4EBP) phosphorylation. In 4EBP knock-out cells, the effect of cordycepin on translation is strongly reduced, confirming the role of this modification. In addition, the AMP-activated kinase (AMPK) was shown to be activated. Inhibition of AMPK prevented translation repression by cordycepin and abolished 4EBP1 dephosphorylation, indicating that the effect of cordycepin on mTOR signaling and protein synthesis is mediated by AMPK activation. We conclude that many of the reported biological effects of cordycepin are likely to be due to its effects on mTOR and AMPK signaling.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Adhesion/drug effects , Deoxyadenosines/pharmacology , Protein Synthesis Inhibitors/pharmacology , Signal Transduction/drug effects , Actin Cytoskeleton/drug effects , Adenylate Kinase/metabolism , Animals , Cell Division/drug effects , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Mice , NIH 3T3 Cells , Polyadenylation/drug effects , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/drug effects , TOR Serine-Threonine Kinases
7.
J Biol Chem ; 283(45): 30482-92, 2008 Nov 07.
Article in English | MEDLINE | ID: mdl-18676370

ABSTRACT

Signaling through mammalian target of rapamycin complex 1 (mTORC1) is stimulated by amino acids and insulin. Insulin inactivates TSC1/2, the GTPase-activator complex for Rheb, and Rheb.GTP activates mTORC1. It is not clear how amino acids regulate mTORC1. FKBP38 (immunophilin FK506-binding protein, 38 kDa), was recently reported to exert a negative effect on mTORC1 function that is relieved by its binding to Rheb.GTP. We confirm that Rheb binds wild type FKBP38, but inactive Rheb mutants showed contrasting abilities to bind FKBP38. We were unable to observe any regulation of FKBP38/mTOR binding by amino acids or insulin. Furthermore, FKBP38 did not inhibit mTORC1 signaling. The translationally controlled tumor protein (TCTP) in Drosophila was recently reported to act as the guanine nucleotide-exchange factor for Rheb. We have studied the role of TCTP in mammalian TORC1 signaling and its control by amino acids. Reducing TCTP levels did not reproducibly affect mTORC1 signaling in amino acid-replete/insulin-stimulated cells. Moreover, overexpressing TCTP did not rescue mTORC1 signaling in amino acid-starved cells. In addition, we were unable to see any stable interaction between TCTP and Rheb or mTORC1. Accumulation of uncharged tRNA has been previously proposed to be involved in the inhibition of mTORC1 signaling during amino acid starvation. To test this hypothesis, we used a Chinese hamster ovary cell line containing a temperature-sensitive mutation in leucyl-tRNA synthetase. Leucine deprivation markedly inhibited mTORC1 signaling in these cells, but shifting the cells to the nonpermissive temperature for the synthetase did not. These data indicate that uncharged tRNA(Leu) does not switch off mTORC1 signaling and suggest that mTORC1 is controlled by a distinct pathway that senses the availability of amino acids. Our data also indicate that, in the mammalian cell lines tested here, neither TCTP nor FKBP38 regulates mTORC1 signaling.


Subject(s)
Amino Acids/metabolism , Multiprotein Complexes/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Amino Acids/pharmacology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , CHO Cells , Cricetinae , Cricetulus , Humans , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Insulin/pharmacology , Leucine-tRNA Ligase/genetics , Leucine-tRNA Ligase/metabolism , Mechanistic Target of Rapamycin Complex 1 , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Multiprotein Complexes/genetics , Mutation , Neuropeptides/genetics , Neuropeptides/metabolism , Protein Binding/physiology , Proteins , RNA, Transfer/genetics , RNA, Transfer/metabolism , Ras Homolog Enriched in Brain Protein , TOR Serine-Threonine Kinases , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism , Telomerase/genetics , Telomerase/metabolism , Transcription Factors/genetics , Tuberous Sclerosis Complex 2 Protein , Tumor Protein, Translationally-Controlled 1 , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
8.
Biol Cell ; 100(5): 279-89, 2008 May.
Article in English | MEDLINE | ID: mdl-18021075

