Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
J Biol Chem ; 290(29): 17822-17837, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26041779

ABSTRACT

Cells respond to shrinkage induced by increased extracellular osmolarity via programmed changes in gene transcription and mRNA translation. The immediate response to this stress includes the induction of expression of the neutral amino acid transporter SNAT2. Increased SNAT2-mediated uptake of neutral amino acids is an essential adaptive mechanism for restoring cell volume. In contrast, stress-induced phosphorylation of the α subunit of the translation initiation factor eIF2 (eIF2α) can promote apoptosis. Here we show that the response to mild hyperosmotic stress involves regulation of the phosphorylation of eIF2α by increased levels of GADD34, a regulatory subunit of protein phosphatase 1 (PP1). The induction of GADD34 was dependent on transcriptional control by the c-Jun-binding cAMP response element in the GADD34 gene promoter and posttranscriptional stabilization of its mRNA. This mechanism differs from the regulation of GADD34 expression by other stresses that involve activating transcription factor 4 (ATF4). ATF4 was not translated during hyperosmotic stress despite an increase in eIF2α phosphorylation. The SNAT2-mediated increase in amino acid uptake was enhanced by increased GADD34 levels in a manner involving decreased eIF2α phosphorylation. It is proposed that the induction of the SNAT2/GADD34 axis enhances cell survival by promoting the immediate adaptive response to stress.


Subject(s)
Amino Acid Transport System A/metabolism , Osmotic Pressure , Protein Phosphatase 1/metabolism , Animals , Cell Line , Cell Survival , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Mice , Phosphorylation , Promoter Regions, Genetic , Protein Phosphatase 1/genetics
2.
J Biol Chem ; 288(24): 17202-13, 2013 Jun 14.
Article in English | MEDLINE | ID: mdl-23645676

ABSTRACT

Endoplasmic reticulum (ER) stress-induced responses are associated with the loss of insulin-producing ß-cells in type 2 diabetes mellitus. ß-Cell survival during ER stress is believed to depend on decreased protein synthesis rates that are mediated via phosphorylation of the translation initiation factor eIF2α. It is reported here that chronic ER stress correlated with increased islet protein synthesis and apoptosis in ß-cells in vivo. Paradoxically, chronic ER stress in ß-cells induced an anabolic transcription program to overcome translational repression by eIF2α phosphorylation. This program included expression of amino acid transporter and aminoacyl-tRNA synthetase genes downstream of the stress-induced ATF4-mediated transcription program. The anabolic response was associated with increased amino acid flux and charging of tRNAs for branched chain and aromatic amino acids (e.g. leucine and tryptophan), the levels of which are early serum indicators of diabetes. We conclude that regulation of amino acid transport in ß-cells during ER stress involves responses leading to increased protein synthesis, which can be protective during acute stress but can lead to apoptosis during chronic stress. These studies suggest that the increased expression of amino acid transporters in islets can serve as early diagnostic biomarkers for the development of diabetes.


Subject(s)
Amino Acids/metabolism , Apoptosis , Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum Stress , Insulin-Secreting Cells/physiology , Activating Transcription Factor 4/metabolism , Amino Acid Transport Systems/genetics , Amino Acid Transport Systems/metabolism , Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , Animals , Cell Survival , Diabetes Mellitus, Type 2/pathology , Eukaryotic Initiation Factor-2/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Biosynthesis , Protein Processing, Post-Translational , RNA, Transfer/metabolism , Transcriptional Activation
3.
J Biol Chem ; 285(22): 17098-111, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20338999

ABSTRACT

Regulation of cell volume is of great importance because persistent swelling or shrinkage leads to cell death. Tissues experience hypertonicity in both physiological (kidney medullar cells) and pathological states (hypernatremia). Hypertonicity induces an adaptive gene expression program that leads to cell volume recovery or apoptosis under persistent stress. We show that the commitment to apoptosis is controlled by phosphorylation of the translation initiation factor eIF2alpha, the master regulator of the stress response. Studies with cultured mouse fibroblasts and cortical neurons show that mutants deficient in eIF2alpha phosphorylation are protected from hypertonicity-induced apoptosis. A novel link is revealed between eIF2alpha phosphorylation and the subcellular distribution of the RNA-binding protein heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1). Stress-induced phosphorylation of eIF2alpha promotes apoptosis by inducing the cytoplasmic accumulation of hnRNP A1, which attenuates internal ribosome entry site-mediated translation of anti-apoptotic mRNAs, including Bcl-xL that was studied here. Hypertonic stress induced the eIF2alpha phosphorylation-independent formation of cytoplasmic stress granules (SGs, structures that harbor translationally arrested mRNAs) and the eIF2alpha phosphorylation-dependent accumulation of hnRNP A1 in SGs. The importance of hnRNP A1 was demonstrated by induction of apoptosis in eIF2alpha phosphorylation-deficient cells that express exogenous cytoplasmic hnRNP A1. We propose that eIF2alpha phosphorylation during hypertonic stress promotes apoptosis by sequestration of specific mRNAs in SGs in a process mediated by the cytoplasmic accumulation of hnRNP A1.


