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










Publication year range
1.
Learn Mem ; 26(9): 332-342, 2019 09.
Article in English | MEDLINE | ID: mdl-31416906

ABSTRACT

Formation of eukaryotic initiation factor 4F (eIF4F) is widely considered to be the rate-limiting step in cap-dependent translation initiation. Components of eIF4F are often up-regulated in various cancers, and much work has been done to elucidate the role of each of the translation initiation factors in cancer cell growth and survival. In fact, many of the basic mechanisms describing how eIF4F is assembled and how it functions to regulate translation initiation were first investigated in cancer cell lines. These same eIF4F translational control pathways also are relevant for neuronal signaling that underlies long-lasting synaptic plasticity and memory, and in neurological diseases where eIF4F and its upstream regulators are dysregulated. Although eIF4F is important in cancer and for brain function, there is not always a clear path to use the results of studies performed in cancer models to inform one of the roles that the same translation factors have in neuronal signaling. Issues arise when extrapolating from cell lines to tissue, and differences are likely to exist in how eIF4F and its upstream regulatory pathways are expressed in the diverse neuronal subtypes found in the brain. This review focuses on summarizing the role of eIF4F and its accessory proteins in cancer, and how this information has been utilized to investigate neuronal signaling, synaptic function, and animal behavior. Certain aspects of eIF4F regulation are consistent across cancer and neuroscience, whereas some results are more complicated to interpret, likely due to differences in the complexity of the brain, its billions of neurons and synapses, and its diverse cell types.


Subject(s)
Brain Neoplasms/genetics , Brain/metabolism , Eukaryotic Initiation Factor-4F/biosynthesis , Gene Expression Regulation, Neoplastic , Protein Biosynthesis , Animals , Humans , RNA, Messenger/genetics , Signal Transduction
2.
Mol Pain ; 7: 70, 2011 Sep 21.
Article in English | MEDLINE | ID: mdl-21936900

ABSTRACT

Neuropathic pain is a debilitating clinical condition with few efficacious treatments, warranting development of novel therapeutics. We hypothesized that dysregulated translation regulation pathways may underlie neuropathic pain. Peripheral nerve injury induced reorganization of translation machinery in the peripheral nervous system of rats and mice, including enhanced mTOR and ERK activity, increased phosphorylation of mTOR and ERK downstream targets, augmented eIF4F complex formation and enhanced nascent protein synthesis. The AMP activated protein kinase (AMPK) activators, metformin and A769662, inhibited translation regulation signaling pathways, eIF4F complex formation, nascent protein synthesis in injured nerves and sodium channel-dependent excitability of sensory neurons resulting in a resolution of neuropathic allodynia. Therefore, injury-induced dysregulation of translation control underlies pathology leading to neuropathic pain and reveals AMPK as a novel therapeutic target for the potential treatment of neuropathic pain.


Subject(s)
Molecular Targeted Therapy , Neuralgia/drug therapy , Neuralgia/enzymology , Protein Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Action Potentials/drug effects , Animals , Blotting, Western , Disease Models, Animal , Enzyme Activators/pharmacology , Enzyme Activators/therapeutic use , Eukaryotic Initiation Factor-4F/biosynthesis , Hyperalgesia/complications , Hyperalgesia/drug therapy , Hyperalgesia/pathology , Male , Mice , Mice, Inbred ICR , Neuralgia/physiopathology , Protein Biosynthesis/drug effects , Rats , Rats, Sprague-Dawley , Sciatic Nerve/drug effects , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/pathology , Signal Transduction/drug effects
3.
Proc Natl Acad Sci U S A ; 108(3): 1046-51, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21191102