ABSTRACT

BACKGROUND INFORMATION: The translational inhibitor protein 4E-BP1 [eIF4E (eukaryotic initiation factor 4E)-binding protein 1] regulates the availability of polypeptide chain initiation factor eIF4E for protein synthesis. Initiation factor eIF4E binds the 5' cap structure present on all cellular mRNAs. Its ability to associate with initiation factors eIF4G and eIF4A, forming the eIF4F complex, brings the mRNA to the 43S complex during the initiation of translation. Binding of eIF4E to eIF4G is inhibited in a competitive manner by 4E-BP1. Phosphorylation of 4E-BP1 decreases the affinity of this protein for eIF4E, thus favouring the binding of eIF4G and enhancing translation. We have previously shown that induction or activation of the tumour suppressor protein p53 rapidly leads to 4E-BP1 dephosphorylation, resulting in sequestration of eIF4E, decreased formation of the eIF4F complex and inhibition of protein synthesis. RESULTS: We now report that activation of p53 also results in modification of 4E-BP1 to a truncated form. Unlike full-length 4E-BP1, which is reversibly phosphorylated at multiple sites, the truncated protein is almost completely unphosphorylated. Moreover, the latter interacts with eIF4E in preference to full-length 4E-BP1. Inhibitor studies indicate that the p53-induced cleavage of 4E-BP1 is mediated by the proteasome and is blocked by conditions that inhibit the dephosphorylation of full-length 4E-BP1. Measurements of the turnover of 4E-BP1 indicate that the truncated form is much more stable than the full-length protein. CONCLUSIONS: The results suggest a model in which proteasome activity gives rise to a stable, hypophosphorylated and truncated form of 4E-BP1, which may exert a long-term inhibitory effect on the availability of eIF4E, thus contributing to the inhibition of protein synthesis and the growth-inhibitory and pro-apoptotic effects of p53.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Leukemia, Erythroblastic, Acute/metabolism , Phosphoproteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Tumor Suppressor Protein p53/metabolism , Adaptor Proteins, Signal Transducing/drug effects , Animals , Cell Cycle Proteins , Cell Line, Tumor , Mice , Phosphoproteins/drug effects , Phosphorylation , Tumor Suppressor Protein p53/pharmacology
9.
FEBS J ; 273(16): 3698-708, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16911520

ABSTRACT

Tumour cells are often sensitized by interferons to the effects of tumour necrosis factor-alpha-related apoptosis-inducing ligand (TRAIL). We have demonstrated previously that TRAIL has an inhibitory effect on protein synthesis [Jeffrey IW, Bushell M, Tilleray VJ, Morley S & Clemens MJ (2002) Cancer Res62, 2272-2280] and we have therefore examined the consequences of prior interferon-alpha treatment for the sensitivity of translation to inhibition by TRAIL. Interferon treatment alone has only a minor effect on protein synthesis but it sensitizes both MCF-7 cells and HeLa cells to the downregulation of translation by TRAIL. The inhibition of translation is characterized by increased phosphorylation of the alpha subunit of eukaryotic initiation factor eIF2 and dephosphorylation of the eIF4E-binding protein 4E-BP1. Both of these effects, as well as the decrease in overall protein synthesis, require caspase-8 activity, although they precede overt apoptosis by several hours. Interferon-alpha enhances the level and/or the extent of activation of caspase-8 by TRAIL, thus providing a likely explanation for the sensitization of cells to the inhibition of translation.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/pharmacology , Interferon-alpha/pharmacology , Membrane Glycoproteins/pharmacology , Protein Biosynthesis/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Caspase 8 , Caspases/metabolism , Cell Line, Tumor , Down-Regulation/drug effects , Drug Synergism , Humans , TNF-Related Apoptosis-Inducing Ligand
10.
FEBS Lett ; 580(7): 1766-70, 2006 Mar 20.
Article in English | MEDLINE | ID: mdl-16504179

ABSTRACT

Activation of an over-expressed mutant form of the tumour suppressor protein p53 has been shown to inhibit protein synthesis. To determine whether this effect is due only to high level expression or the mutant nature of the protein, we have used a doxycycline-inducible lung carcinoma cell line capable of expressing wild-type p53. We now show that levels of wild-type p53 similar to those expressed endogenously also inhibit protein synthesis. The mechanism involves dephosphorylation and accumulation of the translational inhibitor 4E-BP1, and increased association of 4E-BP1 with initiation factor eIF4E. The inhibition of translation is not a consequence of p53-mediated apoptosis.