Subject(s)
Apoptosis , Eukaryotic Initiation Factor-2/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Osmosis , Animals , Cytoplasm/metabolism , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterozygote , Mice , Microscopy, Fluorescence/methods , Models, Biological , Osmotic Pressure , Phosphorylation , Plasmids/metabolism , RNA, Messenger/metabolism , Signal Transduction
4.
Biochem J ; 429(1): 215-24, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20408811

ABSTRACT

Expression of the Cat-1 gene (cationic amino acid transporter-1) is induced in proliferating cells and in response to a variety of stress conditions. The expression of the gene is mediated via a TATA-less promoter. In the present study we show that an Sp1 (specificity protein 1)-binding site within a GC-rich region of the Cat-1 gene controls its basal expression and is important for induction of the gene during the UPR (unfolded protein response). We have shown previously that induction of Cat-1 gene expression during the UPR requires phosphorylation of the translation initiation factor eIF2alpha (eukaryotic initiation factor 2alpha) by PERK (protein-kinase-receptor-like endoplasmic reticulum kinase), one of the signalling pathways activated during the UPR. This leads to increased translation of the transcription factor ATF4 (activating transcription factor 4). We also show that a second signalling pathway is required for sustained transcriptional induction of the Cat-1 gene during the UPR, namely activation of IRE1 (inositol-requiring enzyme 1) leading to alternative splicing of the mRNA for the transcription factor XBP1 (X-box-binding protein 1). The resulting XBP1s (spliced XBP1) can bind to an ERSE (endoplasmic-reticulum-stress-response-element), ERSE-II-like, that was identified within the Cat-1 promoter. Surprisingly, eIF2alpha phosphorylation is required for accumulation of XBP1s. We propose that the signalling via phosphorylated eIF2alpha is required for maximum induction of Cat-1 transcription during the UPR by inducing the accumulation of both ATF4 and XBP1s.


Subject(s)
Cationic Amino Acid Transporter 1/physiology , Endoplasmic Reticulum/physiology , Stress, Physiological/physiology , Transcription, Genetic/physiology , Animals , Base Sequence , Fibroblasts/physiology , Mice , Molecular Sequence Data , Rats , Time Factors
5.
J Biol Chem ; 284(47): 32312-20, 2009 Nov 20.
Article in English | MEDLINE | ID: mdl-19720825

ABSTRACT

Expression of the arginine/lysine transporter Cat-1 is highly induced in proliferating and stressed cells via mechanisms that include transcriptional activation. A bifunctional INE (intronic element) within the first intron of the Cat-1 gene was identified and characterized in this study. The INE had high sequence homology to an amino acid response element and was shown to act as a transcriptional enhancer in unstressed cells by binding the transcription factor, purine-rich element binding protein A (Pur alpha). During endoplasmic reticulum stress, binding of Pur alpha to the INE decreased; the element acted as a positive regulator in early stress by binding of the transcription factor ATF4 and as a negative regulator in prolonged stress by binding the stress-induced C/EBP family member, CHOP. We conclude that transcriptional control of the Cat-1 gene is tightly controlled by multiple cis-DNA elements, contributing to regulation of cationic amino acid transport for cell growth and proliferation. In addition, we propose that genes may use stress-response elements such as the INE to support basal expression in the absence of stress.