ABSTRACT

Deregulation of cap-dependent translation is associated with cancer initiation and progression. The rate-limiting step of protein synthesis is the loading of ribosomes onto mRNA templates stimulated by the heterotrimeric complex, eukaryotic initiation factor (eIF)4F. This step represents an attractive target for anticancer drug discovery because it resides at the nexus of the TOR signaling pathway. We have undertaken an ultra-high-throughput screen to identify inhibitors that prevent assembly of the eIF4F complex. One of the identified compounds blocks interaction between two subunits of eIF4F. As a consequence, cap-dependent translation is inhibited. This compound can reverse tumor chemoresistance in a genetically engineered lymphoma mouse model by sensitizing cells to the proapoptotic action of DNA damage. Molecular modeling experiments provide insight into the mechanism of action of this small molecule inhibitor. Our experiments validate targeting the eIF4F complex as a strategy for cancer therapy to modulate chemosensitivity.


Subject(s)
Benzoates/pharmacology , Eukaryotic Initiation Factor-4F/antagonists & inhibitors , Lymphoma/metabolism , Models, Molecular , Protein Biosynthesis/physiology , Animals , Blotting, Western , Eukaryotic Initiation Factor-4F/biosynthesis , Eukaryotic Initiation Factor-4G/antagonists & inhibitors , High-Throughput Screening Assays , Lymphoma/drug therapy , Mice , Protein Binding
4.
Cancer Res ; 69(19): 7491-4, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19773439

ABSTRACT

The Myc/Max/Mad family of transcription factors and the eukaryotic initiation factor 4F (4F) complex play fundamental roles in regulating cell growth, proliferation, differentiation, and oncogenic transformation. Recent findings indicate that the role of Myc during cell growth and proliferation is linked to an increase in eIF4F activity in a feedforward relationship, providing a possible molecular mechanism of cell transformation by Myc. Developing therapeutics to inhibit eIF4F and/or Myc could be a potential treatment for a wide range of human cancers.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Eukaryotic Initiation Factor-4F/antagonists & inhibitors , Eukaryotic Initiation Factor-4F/metabolism , Neoplasms/metabolism , Neoplasms/therapy , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Growth Processes/physiology , Eukaryotic Initiation Factor-4F/biosynthesis , Eukaryotic Initiation Factor-4F/genetics , Humans
5.
Am J Physiol Endocrinol Metab ; 292(6): E1647-55, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17284572

ABSTRACT

An important function of growth hormone (GH) is to promote cell and tissue growth, and a key component of these effects is the stimulation of protein synthesis. In this study, we demonstrate that, in H4IIE hepatoma cells, GH acutely activated protein synthesis through signaling via the mammalian target of rapamycin (mTOR) and specifically through the rapamycin-sensitive mTOR complex 1 (mTORC1). GH treatment enhanced the phosphorylation of two targets of mTOR signaling, 4E-BP1 and ribosomal protein S6. Phosphorylation of S6 and 4E-BP1 was maximal at 30-45 min and 10-20 min after GH stimulation, respectively. Both proteins modulate components of the translational machinery. The GH-induced phosphorylation of 4E-BP1 led to its dissociation from eIF4E and increased binding of eIF4E to eIF4G to form (active) eIF4F complexes. The ability of GH to stimulate the phosphorylation of S6 and 4E-BP1 was blocked by rapamycin. GH also led to the dephosphorylation of a third translational component linked to mTORC1, the elongation factor eEF2. Its regulation followed complex biphasic kinetics, both phases of which required mTOR signaling. GH rapidly activated both the MAP kinase (ERK) and PI 3-kinase pathways. Signaling through PI 3-kinase alone was, however, sufficient to activate the downstream mTORC1 pathway. Consistent with this, GH increased the phosphorylation of TSC2, an upstream regulator of mTORC1, at sites that are targets for Akt/PKB. Finally, the activation of overall protein synthesis by GH in H4IIE cells was essentially completely inhibited by wortmannin or rapamycin. These results demonstrate for the first time that mTORC1 plays a major role in the rapid activation of protein synthesis by GH.