Subject(s)
Lung Neoplasms/pathology , Protein Biosynthesis , Tumor Suppressor Protein p53/physiology , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Cell Cycle Proteins , Cell Line, Tumor , Doxycycline , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Regulation , Humans , Phosphoproteins/metabolism , Tumor Suppressor Protein p53/genetics
11.
Int J Biochem Cell Biol ; 38(2): 164-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16271499

ABSTRACT

Epstein-Barr virus is a potent mitogen for human B lymphocytes and is associated with a large number of human malignancies. This large virus expresses several genes that may contribute to the transformed phenotype of infected cells and it possesses multiple strategies for the inhibition of apoptosis. The interferon-inducible protein kinase PKR is an important target for the anti-apoptotic actions of the virus and its activity is regulated by the small untranslated RNA, EBER-1. This review summarizes the mechanisms of action of EBER-1 and other Epstein-Barr virus gene products in the regulation of apoptosis, and presents a model of how EBER-1 exerts its effects on PKR activity.


Subject(s)
Apoptosis/physiology , Cell Transformation, Neoplastic , Herpesvirus 4, Human/metabolism , B-Lymphocytes/metabolism , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/pathogenicity , Humans , Nucleic Acid Conformation , RNA, Viral/genetics , RNA, Viral/metabolism , eIF-2 Kinase/metabolism
12.
Oncogene ; 24(30): 4839-50, 2005 Jul 14.
Article in English | MEDLINE | ID: mdl-15897901

ABSTRACT

Activation of a temperature-sensitive form of mouse p53 in murine erythroleukaemia cells rapidly inhibits protein synthesis and causes early dephosphorylation and cleavage of protein synthesis initiation factor eIF4GI and the eIF4E-binding protein 4E-BP1. Dephosphorylated 4E-BP1 and the cleaved products of 4E-BP1 and eIF4GI associate with eIF4E under these conditions, concomitant with decreased interaction of full-length eIF4GI with eIF4E. These changes may play an important role in preventing formation of the eIF4F complex and thus the initiation of protein synthesis. As observed previously for eIF4GI, the cleavage of 4E-BP1 is insensitive to the general caspase inhibitor z-VAD.FMK, consistent with a caspase-independent mechanism of factor modification and regulation of protein synthesis. Comparison of the p53-induced patterns of eIF4GI and 4E-BP1 dephosphorylation and cleavage with those caused by the mTOR inhibitor rapamycin indicates that p53 activation and rapamycin have distinct but additive effects. Moreover, p53 activation inhibits rapamycin-insensitive protein kinase activity against 4E-BP1. P53 and rapamycin have additive effects on the inhibition of overall protein synthesis. These data suggest that the inhibition of protein synthesis by p53 is largely independent of the regulation of rapamycin-sensitive mTOR in the system under investigation.


Subject(s)
Carrier Proteins/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Phosphoproteins/metabolism , Protein Biosynthesis , Tumor Suppressor Protein p53/metabolism , Adaptor Proteins, Signal Transducing , Animals , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins , Eukaryotic Initiation Factor-4E/biosynthesis , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factors , Mice , Phosphoproteins/genetics , Phosphorylation/drug effects , Protein Kinases/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Temperature , Tumor Suppressor Protein p53/genetics
13.
Semin Cell Dev Biol ; 16(1): 13-20, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15659335

ABSTRACT

Protein synthesis is regulated at the translational level by a variety of mechanisms in virus-infected cells. Viruses often induce the shut-off of host translation in order to favour the expression of their own genetic information, but cells possess a number of strategies for counteracting such effects of infection. Important regulatory mechanisms include the phosphorylation of the alpha subunit of polypeptide chain initiation factor eIF2, RNA degradation mediated by the 2'5'-oligoadenylate/RNase L system, control of availability of the cap-binding protein eIF4E by its interaction with the 4E-binding proteins and specific proteolytic cleavage of several key initiation factors. Most of these mechanisms are also utilised in uninfected cells in response to a variety of physiological stresses and during the early stages of apoptosis. Thus, mechanisms of translational control during virus infection can provide models for the cellular stress responses observed in a wide range of other circumstances.