Subject(s)
Cationic Amino Acid Transporter 1/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation , Nerve Tissue Proteins/genetics , Transcription Factors/genetics , Activating Transcription Factor 4/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , DNA/chemistry , Endoplasmic Reticulum/metabolism , Enhancer Elements, Genetic , Humans , Introns , Mice , Rats , Transcription Factor CHOP/metabolism
6.
RNA ; 14(3): 593-602, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18230759

ABSTRACT

In vitro translation systems are used to investigate translational mechanisms and to synthesize proteins for characterization. Most available mammalian cell-free systems have reduced efficiency due to decreased translation initiation caused by phosphorylation of the initiation factor eIF2alpha on Ser51. We describe here a novel cell-free protein synthesis system using extracts from cultured mouse embryonic fibroblasts that are homozygous for the Ser51 to- Ala mutation in eIF2alpha (A/A cells). The translation efficiency of a capped and polyadenylated firefly luciferase mRNA in A/A cell extracts was 30-fold higher than in wild-type extracts. Protein synthesis in extracts from A/A cells was active for at least 2 h and generated up to 20 microg/mL of luciferase protein. Additionally, the A/A cell-free system faithfully recapitulated the selectivity of in vivo translation for mRNA features; translation was stimulated by a 5'-end cap (m7GpppN) and a 3'-end poly(A) tail in a synergistic manner. The system also showed similar efficiencies of cap-dependent and IRES-mediated translation (EMCV IRES). Significantly, the A/A cell-free system supported the post-translational modification of proteins, as shown by glycosylation of the HIV type-1 gp120 and cleavage of the signal peptide from beta-lactamase. We propose that cell-free systems from A/A cells can be a useful tool for investigating mechanisms of mammalian mRNA translation and for the production of recombinant proteins for molecular studies. In addition, cell-free systems from differentiated cells with the Ser51Ala mutation should provide a means for investigating cell type-specific features of protein synthesis.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Protein Biosynthesis , Amino Acid Substitution , Animals , Base Sequence , Cell-Free System , Eukaryotic Initiation Factor-2/genetics , In Vitro Techniques , Mice , Phosphorylation , Plasmids/genetics , Protein Processing, Post-Translational , RNA Caps/genetics , RNA Caps/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
7.
Biochem J ; 402(1): 163-73, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17042743

ABSTRACT

The adaptive response to amino acid limitation in mammalian cells inhibits global protein synthesis and promotes the expression of proteins that protect cells from stress. The arginine/lysine transporter, cat-1, is induced during amino acid starvation by transcriptional and post-transcriptional mechanisms. It is shown in the present study that the transient induction of cat-1 transcription is regulated by the stress response pathway that involves phosphorylation of the translation initiation factor, eIF2 (eukaryotic initiation factor-2). This phosphorylation induces expression of the bZIP (basic leucine zipper protein) transcription factors C/EBP (CCAAT/enhancer-binding protein)-beta and ATF (activating transcription factor) 4, which in turn induces ATF3. Transfection experiments in control and mutant cells, and chromatin immunoprecipitations showed that ATF4 activates, whereas ATF3 represses cat-1 transcription, via an AARE (amino acid response element), TGATGAAAC, in the first exon of the cat-1 gene, which functions both in the endogenous and in a heterologous promoter. ATF4 and C/EBPbeta activated transcription when expressed in transfected cells and they bound as heterodimers to the AARE in vitro. The induction of transcription by ATF4 was inhibited by ATF3, which also bound to the AARE as a heterodimer with C/EBPbeta. These results suggest that the transient increase in cat-1 transcription is due to transcriptional activation caused by ATF4 followed by transcriptional repression by ATF3 via a feedback mechanism.


Subject(s)
Amino Acids/metabolism , Arginine/metabolism , Cationic Amino Acid Transporter 1/genetics , Gene Expression Regulation , Lysine/metabolism , Activating Transcription Factor 3/metabolism , Activating Transcription Factor 4/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cationic Amino Acid Transporter 1/metabolism , Dimerization , Eukaryotic Initiation Factor-2/metabolism , Feedback, Physiological , Phosphorylation , Promoter Regions, Genetic , RNA, Messenger/metabolism , Rats , Transcription, Genetic , Transfection , Tumor Cells, Cultured
8.
Cell Rep ; 21(10): 2895-2910, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29212034

ABSTRACT

GADD34, a stress-induced regulatory subunit of the phosphatase PP1, is known to function in hyperosmotic stress through its well-known role in the integrated stress response (ISR) pathway. Adaptation to hyperosmotic stress is important for the health of corneal epithelial cells exposed to changes in extracellular osmolarity, with maladaptation leading to dry eye syndrome. This adaptation includes induction of SNAT2, an endoplasmic reticulum (ER)-Golgi-processed protein, which helps to reverse the stress-induced loss of cell volume and promote homeostasis through amino acid uptake. Here, we show that GADD34 promotes the processing of proteins synthesized on the ER during hyperosmotic stress independent of its action in the ISR. We show that GADD34/PP1 phosphatase activity reverses hyperosmotic-stress-induced Golgi fragmentation and is important for cis- to trans-Golgi trafficking of SNAT2, thereby promoting SNAT2 plasma membrane localization and function. These results suggest that GADD34 is a protective molecule for ocular diseases such as dry eye syndrome.