Subject(s)
Growth Hormone/pharmacology , Protein Biosynthesis/drug effects , Transcription Factors/metabolism , Animals , Carrier Proteins/metabolism , Cell Line, Tumor , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4F/biosynthesis , Eukaryotic Initiation Factor-4G/metabolism , Growth Hormone/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins , Kinetics , Peptide Elongation Factor 2/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Rats , Recombinant Proteins/pharmacology , Ribosomal Protein S6/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Sirolimus/pharmacology , Time Factors
6.
Genes Dev ; 20(4): 461-72, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16481474

ABSTRACT

Recruitment of the 40S ribosome to the 5' end of a eukaryotic mRNA requires assembly of translation initiation factors eIF4E, the cap-binding protein, together with eIF4A and eIF4G into a complex termed eIF4F. While the translational repressor 4E-BP1 regulates binding of eIF4E to eIF4G, the forces required to construct an eIF4F complex remain unidentified. Here, we establish that the herpes simplex virus-1 (HSV-1) ICP6 polypeptide associates with eIF4G to promote eIF4F complex assembly. Strikingly, release of eIF4E from the 4E-BP1 repressor is insufficient to drive complex formation, suggesting that ICP6 is an eIF4F-assembly chaperone. This is the first example of a translation initiation factor-associated protein that promotes active complex assembly and defines a new, controllable step in the initiation of translation. Homology of the N-terminal, eIF4G-binding segment of ICP6 with cellular chaperones suggest that factors capable of interacting with eIF4G and promoting eIF4F complex assembly may play important roles in a variety of processes where translation complexes need to be remodeled or assembled on populations of newly synthesized or derepressed mRNAs, including development, differentiation, and the response to a broad spectrum of environmental cues.


Subject(s)
Eukaryotic Initiation Factor-4F/biosynthesis , Eukaryotic Initiation Factor-4G/metabolism , Herpesvirus 1, Human/metabolism , Molecular Chaperones/metabolism , Viral Proteins/metabolism , Cell Line , Chromatography, Agarose , Cloning, Molecular , Humans , Immunoblotting , Immunoprecipitation
7.
Am J Physiol Endocrinol Metab ; 290(4): E631-42, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16263769

ABSTRACT

Feeding promotes protein accretion in skeletal muscle through a stimulation of the mRNA translation initiation phase of protein synthesis either secondarily to nutrient-induced rises in insulin or owing to direct effects of nutrients themselves. The present set of experiments establishes the effects of meal feeding on potential signal transduction pathways that may be important in accelerating mRNA translation initiation. Gastrocnemius muscle from male Sprague-Dawley rats trained to consume a meal consisting of rat chow was sampled before, during, and after the meal. Meal feeding enhanced the assembly of the active eIF4G.eIF4E complex, which returned to basal levels within 3 h of removal of food. The increased assembly of the active eIF4G.eIF4E complex was associated with a marked 10-fold rise in phosphorylation of eIF4G(Ser(1108)) and a decreased assembly of inactive 4E-BP1.eIF4E complex. The reduced assembly of 4E-BP1.eIF4E complex was associated with a 75-fold increase in phosphorylation of 4E-BP1 in the gamma-form during feeding. Phosphorylation of S6K1 on Ser(789) was increased by meal feeding, although the extent of phosphorylation was greater at 0.5 h after feeding than after 1 h. Phosphorylation of mammalian target of rapamycin (mTOR) on Ser(2448) or Ser(2481), an upstream kinase responsible for phosphorylating both S6K1 and 4E-BP1, was increased at all times during meal feeding, although the extent of phosphorylation was greater at 0.5 h after feeding than after 1 h. Phosphorylation of PKB, an upstream kinase responsible for phosphorylating mTOR, was elevated only after 0.5 h of meal feeding for Thr(308), whereas phosphorylation Ser(473) was significantly elevated at only 0.5 and 1 h after initiation of feeding. We conclude from these studies that meal feeding stimulates two signal pathways in skeletal muscle that lead to elevated eIF4G.eIF4E complex assembly through increased phosphorylation of eIF4G and decreased association of 4E-BP1 with eIF4E.