Subject(s)
Eukaryotic Cells/virology , Protein Biosynthesis , Viral Proteins/genetics , 2',5'-Oligoadenylate Synthetase/metabolism , Animals , Antineoplastic Agents/metabolism , Endoribonucleases/metabolism , Models, Genetic , Viral Proteins/biosynthesis , eIF-2 Kinase/metabolism
14.
Eur J Biochem ; 271(10): 1895-905, 2004 May.
Article in English | MEDLINE | ID: mdl-15128299

ABSTRACT

Epstein-Barr virus (EBV) is a potent mitogenic and antiapoptotic agent for B lymphocytes and is associated with several different types of human tumour. The abundantly expressed small viral RNA, EBER-1, binds to the growth inhibitory and pro-apoptotic protein kinase R (PKR) and blocks activation of the latter by double-stranded RNA. Recent evidence has suggested that expression of EBER-1 alone in EBV-negative B cells promotes a tumorigenic phenotype and that this may be related to inhibition of the pro-apoptotic effects of PKR. The ribosomal protein L22 binds to EBER-1 in virus-infected cells, but the significance of this has not previously been established. We report here that L22 and PKR compete for a common binding site on EBER-1. As a result of this competition, L22 interferes with the ability of the small RNA to inhibit the activation of PKR by dsRNA. Transient expression of EBER-1 in murine embryonic fibroblasts stimulates reporter gene expression and partially reverses the inhibitory effect of PKR. However, EBER-1 is also stimulatory when transfected into PKR knockout cells, suggesting an additional, PKR-independent, mode of action of the small RNA. Expression of L22 prevents both the PKR-dependent and -independent effects of EBER-1 in vivo. These results suggest that the association of L22 with EBER-1 in EBV-infected cells can attenuate the biological effects of the viral RNA. Such effects include both the inhibition of PKR and additional mechanism(s) by which EBER-1 stimulates gene expression.


Subject(s)
RNA, Viral/antagonists & inhibitors , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , Ribosomal Proteins/metabolism , Animals , Binding, Competitive , Cells, Cultured , Enzyme Activation/drug effects , Fibroblasts/metabolism , Gene Expression , Genes, Reporter , HeLa Cells , Herpesvirus 4, Human/genetics , Humans , Mice , RNA, Double-Stranded/metabolism , RNA, Double-Stranded/pharmacology , RNA, Viral/genetics , RNA-Binding Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribosomal Proteins/genetics , Transfection , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
15.
Oncogene ; 23(18): 3180-8, 2004 Apr 19.
Article in English | MEDLINE | ID: mdl-15094767

ABSTRACT

There is increasing evidence that deregulation of gene expression at the level of mRNA translation can contribute to cell transformation and the malignant phenotype. Two steps in the pathway of polypeptide chain initiation, viz. the assembly of the 43S initiation complex catalysed by polypeptide chain initiation factor eIF2 and the binding of eIF4E to eIF4G during the recruitment of mRNA to the ribosome, have been shown to be likely targets for changes associated with tumorigenesis. The activity of eIF2 is controlled by changes in phosphorylation of the alpha subunit of this factor. The availability of eIF4E for binding to eIF4G is regulated by the phosphorylation of a small family of eIF4E-binding proteins (the 4E-BPs). The activities of the protein kinases and/or phosphatases responsible for the (de)phosphorylation of these substrates may in turn be controlled by cellular and viral oncogenes and tumour-suppressor genes. This review will describe recent aspects of the mechanisms involved, with particular emphasis on the regulation of the eIF2 alpha kinase PKR and the control of 4E-BP phosphorylation by viral gene products, growth-inhibitory cytokines and the tumour-suppressor protein p53.