Subject(s)
Amino Acid Transport System A/metabolism , Protein Phosphatase 1/metabolism , Amino Acid Transport System A/genetics , Amino Acids/metabolism , Blotting, Western , Humans , Osmosis/physiology , Protein Phosphatase 1/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Reverse Transcriptase Polymerase Chain Reaction
9.
Int J Biochem Cell Biol ; 59: 135-41, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25541374

ABSTRACT

PERK, PKR, HRI and GCN2 are the four mammalian kinases that phosphorylate the α subunit of the eukaryotic translation initiation factor 2 (eIF2α) on Ser51. This phosphorylation event is conserved among many species and attenuates protein synthesis in response to diverse stress conditions. In contrast, Saccharmyces cerevisiae expresses only the GCN2 kinase. It was demonstrated previously in S. cerevisiae that single point mutations in eIF2α's N-terminus severely impaired phosphorylation at Ser51. To assess whether similar recognition patterns are present in mammalian eIF2α, we expressed human eIF2α's with these mutations in mouse embryonic fibroblasts and assessed their phosphorylation under diverse stress conditions. Some of the mutations prevented the stress-induced phosphorylation of eIF2α by all mammalian kinases, thus defining amino acid residues in eIF2α (Gly 30, Leu 50, and Asp 83) that are required for substrate recognition. We also identified residues that were less critical or not required for recognition by the mammalian kinases (Ala 31, Met 44, Lys 79, and Tyr 81), even though they were essential for recognition of the yeast eIF2α by GCN2. We propose that mammalian eIF2α kinases evolved to maximize their interactions with the evolutionarily conserved Ser51 residue of eIF2α in response to diverse stress conditions, thus adding to the complex signaling pathways that mammalian cells have over simpler organisms.


Subject(s)
Amino Acids/metabolism , Eukaryotic Initiation Factor-2/chemistry , Eukaryotic Initiation Factor-2/metabolism , Saccharomyces cerevisiae/metabolism , Stress, Physiological , Animals , Endoplasmic Reticulum Stress/drug effects , HEK293 Cells , Humans , Hypertonic Solutions/pharmacology , Mice , Models, Molecular , Mutant Proteins/metabolism , Mutation/genetics , Oxidative Stress/drug effects , Phosphorylation/drug effects , Poly I-C/pharmacology , Reproducibility of Results , Stress, Physiological/drug effects , Structure-Activity Relationship
10.
Adv Nutr ; 5(5): 558-60, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25469394

ABSTRACT

Responding to nutrient availability is an important homeostatic mechanism in the growth, development, and function of cells and tissues. However, these adaptations can also play a role in the development of disease. Our symposium, "Cellular Responses to Nutrients and Development of Disease," presented research about how cells sense nutrients and how the resulting signal transduction controls cellular processes from gene transcription to impacting various pathophysiologic processes. Dr. Michael Kilberg discussed the transcription program triggered by amino acid limitation that leads to growth arrest in normal cells and sustained growth in tumor cells. Dr. Noa Noy elaborated on the role of lipid-binding proteins in retinoic acid signaling, focusing on fatty acid-binding protein 5 (FABP5), which promotes cell growth by delivering this molecule to the nuclear receptor peroxisome proliferator-activated receptor δ (PPARδ). Dr. Li-Na Wei discussed the many functions of the protein receptor interacting protein 140 (RIP140) as a coregulator of nuclear receptors and as a cytoplasmic protein that regulates insulin-stimulated glucose uptake, lipolysis, and inflammation. Dr. Ruma Banerjee presented state-of-the-art approaches for studying the gaseous signaling molecule hydrogen sulfide (H2S), discussing its concentrations, metabolism, and functions in the regulation of redox signaling. Finally, Dr. Maria Hatzoglou described how the stress-induced increases in amino acid transport, mammalian target of rapamycin (mTOR) signaling, and protein synthesis in pancreatic ß-cells can contribute to the progression of diabetes.