Subject(s)
Eating/physiology , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4F/biosynthesis , Eukaryotic Initiation Factor-4G/metabolism , Muscle, Skeletal/metabolism , Amino Acids/blood , Animals , Blotting, Western , Carrier Proteins/metabolism , Eukaryotic Initiation Factor-4F/metabolism , Insulin/blood , Intracellular Signaling Peptides and Proteins , Male , Phosphoproteins/metabolism , Phosphorylation , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases/metabolism , TOR Serine-Threonine Kinases , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism
8.
Mol Biochem Parasitol ; 140(1): 23-41, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15694484

ABSTRACT

In eukaryotes protein synthesis initiates with the binding of the multimeric translation initiation complex eIF4F - eIF4E, eIF4A and eIF4G - to the monomethylated cap present on the 5' end of mRNAs. eIF4E interacts directly with the cap nucleotide, while eIF4A is a highly conserved RNA helicase and eIF4G acts as a scaffold for the complex with binding sites for both eIF4E and eIF4A. eIF4F binding to the mRNA recruits the small ribosomal subunit to its 5' end. Little is known in detail of protein synthesis in the protozoan parasites belonging to the family Trypanosomatidae. However, the presence of the highly modified cap structure, cap4, and the spliced leader sequence on the 5' ends of all mRNAs suggests possible differences in mRNA recruitment by ribosomes. We identified several potential eIF4F homologues by searching Leishmania major databases: four eIF4Es (LmEIF4E1-4), two eIF4As (LmEIF4A1-2) and five eIF4Gs (LmEIF4G1-5). We report the initial characterisation of LmEIF4E1-3, LmEIF4A1-2 and LmEIF4G3. First, the expression of these proteins in L. major promastigotes was quantitated by Western blotting using isoform specific antibodies. LmEIF4A1 and LmEIF4E3 are very abundant, LmEIF4G3 is moderately abundant and LmEIF4E1/LmEIF4E2/LmEIF4A2 are rare or not detected. In cap-binding assays, only LmEIF4E1 bound to the 7-methyl-GTP-Sepharose resin. Molecular modelling confirmed that LmEIF4E1 has all the structural features of a cap-binding protein. Finally, pull-down assays were used to investigate the potential interaction between the eIF4A (LmEIF4A1/LmEIF4A2) and eIF4G (LmEIF4G1-3) homologues. Only LmEIF4G3, via the HEAT domain, bound specifically both to LmEIF4A1 as well as to human eIF4A. Therefore for each factor, one of the L. major forms seems to fulfil, in part at least, the expected characteristics of a translational initiation factor.


Subject(s)
Eukaryotic Initiation Factor-4F/biosynthesis , Leishmania major/metabolism , Amino Acid Sequence , Animals , Cell Cycle Proteins , Cloning, Molecular , Eukaryotic Initiation Factor-4F/genetics , Intracellular Signaling Peptides and Proteins , Leishmania major/genetics , Leishmania major/growth & development , Life Cycle Stages , Molecular Sequence Data , Protein Structure, Tertiary/genetics , Protein Subunits/biosynthesis , Protein Subunits/chemistry , Protein Subunits/genetics , Proteins , RNA, Messenger/metabolism , RNA, Protozoan/metabolism , Sequence Alignment
9.
Am J Physiol Gastrointest Liver Physiol ; 287(3): G667-75, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15117679