Subject(s)
Cell Transformation, Neoplastic/genetics , Protein Biosynthesis , Adaptor Proteins, Signal Transducing , Animals , Apoptosis , Carrier Proteins/physiology , Cell Cycle Proteins , Eukaryotic Initiation Factor-2/physiology , Eukaryotic Initiation Factor-4E/physiology , Eukaryotic Initiation Factor-4G/physiology , Humans , Phosphoproteins/physiology , Tumor Suppressor Protein p53/physiology , eIF-2 Kinase/physiology
16.
Eur J Biochem ; 270(15): 3122-32, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12869187

ABSTRACT

Activation of a temperature-sensitive form of p53 in murine erythroleukaemia cells results in a rapid impairment of protein synthesis that precedes inhibition of cell proliferation and loss of cell viability by several hours. The inhibition of translation is associated with specific cleavages of polypeptide chain initiation factors eIF4GI and eIF4B, a phenomenon previously observed in cells induced to undergo apoptosis in response to other stimuli. Although caspase activity is enhanced in the cells in which p53 is activated, both the effects on translation and the cleavages of the initiation factors are completely resistant to inhibition of caspase activity. Moreover, exposure of the cells to a combination of the caspase inhibitor z-VAD.FMK and the survival factor erythropoietin prevents p53-induced cell death but does not reverse the inhibition of protein synthesis. We conclude that the p53-regulated cleavages of eIF4GI and eIF4B, as well as the overall inhibition of protein synthesis, are caspase-independent events that can be dissociated from the induction of apoptosis per se.


Subject(s)
Apoptosis , Protein Biosynthesis , Tumor Suppressor Protein p53/metabolism , Animals , Caspases/metabolism , Cell Division , Eukaryotic Initiation Factor-4F/metabolism , Mice , Mutation , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
17.
J Interferon Cytokine Res ; 23(6): 277-92, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12859854

ABSTRACT

The interferons (IFNs), in addition to their well-known antiviral activities, have important roles in the control of cell proliferation and are effective agents for the treatment of a limited number of malignant diseases. IFNs not only regulate cell growth and division but also influence cell survival through their effects on apoptosis. This review describes the current state of knowledge about the mechanisms of action of these cytokines on the apoptotic machinery, with particular emphasis on the synergism that exists between the IFNs and other proapoptotic agents, such as members of the tumor necrosis factor (TNF) family. The review also discusses the physiologic and clinical implications of the effects of the IFNs on apoptosis for regulation of viral infection and tumor growth.


Subject(s)
Apoptosis , Interferons/physiology , Animals , Cell Differentiation , Humans , Interferons/genetics , Neoplasms/therapy , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction , Virus Diseases/therapy
18.
RNA ; 9(7): 858-70, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12810919

ABSTRACT

Translation of the hepatitis C genome is mediated by internal ribosome entry on the structurally complex 5' untranslated region of the large viral RNA. Initiation of protein synthesis by this mechanism is independent of the cap-binding factor eIF4E, but activity of the initiator Met-tRNA(f)-binding factor eIF2 is still required. HCV protein synthesis is thus potentially sensitive to the inhibition of eIF2 activity that can result from the phosphorylation of the latter by the interferon-inducible, double-stranded RNA-activated protein kinase PKR. Two virally encoded proteins, NS5A and E2, have been shown to reduce this inhibitory effect of PKR by impairing the activation of the kinase. Here we present evidence for a third viral strategy for PKR inhibition. A region of the viral RNA comprising part of the internal ribosome entry site (IRES) is able to bind to PKR in competition with double-stranded RNA and can prevent autophosphorylation and activation of the kinase in vitro. The HCV IRES itself has no PKR-activating ability. Consistent with these findings, cotransfection experiments employing a bicistronic reporter construct and wild-type PKR indicate that expression of the protein kinase is less inhibitory towards HCV IRES-driven protein synthesis than towards cap-dependent protein synthesis. These data suggest a dual function for the viral IRES, with both a structural role in promoting initiation complex formation and a regulatory role in preventing inhibition of initiation by PKR.