Subject(s)
Amino Acids/pharmacology , Chronic Disease/drug therapy , Chronic Disease/prevention & control , Receptors, Cytoplasmic and Nuclear/drug effects , Signal Transduction , Animals , Cell Line , Congresses as Topic , Disease Models, Animal , Homeostasis , Humans , JNK Mitogen-Activated Protein Kinases , Protein Serine-Threonine Kinases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , TOR Serine-Threonine Kinases/metabolism , Transcription, Genetic
11.
Mol Cell Biol ; 32(5): 992-1003, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22215619

ABSTRACT

The accumulation of unfolded proteins in the endoplasmic reticulum (ER) triggers transcriptional and translational reprogramming. This unfolded protein response (UPR) protects cells during transient stress and can lead to apoptosis during prolonged stress. Two key mediators of the UPR are PKR-like ER kinase (PERK), which phosphorylates the α subunit of eukaryotic translation initiation factor 2 (eIF2α), resulting in decreased protein synthesis, and the α subunit of inositol-requiring enzyme 1 (IRE1α), which initiates cytoplasmic splicing of the mRNA encoding the transcription factor X-box binding protein 1 (XBP1). XBP1 induces transcription of genes involved in protein quality control. This report describes cross talk between these two pathways: phosphorylation of eIF2α was required for maximal induction of spliced XBP1 (XBP1s) protein levels via a mechanism that involved stabilization of XBP1s mRNA. By using mouse embryo fibroblasts deficient in UPR signaling pathways, we demonstrate that stress-induced stabilization of XBP1s mRNA requires cytoplasmic splicing of the mRNA and inhibition of its translation. Because the XBP1s protein promotes transcription of its own gene, the UPR-induced mRNA stabilization is part of a positive feedback loop that induces XBP1s protein accumulation and transcription of target genes during stress. We propose a model in which eIF2α phosphorylation-mediated control of mRNA turnover is a molecular switch that regulates the stress response transcription program and the ER's capacity for protein folding during stress.


Subject(s)
DNA-Binding Proteins/genetics , Endoplasmic Reticulum Stress/genetics , Protein Biosynthesis , RNA Splicing , Transcription Factors/genetics , Unfolded Protein Response , Animals , Cell Line , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Mice , Models, Molecular , Phosphorylation , Protein Folding , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regulatory Factor X Transcription Factors , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic , X-Box Binding Protein 1
12.
Mol Cell Biol ; 29(10): 2899-912, 2009 May.
Article in English | MEDLINE | ID: mdl-19273590

ABSTRACT

The response to amino acid starvation involves the global decrease of protein synthesis and an increase in the translation of some mRNAs that contain an internal ribosome entry site (IRES). It was previously shown that translation of the mRNA for the arginine/lysine amino acid transporter Cat-1 increases during amino acid starvation via a mechanism that utilizes an IRES in the 5' untranslated region of the Cat-1 mRNA. It is shown here that polypyrimidine tract binding protein (PTB) and an hnRNA binding protein, heterogeneous nuclear ribonucleoprotein L (hnRNP L), promote the efficient translation of Cat-1 mRNA during amino acid starvation. Association of both proteins with Cat-1 mRNA increased during starvation with kinetics that paralleled that of IRES activation, although the levels and subcellular distribution of the proteins were unchanged. The sequence CUUUCU within the Cat-1 IRES was important for PTB binding and for the induction of translation during amino acid starvation. Binding of hnRNP L to the IRES or the Cat-1 mRNA in vivo was independent of PTB binding but was not sufficient to increase IRES activity or Cat-1 mRNA translation during amino acid starvation. In contrast, binding of PTB to the Cat-1 mRNA in vivo required hnRNP L. A wider role of hnRNP L in mRNA translation was suggested by the decrease of global protein synthesis in cells with reduced hnRNP L levels. It is proposed that PTB and hnRNP L are positive regulators of Cat-1 mRNA translation via the IRES under stress conditions that cause a global decrease of protein synthesis.