ABSTRACT

To determine the mechanism of meal-regulated synthesis of pancreatic digestive enzymes, we studied the effect of fasting and refeeding on pancreatic protein synthesis, relative mRNA levels of digestive enzymes, and activation of the translational machinery. With the use of the flooding dose technique with L-[3H]phenylalanine, morning protein synthesis in the pancreas of Institute for Cancer Research mice fed ad libitum was 7.9 +/- 0.3 nmol phenylalanine.10 min(-1).mg protein(-1). Prior fasting for 18 h reduced total protein synthesis to 70 +/- 1.4% of this value. Refeeding for 2 h, during which the mice consumed 29% of their daily food intake, increased protein synthesis to 117.3 +/- 4.9% of the control level. Pancreatic mRNA levels of amylase, lipases, trypsins, chymotrypsin, elastases, as well as those for several housekeeping genes tested were not significantly changed after refeeding compared with fasted mice. By contrast, the major translational control pathway involving Akt, mTOR, and S6K was strongly regulated by fasting and refeeding. Fasting for 18 h decreased phosphorylation of ribosomal protein S6 to almost undetectable levels, and refeeding highly increased it. The most highly phosphorylated form of the eIF4E binding protein (4E-BP1) made up the 14.6% of total 4E-BP1 in normally fed animals, was only 2.8% after fasting, and was increased to 21.4% after refeeding. This was correlated with an increase in the formation of the eIF4E-eIF4G complex after refeeding. By contrast, feeding did not affect eIF2B activity. Thus food intake stimulates pancreatic protein synthesis and translational effectors without increasing digestive enzyme mRNA levels.


Subject(s)
Eating/physiology , Pancreas/metabolism , Protein Biosynthesis/physiology , RNA, Messenger/biosynthesis , Animals , DNA Primers , Eukaryotic Initiation Factor-4F/biosynthesis , Eukaryotic Initiation Factor-4F/genetics , Male , Mice , Mice, Inbred ICR , Pancreas/enzymology , Phosphorylation , Precipitin Tests , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptors, G-Protein-Coupled/physiology , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Protein S6 Kinases, 70-kDa/biosynthesis , Ribosomal Protein S6 Kinases, 70-kDa/genetics
10.
Am J Physiol Endocrinol Metab ; 285(1): E40-53, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12637260

ABSTRACT

Previous studies have shown that intravenous infusion of insulin and/or amino acids reproduces the feeding-induced stimulation of muscle protein synthesis in neonates and that insulin and amino acids act independently to produce this effect. The goal of the present study was to delineate the regulatory roles of insulin and amino acids on muscle protein synthesis in neonates by examining translational control mechanisms, specifically the eukaryotic translation initiation factors (eIFs), which enable coupling of initiator methionyl-tRNAi and mRNA to the 40S ribosomal subunit. Insulin secretion was blocked by somatostatin in fasted 7-day-old pigs (n = 8-12/group), insulin was infused to achieve plasma levels of approximately 0, 2, 6, and 30 microU/ml, and amino acids were clamped at fasting or fed levels or, at the high insulin dose, below fasting. Both insulin and amino acids increased the phosphorylation of ribosomal protein S6 kinase (S6K1) and the eIF4E-binding protein (4E-BP1), decreased the binding of 4E-BP1 to eIF4E, increased eIF4E binding to eIF4G, and increased fractional protein synthesis rates but did not affect eIF2B activity. In the absence of insulin, amino acids had no effect on these translation initiation factors but increased the protein synthesis rates. Raising insulin from below fasting to fasting levels generally did not alter translation initiation factor activity but raised protein synthesis rates. The phosphorylation of S6K1 and 4E-BP1 and the amount of 4E-BP1 bound to eIF4E and eIF4E bound to eIF4G were correlated with insulin level, amino acid level, and protein synthesis rate. Thus insulin and amino acids regulate muscle protein synthesis in skeletal muscle of neonates by modulating the availability of eIF4E for 48S ribosomal complex assembly, although other processes also must be involved.


Subject(s)
Amino Acids/pharmacology , Animals, Newborn/physiology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Muscle Proteins/biosynthesis , Muscle, Skeletal/metabolism , Protein Biosynthesis/drug effects , Algorithms , Amino Acids/metabolism , Amino Acids, Branched-Chain/metabolism , Animals , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4F/biosynthesis , Eukaryotic Initiation Factor-4F/genetics , Female , Glucose Clamp Technique , Immunoblotting , Muscle, Skeletal/drug effects , Pancrelipase/physiology , Phosphorylation , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...