Subject(s)
Hepacivirus/physiology , RNA, Viral/genetics , Ribosomes/virology , eIF-2 Kinase/antagonists & inhibitors , Animals , Base Sequence , Cells, Cultured , HeLa Cells , Hepacivirus/genetics , Humans , Mice , Molecular Sequence Data , Nucleic Acid Conformation , Protein Binding , RNA, Viral/chemistry , RNA, Viral/metabolism , Transcription, Genetic , eIF-2 Kinase/isolation & purification
19.
Oncogene ; 21(34): 5325-34, 2002 Aug 08.
Article in English | MEDLINE | ID: mdl-12149653

ABSTRACT

p53 is an important regulator of cell cycle progression and apoptosis, and inactivation of p53 is associated with tumorigenesis. Although p53 exerts many of its effects through regulation of transcription, this protein is also found in association with ribosomes and several mRNAs have been identified that are translationally controlled in a p53-dependent manner. We have utilized murine erythroleukemic cells that express a temperature-sensitive p53 protein to determine whether p53 also functions at the level of translation. The data presented here demonstrate that p53 causes a rapid decrease in translation initiation. Analysis of several potential mechanisms for regulating protein synthesis shows that p53 has selective effects on the phosphorylation of the eIF4E-binding protein, 4E-BP1, and the activity of the p70 ribosomal protein S6 kinase. These data provide evidence that modulation of translational activity constitutes a further mechanism by which the growth inhibitory effects of p53 may be mediated.


Subject(s)
Carrier Proteins/metabolism , Leukemia, Erythroblastic, Acute/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases , Ribosomal Protein S6 Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Adaptor Proteins, Signal Transducing , Amino Acids/metabolism , Carrier Proteins/genetics , Cell Cycle Proteins , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , DNA-Binding Proteins/metabolism , Erythrocytes/cytology , Erythrocytes/metabolism , Eukaryotic Initiation Factor-4E , Gene Expression Regulation , Humans , Peptide Initiation Factors/genetics , Peptide Initiation Factors/metabolism , Phosphoproteins/genetics , Phosphorylation , Protein Biosynthesis , Protein Kinases , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Repressor Proteins/genetics , Repressor Proteins/metabolism , Ribosomes/metabolism , TOR Serine-Threonine Kinases , Temperature , Transcription Factors/metabolism
20.
Virology ; 297(2): 253-69, 2002 Jun 05.
Article in English | MEDLINE | ID: mdl-12083824

ABSTRACT

Recent studies have suggested a role for the Epstein-Barr virus-encoded RNA EBER-1 in malignant transformation. EBER-1 inhibits the activity of the protein kinase PKR, an inhibitor of protein synthesis with tumour suppressor properties. In human 293 cells and murine embryonic fibroblasts, transient expression of EBER-1 promoted total protein synthesis and enhanced the expression of cotransfected reporter genes. However reporter gene expression was stimulated equally well in cells from control and PKR knockout mice. NIH 3T3 cells stably expressing EBER-1 exhibited a greatly increased frequency of colony formation in soft agar, and protein synthesis in these cells was relatively resistant to inhibition by the calcium ionophore A23187. Nevertheless clones containing a high concentration of EBER-1 were not invariably tumourigenic. We conclude that EBER-1 can enhance protein synthesis by a PKR-independent mechanism and that, although this RNA may contribute to the oncogenic potential of Epstein-Barr virus, its expression is not always sufficient for malignant transformation.


Subject(s)
Fibroblasts/cytology , Fibroblasts/virology , Herpesvirus 4, Human/pathogenicity , Protein Biosynthesis , RNA, Viral/physiology , Animals , Cell Division , Cell Line , Cell Line, Transformed , Cell Transformation, Viral , Fibroblasts/metabolism , Gene Expression Regulation , Humans , Mice , Neoplasms/physiopathology , Transfection , eIF-2 Kinase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...