Subject(s)
Amino Acids/metabolism , Cationic Amino Acid Transporter 1/metabolism , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , 5' Untranslated Regions , Animals , Cationic Amino Acid Transporter 1/genetics , Cell Line , Gene Expression Regulation , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Mice , Nucleic Acid Conformation , Polypyrimidine Tract-Binding Protein/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ribosomes/metabolism
13.
J Biol Chem ; 283(33): 22443-56, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18550528

ABSTRACT

The accumulation of unfolded proteins in the endoplasmic reticulum (ER) triggers a stress response program that protects cells early in the response and can lead to apoptosis during prolonged stress. The basic leucine zipper transcription factor, CCAAT/enhancer-binding protein beta (C/EBPbeta), is one of the genes with increased expression during ER stress. Translation of the C/EBPbeta mRNA from different initiation codons leads to the synthesis of two transcriptional activators (LAP-1 and -2) and a transcriptional repressor (LIP). The LIP/LAP ratio is a critical factor in C/EBPbeta-mediated gene transcription. It is shown here that the LIP/LAP ratio decreased by 5-fold during the early phase of ER stress and increased by 20-fold during the late phase, mostly because of changes in LIP levels. The early decrease in LIP required degradation via the proteasome pathway and phosphorylation of the translation initiation factor, eIF2alpha. The increased LIP levels during the late phase were due to increased synthesis and increased stability of the protein. It is proposed that regulation of synthesis and degradation rates during ER stress controls the LIP/LAP ratio. The importance of C/EBPbeta in the ER-stress response program was demonstrated using C/EBPbeta-deficient mouse embryonic fibroblasts. It is shown that C/EBPbeta attenuates expression of pro-survival ATF4 target genes in late ER stress and enhances expression of cell death-associated genes downstream of CHOP. The inhibitory effect of LIP on ATF4-induced transcription was demonstrated for the cat-1 amino acid transporter gene. We conclude that regulation of LIP/LAP ratios during ER stress is a novel mechanism for modulating the cellular stress response.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/physiology , Endoplasmic Reticulum/physiology , Liver/physiology , Transcription, Genetic , Animals , CCAAT-Enhancer-Binding Protein-beta/deficiency , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Line, Tumor , Genes, Reporter , Glioma/genetics , Luciferases/genetics , Plasmids , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Rats
14.
J Biol Chem ; 281(26): 17929-40, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16621798

ABSTRACT

Nutritional stress caused by amino acid starvation involves a coordinated cellular response that includes the global decrease of protein synthesis and the increased production of cell defense proteins. Part of this response is the induction of transport system A for neutral amino acids that leads to the recovery of cell volume and amino acid levels once extracellular amino acid availability is restored. Hypertonic stress also increases system A activity as a mechanism to promote a rapid recovery of cell volume. Both a starvation-dependent and a hypertonic increase of system A transport activity are due to the induction of SNAT2, the ubiquitous member of SLC38 family. The molecular mechanisms underlying SNAT2 induction were investigated in tissue culture cells. We show that the increase in system A transport activity and SNAT2 mRNA levels upon amino acid starvation were blunted in cells with a mutant eIF2alpha that cannot be phosphorylated. In contrast, the induction of system A activity and SNAT2 mRNA levels by hypertonic stress were independent of eIF2alpha phosphorylation. The translational control of the SNAT2 mRNA during amino acid starvation was also investigated. It is shown that the 5'-untranslated region contains an internal ribosome entry site that is constitutively active in amino acid-fed and -deficient cells and in a cell-free system. We also show that amino acid starvation caused a 2.5-fold increase in mRNA and protein expression from a reporter construct containing both the SNAT2 intronic amino acid response element and the SNAT2-untranslated region. We conclude that the adaptive response of system A activity to amino acid starvation requires eukaryotic initiation factor 2alpha phosphorylation, increased gene transcription, and internal ribosome entry site-mediated translation. In contrast, the response to hypertonic stress does not involve eukaryotic initiation factor 2alpha phosphorylation, suggesting that SNAT2 expression can be modulated by specific signaling pathways in response to different stresses.


Subject(s)
Amino Acid Transport System A/genetics , Amino Acid Transport System A/metabolism , Amino Acids/metabolism , Eukaryotic Initiation Factor-2/metabolism , Protein Biosynthesis/physiology , 5' Untranslated Regions , Animals , Cell-Free System , Gene Expression Regulation/physiology , Genes, Reporter , Glioma , HeLa Cells , Humans , Hypertonic Solutions , Osmotic Pressure , Phosphorylation , RNA, Messenger/metabolism , Ribosomes/physiology , Signal Transduction/physiology , Transcriptional Activation/physiology
15.
Mol Cell ; 17(3): 405-16, 2005 Feb 04.
Article in English | MEDLINE | ID: mdl-15694341

ABSTRACT

It was previously shown that the mRNA for the cat-1 Arg/Lys transporter is translated from an internal ribosome entry site (IRES) that is regulated by cellular stress. Amino acid starvation stimulated cat-1 translation via a mechanism that requires translation of an ORF in the mRNA leader and remodeling of the leader to form an active IRES (the "zipper model" of translational control). It is shown here that slowing of the leader peptide elongation rate, either by cycloheximide or the introduction of rare codons, stimulated translation of the downstream ORF. These results suggest that ribosome stalling in the upstream ORF causes mRNA remodeling and formation of an active IRES. This control is reminiscent of translation attenuation in prokaryotic operons, where inhibition of translation elongation can regulate both mRNA translation and gene transcription by altering mRNA structure.


Subject(s)
Protein Biosynthesis , Ribosomes/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cationic Amino Acid Transporter 1/genetics , Cell Line , Codon, Initiator/genetics , Codon, Terminator/genetics , Cycloheximide/pharmacology , Eukaryotic Cells/metabolism , Eukaryotic Initiation Factor-2/metabolism , In Vitro Techniques , Mice , Models, Biological , Molecular Sequence Data , Nucleic Acid Conformation , Open Reading Frames , Phosphorylation , Prokaryotic Cells/metabolism , Protein Biosynthesis/drug effects , RNA, Messenger/chemistry , RNA, Messenger/genetics , Rabbits , Rats , Transfection
16.
Curr Protoc Cell Biol ; Chapter 7: Unit 7.8, 2002 Feb.
Article in English | MEDLINE | ID: mdl-18228410

ABSTRACT

This unit describes methods for preparation of glycoproteins metabolically labeled with radioactive sugars, sulfate, and phosphate. Methods for liberation of both N- and O-linked glycans are also described. These protocols can be used to generate materials for characterization of glycoprotein glycans from cultured cells.


Subject(s)
Clinical Laboratory Techniques , Glycoproteins/chemistry , Isotope Labeling/methods , Carbohydrate Metabolism , Carbohydrates/chemistry , Glycoproteins/metabolism , Metabolism , Radioisotopes , Research Design
17.
Mol Cell ; 12(4): 796-7, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14580329

ABSTRACT

When growth factors are removed from many mammalian cells, growth ceases and apoptosis is induced. The small GTPase rab7, which regulates endocytic membrane traffic, participates in this process by mediating the regulated internalization and degradation of nutrient transporters. This process triggers nutrient starvation that helps to induce cell death.


Subject(s)
Apoptosis/physiology , Cell Division/physiology , Growth Substances/physiology , rab GTP-Binding Proteins/physiology , Animals , Energy Metabolism/physiology , Humans , Protein Transport/physiology , rab7 GTP-Binding Proteins
18.
J Biol Chem ; 277(21): 19198-205, 2002 May 24.
Article in English | MEDLINE | ID: mdl-11877448

ABSTRACT

Initiation of translation from most cellular mRNAs occurs via scanning; the 40 S ribosomal subunit binds to the m(7)G-cap and then moves along the mRNA until an initiation codon is encountered. Some cellular mRNAs contain internal ribosome entry sequences (IRESs) within their 5'-untranslated regions, which allow initiation independently of the 5'-cap. This study investigated the ability of cellular stress to regulate the activity of IRESs in cellular mRNAs. Three stresses were studied that cause the phosphorylation of the translation initiation factor, eIF2alpha, by activating specific kinases: (i) amino acid starvation, which activates GCN2; (ii) endoplasmic reticulum (ER) stress, which activates PKR-like ER kinase, PERK kinase; and (iii) double-stranded RNA, which activates double-stranded RNA-dependent protein kinase (PKR) by mimicking viral infection. Amino acid starvation and ER stress caused transient phosphorylation of eIF2alpha during the first hour of treatment, whereas double-stranded RNA caused a sustained phosphorylation of eIF2alpha after 2 h. The effects of these treatments on IRES-mediated initiation were investigated using bicistronic mRNA expression vectors. No effect was seen for the IRESs from the mRNAs for the chaperone BiP and the protein kinase Pim-1. In contrast, translation mediated by the IRESs from the cationic amino acid transporter, cat-1, and of the cricket paralysis virus intergenic region, were stimulated 3- to 10-fold by all three treatments. eIF2alpha phosphorylation was required for the response because inactivation of phosphorylation prevented the stimulation. It is concluded that cellular stress can stimulate translation from some cellular IRESs via a mechanism that requires the phosphorylation of eIF2alpha. Moreover, there are distinct regulatory patterns for different cellular mRNAs that contain IRESs within their 5'-untranslated regions.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Protein Biosynthesis , Ribosomes/metabolism , Animals , Cells, Cultured , Oxidative Stress , Phosphorylation , Rats , Tumor Cells, Cultured
19.
J Biol Chem ; 277(14): 11780-7, 2002 Apr 05.
Article in English | MEDLINE | ID: mdl-11781318

ABSTRACT

The cationic amino acid transporter, Cat-1, is a high affinity transporter of the essential amino acids, arginine and lysine. Expression of the cat-1 gene is known to be regulated by amino acid availability. It is shown here that cat-1 gene expression is also induced by Glc limitation, which causes a 7-fold increase in cat-1 mRNA, a 30-fold induction of Cat-1 protein levels, and a 4-fold stimulation of arginine uptake. Glc limitation is known to induce the unfolded protein response (UPR) by altering protein glycosylation in the endoplasmic reticulum (ER). The studies here demonstrate that synthesis of Cat-1 occurs during the UPR when global protein synthesis is inhibited. The 5'-UTR of the cat-1 mRNA contains an internal ribosomal entry site (IRES) that is activated by amino acid starvation by a mechanism that involves phosphorylation of the translation initiation factor, eukaryotic initiation factor 2alpha, by the GCN2 kinase. It is shown here that translation from the cat-1/IRES is also induced by Glc deprivation in a manner dependent upon phosphorylation of eukaryotic initiation factor 2alpha by the transmembrane ER kinase, PERK. Because PERK is a key constituent of the UPR, it is concluded that induction of cat-1 gene expression is part of the adaptive response of cells to ER stress. These results also demonstrate that regulation of IRES activity in cellular mRNAs is part of the mechanism by which the UPR protects cells from unfolded proteins in the ER.


Subject(s)
Cell Cycle Proteins/metabolism , Glucose/metabolism , Phosphoproteins/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Ribosomes/metabolism , eIF-2 Kinase/metabolism , 3T3 Cells , Animals , Binding Sites , Blotting, Northern , Blotting, Western , Dose-Response Relationship, Drug , Endoplasmic Reticulum/metabolism , GTPase-Activating Proteins , Genes, Dominant , Genetic Vectors , Glycosylation , Ions , Luciferases/metabolism , Mice , Models, Genetic , Phosphorylation , Protein Binding , Rats , Time Factors , Transfection , Tumor Cells, Cultured
20.
J Biol Chem ; 277(44): 41539-46, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12196519

ABSTRACT

The cationic amino acid transporter, Cat-1, is a high affinity transporter of the essential amino acids, arginine and lysine. Expression of the cat-1 gene increases during nutritional stress as part of the adaptive response to starvation. Amino acid limitation induces coordinate increases in stability and translation of the cat-1 mRNA, at a time when global protein synthesis decreases. It is shown here that increased cat-1 mRNA stability requires an 11 nucleotide AU-rich element within the distal 217 bases of the 3'-untranslated region. When this 217-nucleotide nutrient sensor AU-rich element (NS-ARE) is present in a chimeric mRNA it confers mRNA stabilization during amino acid starvation. HuR is a member of the ELAV family of RNA-binding proteins that has been implicated in regulating the stability of ARE-containing mRNAs. We show here that the cytoplasmic concentration of HuR increases during amino acid starvation, at a time when total cellular HuR levels decrease. In addition, RNA gel shift experiments in vitro demonstrated that HuR binds to the NS-ARE and binding was dependent on the 11 residue AU-rich element. Moreover, HuR binding to the NS-ARE in extracts from amino acid-starved cells increased in parallel with the accumulation of cytoplasmic HuR. It is proposed that an adaptive response of cells to nutritional stress results in increased mRNA stability mediated by HuR binding to the NS-ARE.


Subject(s)
3' Untranslated Regions/metabolism , Amino Acids/metabolism , Antigens, Surface , Cationic Amino Acid Transporter 1/genetics , Cytoplasm/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Animals , Base Sequence , ELAV Proteins , ELAV-Like Protein 1 , Electrophoretic Mobility Shift Assay , Molecular Sequence Data , RNA Stability , RNA, Messenger/chemistry , Rats , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL