Skip to main content
Physiological Genomics logoLink to Physiological Genomics
. 2009 Aug;38(3):328–341. doi: 10.1152/physiolgenomics.90396.2008

eIF2α kinases GCN2 and PERK modulate transcription and translation of distinct sets of mRNAs in mouse liver

An N Dang Do 1, Scot R Kimball 1, Douglas R Cavener 2, Leonard S Jefferson 1
PMCID: PMC3171828  PMID: 19509078

Abstract

In eukaryotes, selective derepression of mRNA translation through altered utilization of upstream open reading frames (uORF) or internal ribosomal entry sites (IRES) regulatory motifs following exposure to stress is regulated at the initiation stage through the increased phosphorylation of eukaryotic initiation factor 2 on its α-subunit (eIF2α). While there is only one known eIF2α kinase in yeast, general control nonderepressible 2 (GCN2), mammals have evolved to express at least four: GCN2, heme-regulated inhibitor kinase (HRI), double-stranded RNA-activated protein kinase (PKR), and PKR-like endoplasmic reticulum-resident kinase (PERK). So far, the main known distinction among these four kinases is their activation in response to different acute stressors. In the present study, we used the in situ perfused mouse liver model and hybridization array analyses to assess the general translational response to stress regulated by two of these kinases, GCN2 and PERK, and to differentiate between the downstream effects of activating GCN2 versus PERK. The resulting data showed that at least 2.5% of mouse liver mRNAs are subject to derepressed translation following stress. In addition, the data demonstrated that eIF2α kinases GCN2 and PERK differentially regulate mRNA transcription and translation, which in the latter case suggests that increased eIF2α phosphorylation is not sufficient for derepression of translation. These findings open an avenue for more focused future research toward groups of mRNAs that code for the early cellular stress response proteins.

Keywords: mRNA translation, endoplasmic reticulum stress, amino acid deprivation, eukaryotic initiation factor 2α phosphorylation


global rates of protein synthesis are repressed in response to a variety of cell stresses, including UV irradiation (14), infection by certain viruses (17, 21, 33, 56, 59, 64, 71, 77), and deprivation of essential amino acids (18, 35, 36). In part, the repression of global protein synthesis associated with such stresses is a consequence of phosphorylation of Ser51 on the α-subunit of the translation initiation factor eukaryotic initiation factor (eIF)2 (33, 56, 59, 64, 81). During one of the first steps in the initiation of mRNA translation, eIF2 binds to GTP and the initiator methionyl-tRNAi to form the ternary complex that subsequently binds to the 40S ribosomal subunit (29). In a later step in initiation, the GTP bound to eIF2 is hydrolyzed to GDP, and the resulting eIF2·GDP complex is released from the 40S ribosomal subunit. For initiation to recur, the GDP bound to eIF2 must be exchanged for GTP, a reaction catalyzed by another initiation factor, eIF2B. Phosphorylation of eIF2α on Ser51 converts eIF2 from a substrate into a competitive inhibitor of eIF2B, leading to repression of global rates of protein synthesis (70). Paradoxically, inhibition of eIF2B by phosphorylated eIF2 leads to upregulated translation of mRNAs with particular motifs in their 5′-untranslated region (UTR), such as upstream open reading frames (uORF) and internal ribosomal entry sites (IRES) (41, 68, 73, 74). Although information concerning the number and identity of mRNAs whose translation is upregulated in response to eIF2α phosphorylation is limited, those that have been identified typically encode proteins involved in cellular adaptation to the stress (41).

The genome of the yeast Saccharomyces cerevisiae encodes a single eIF2α kinase, general control nonderepressible 2 (GCN2) (15). GCN2 is activated in response to nutrient deprivation and, in particular, to deprivation of amino acids (39). In contrast, mammals have four distinct eIF2α kinases: a mammalian ortholog of GCN2, the double-stranded RNA-activated protein kinase (PKR), the heme-controlled inhibitor (HRI), and the PKR-like endoplasmic reticulum (ER)-resident kinase (PERK) (reviewed in Ref. 76). Each of these kinases is activated in response to distinct stresses: GCN2 by deprivation of essential amino acids, PKR by infection with certain viruses, HRI during heme limitation, and PERK by ER stress [e.g., during the unfolded protein response (UPR)]. Phosphorylation of eIF2α by any of the four kinases results in inhibition of eIF2B and repressed global rates of protein synthesis. In addition, phosphorylation of eIF2α is thought to be important in effecting a recovery from the stress. For example, phosphorylation of eIF2α by PERK in response to accumulation of unfolded proteins in the lumen of the ER represses the synthesis of proteins destined for that subcellular location, thereby minimizing further accumulation of unfolded proteins (28). However, PERK-mediated phosphorylation of eIF2α also leads to increased translation of mRNAs encoding certain transcription factors such as activating transcription factor (ATF)4 (aka C/ATF/CREB2/TAXREB67) and ATF5 (aka AFTA/Atf7/Arfx/ODA-10) that promote the transcription of genes encoding proteins such as growth arrest and DNA-damage-inducible protein (GADD)34 (aka PPR1R15A) (55). GADD34 targets protein phosphatase 1 to eIF2, resulting in dephosphorylation of the protein and restoration of global protein synthesis. If this second function, i.e., increased translation of mRNAs encoding specific proteins, is as crucial as that of repressing global protein synthesis, it is logical to surmise that each of the eIF2α kinases would effect a recovery program specific to its activating stress, hence influencing the translation efficiency of different subsets of mRNAs.

In the present study we hypothesized that activation of different eIF2α kinases affects the translational regulation of both a general subset of mRNAs involved in determining cell fate as well as a specific subset of mRNAs involved in responding to the activating stress. To test the hypothesis, we performed in situ liver perfusion studies using wild-type mice and mice lacking either GCN2 (Gcn2−/−) or PERK (Perk−/−). GCN2 was activated by deprivation of the essential amino acid Met, and PERK was activated with 2,5-di-tert-butylhydroquinone (tBuHQ), a selective inhibitor of ATP-dependent microsomal Ca2+ sequestration and activator of ER stress (32, 51). Overall, the results support the hypothesis, i.e., activation of GCN2 resulted in altered translation of both overlapping and distinct mRNAs compared with activation of PERK.

MATERIALS AND METHODS

Animals.

All animals received water and food (Harland) ad libitum and were maintained in accordance with the protocol approved by the Pennsylvania State College of Medicine's Institutional Animal Care and Use Committee. Global Gcn2−/− and wild-type littermate mice, 3–6 mo old, were generated according to a previously published protocol (81). The liver-specific knockout of Perk was generated by crossing the AlbCre deletor strain by the floxed Perk allele (Perk f), yielding AlbCre Perkf/f and wild-type littermate mice, also 3–6 mo old, as previously described (80). The AlbCre Perkf/f mice are referred to as Perk−/− here. Both Gcn2 and Perk mice are on a mixed C57BL/6J-129SvEvTac genetic background that has been intercrossed for several generations, and thus have a similar genetic background.

Liver perfusion.

Livers were perfused with a single-pass protocol as described previously (20, 72) with slight modifications. The perfusion medium contained a 2-to-1 ratio of buffer A [modified Ringer II buffer (mM): 110 NaCl, 4.4 KCl, 2.36 CaCl2, 1.1 KH2PO4, 1.1 MgSO4, 25 NaHCO3, and 11 glucose, with 3% albumin] and washed bovine red blood cells. For perfusions of Gcn2+/+ and −/− mice, Met was omitted from the perfusate for the treatment condition. For perfusions of Perk+/+ and −/− mice, the final CaCl2 concentration was lowered to 0.1 mM in buffer A. To induce ER stress, tBuHQ dissolved in DMSO was added to the perfusate immediately before perfusion to yield a final concentration of 40 μM. An equal volume of DMSO was added to the control perfusate. All perfusions were performed at 37°C for 35 min.

eIF2B guanine nucleotide exchange assay.

The guanine nucleotide exchange activity of eIF2B was measured with published protocols (13, 37). Perfused liver samples were homogenized with a Dounce homogenizer in 4 volumes of buffer B (mM: 45 HEPES, pH 7.4, 0.375 MgOAc, 0.075 EDTA, 95 KOAc, and 2 digitonin, with 10% glycerol and 3 μM microcystin), and the homogenate was centrifuged at 10,000 g for 10 min at 4°C. Supernatant (25 μl) was added to a tube containing 97 μl of H2O and 140 μl of eIF2B assay buffer (mM: 52.1 MOPS, pH 7.4, 0.22 GDP, 104 KCl, 1.04 DTT, and 2.08 MgOAc, with 0.21 mg/ml bovine serum albumin), and the mixture was preincubated at 30°C for 1 min. The reaction was started by the addition of 35 μl of [3H]GDP·eIF2 binary complex followed by incubation at 30°C. Aliquots were removed from the assay reaction mixture into 2.5 ml of wash buffer (mM: 50 MOPS, pH 7.4, 2 MgOAc, 100 KCl, 1 DTT) at 0, 30, 60, and 90 s and vacuum filtered through 0.45-μm cellulose nitrate membrane filters (Whatman International, Maidstone, UK). The membranes were washed twice with 2.5 ml of wash buffer, dissolved in Filtron-X (National Diagnostics, Atlanta, GA) and the radioactivity contained thereon was measured in a scintillation counter (Beckman LS 6500 multipurpose scintillation counter).

SDS-polyacrylamide gel electrophoresis and Western blot analysis.

Perfused liver samples were homogenized in 7 volumes of buffer C (mM: 20 HEPES, pH 7.4, 2 EGTA, 50 NaF, 100 KCl, 0.2 EDTA, 50 β-glycerophosphate, 1 DTT, 0.1 PMSF, 1 benzamidine, and 0.5 NaVO4, with 10 μl/ml Sigma protease inhibitor cocktail). The homogenate was centrifuged at 1,000 g for 3 min at 4°C, and an aliquot of the supernatant was added to an equal volume of SDS sample buffer [0.125 M Tris·HCl, pH 6.8, 25% (vol/vol) glycerol, 2.5% SDS, 2.5% (vol/vol) β-mercaptoethanol, 0.2% bromophenol blue] and boiled at >95°C for 4 min. Protein samples were stored at −70°C until resolved by SDS-PAGE. After electrophoresis, proteins in the gel were transferred onto 0.45-μm polyvinylidene difluoride (PVDF) membranes (Pall Life Sciences, Exton, PA) and the membranes were blocked with 5% nonfat dry milk. Membranes were then incubated with primary antibody overnight at 4°C using the following antibodies: anti-phospho-Ser51-eIF2α (Biosource International, Invitrogen, Carlsbad, CA) and monoclonal anti-eIF2α (63). The next day membranes were incubated with secondary antibody at room temperature for 1 h with horseradish peroxidase-conjugated anti-rabbit or -mouse antibodies (Bethyl, Montgomery, TX), respectively. Membranes were developed with enhanced chemiluminescence reagents (Amersham Biosciences, Piscataway, NJ), and images were captured with GeneGnome software (SynGene, Frederick, MD) and quantitated with GeneTools software (SynGene).

Sucrose density gradient centrifugation.

Discontinuous nine-step 20% [mM: 10 HEPES, pH 7.4, 250 KCl, 5 MgCl2, 0.5 EDTA, with 20% (wt/wt) sucrose] to 47% [mM: 10 HEPES, pH 7.4, 250 KCl, 5 MgCl2, 0.5 EDTA, with 47% (wt/wt) sucrose] sucrose density gradients were used to separate mRNAs based on the number of bound ribosomes, an indicator of translational state. Perfused liver samples (1 g) were homogenized with a Dounce homogenizer in 3 volumes of buffer D (mM: 50 HEPES, pH 7.4, 250 KCl, 5 MgCl2, 250 sucrose, 100 μg/ml cycloheximide), and the homogenate was centrifuged at 3,000 g for 15 min at 4°C. The supernatant (1 ml) was removed to a separate tube, and 10% (wt/vol) Triton X-100 and 13% (wt/wt) sodium deoxycholate (100 μl each) were added and gently mixed. Detergent-containing supernatant (1 ml) was loaded onto the 20–47% sucrose density gradients and centrifuged at 141,371 g for 3 h 38 min at 4°C for resolution of polysomes or 91,287 g for 19 h 9 min at 4°C for resolution of ribosomal subunits and monomers. After centrifugation, gradients were fractionated with an ISCO UV gradient fractionator while the absorbance at 254 nm was continuously monitored (Teledyne Isco, Lincoln, NE). Quantitation of the area under the absorbance curves was performed with the Un-Scan-It software program (version 4.3, Silk Scientific, Orem, UT).

RNA extraction.

Total RNA was extracted from 1 ml of detergent-containing supernatant or from the sucrose density gradient fractions with acid phenol:chloroform (Ambion, Austin, TX). Per milliliter of sample, 40 μl of 0.5 M EDTA, 25 μl of 20% SDS, and 1 volume of acid phenol:chloroform were added. The mixture was centrifuged at 3,100 g for 30 min at 4°C, the top clear layer was removed, and RNA was extracted a second time by the addition of 1 volume of acid phenol:chloroform. RNA was precipitated overnight in 2.5 volumes of 100% ethanol and 0.1 volume of 5 M NH4OAc (Ambion). RNA was pelleted, washed twice with 75% ethanol, and resuspended in RNA storage solution plus anti-RNase (Ambion). The concentration and purity of RNA samples were measured with an Agilent 2100 Bioanalyzer in the Pennsylvania State University College of Medicine Functional Genomics Core Facility before hybridization array and quantitative (q)RT-PCR analysis.

Hybridization array analysis.

Total RNA from two perfused liver samples per condition (unfractionated, nonpolysomal, subpolysomal, or polysomal) was pooled to minimize biological variation, and the experiment was performed in duplicate. Twelve micrograms of total RNA was used in the initial cDNA conversion step. All steps including double-strand cDNA synthesis, in vitro labeling, and cRNA fragmentation were done with the One-Cycle Target Labeling and Control Reagents kit and protocol (Affymetrix, Santa Clara, CA). Samples were hybridized to GeneChip Mouse Genome 430 2.0 arrays (Affymetrix) in a GeneChip Hybridization Oven 640 and washed and stained in a GeneChip Fluidics Station 400, with reagents from the above-mentioned kit. Arrays were scanned with a GCS 3000 scanner, using GCOS 1.2 software, in the Pennsylvania State University DNA Microarray Facility. Hybridization array data have been submitted to the National Center for Biotechnology Information's Gene Expression Omnibus according to the MIAME (Minimum information about a microarray experiment) protocol [Ref. 7; GSE11685, http://www.ncbi.nlm.nih.gov/projects/geo/query/acc.cgi?acc=GSE11685].

Affymetrix Chip files containing qualitative and quantitative values of each probe set on the arrays were imported into GeneSpring (Agilent Technologies, Palo Alto, CA) and sorted for raw expression values >80 in at least 1 of 16 conditions, as an initial filter to eliminate background noise. The resulting lists were imported into Microsoft Excel and analyzed with the Z-score method (10, 48). First, the data were log10 transformed. The transformed values were used in the following calculations: Zscore = (intensityG − mean intensityG1…Gn)/SDG1…Gn, where G is any gene on the hybridization array and G1…Gn is the aggregate measure of all genes, and Zratios = [(ZscoreG1ave)Exp − (ZscoreG1ave)Con]/SD of Z score differencesG1…Gn, where G1ave = average Z score for any particular gene being tested under multiple experimental conditions [experimental (Exp) vs. control (Con)].

Lists of different criteria were generated by sorting the appropriate Z scores in Microsoft Excel. The resulting probe lists were categorized based on their biological process with the GeneSpring Gene Ontology.

Quantitative RT-PCR.

Total RNA (1 μg) was converted into cDNA with the SuperScript First Strand Synthesis Kit (Invitrogen). A wild-type, untreated, unfractionated liver cDNA sample was used to create a standard curve. The remaining cDNA samples were diluted 1:16 and an amount equal to 1/10th of the final reaction volume was used per PCR reaction. For validation of changes in Stch (aka HSPA13) expression observed with the ABI gene expression assay, the plates were run with the TaqMan reporter on an ABI 7000 sequence detection system following the manufacturer-recommended settings (Applied Biosystems, Foster City, CA). For all other qRT-PCR validation reactions using SYBR Green, the final primer concentration was 100 μM. The plates were run on either an MJ Research Opticon 2 or an ABI 7000 thermal cycler with the following settings: 95°C for 15 min, 50 cycles of 94°C for 15 s, 55°C for 20 s, 72°C for 15 s. Primer sequences are listed in Supplemental Table S1.1 Expression level was calculated with the absolute quantitation method (8). The amount of the cDNA of interest was normalized to the average expression amount of β-actin, GAPDH, and Tbp (aka GTF2D1/Gtf2d/SCA17/TFIID) cDNA in the same fraction. The amount of mRNA encoding individual proteins present in fractions from sucrose density gradients was expressed relative to the total amount of that mRNA present in the combined non-, sub-, and polysome fractions.

Statistical analysis.

Statistical analyses were done with the InStat software program (GraphPad Software, La Jolla, CA) as detailed in Figs. 1–9.

RESULTS

Validation of experimental model systems.

GCN2 is activated in response to individual deprivation of any of the essential amino acids (39). However, in liver, certain amino acids, e.g., the branched-chain amino acids and tryptophan (2), also modulate signaling through the mammalian target of rapamycin (mTOR) complex 1 (mTORC1) pathway. Therefore, in pilot studies, mouse livers were perfused with medium lacking individual amino acids, and the effect on phosphorylation of eIF2α, an index of GCN2 activation, and 4E-BP1 and S6K1, indexes of mTORC1 activation, was assessed. In contrast to Leu or Trp, deprivation of Met resulted in a significant increase in eIF2α phosphorylation, with no effect on mTORC1 signaling (data not shown). Consequently, Met deprivation was used as a means of specifically activating GCN2 in subsequent experiments.

To functionally confirm the absence of amino acid-regulated eIF2α kinase activity in livers of Gcn2−/− mice, livers from Gcn2+/+ or Gcn2−/− mice were perfused with medium containing or lacking Met and changes in eIF2α phosphorylation were assessed by Western blot analysis. As shown in Fig. 1A, Met deficiency caused eIF2α phosphorylation to increase ∼2.2-fold in livers from Gcn2+/+ mice. In contrast, Met deficiency had no effect on eIF2α phosphorylation in livers from Gcn2−/− mice. Moreover, eIF2B activity was reduced in livers from Gcn2+/+, but not Gcn2−/−, mice in response to Met deprivation (Fig. 1C). Inhibition of translation initiation relative to elongation/termination leads to disaggregation of polysomes, with a resulting accumulation of 40S and 60S ribosomal subunits as individual subunits or as 80S monosomes (referred to here as the subpolysomal fraction). The inhibition of eIF2B activity observed in the present study would therefore be expected to result in polysome disaggregation. Indeed, as shown in Fig. 1, E and G, in livers from Gcn2+/+ mice, Met deficiency caused a decrease in the number of ribosomes present in the polysome fraction and a corresponding increase in the number of 80S monomers present in the subpolysomal fraction.

Fig. 1.

Fig. 1.

Regulation of eukaryotic initiation factor (eIF)2α, eIF2B, and polysome aggregation in perfused general control nonderepressible 2 (GCN2)- and double-stranded RNA-activated protein kinase (PKR)-like endoplasmic reticulum-resident kinase (PERK)-perfused mouse livers after methionine (Met) deprivation or 2,5-di-tert-butylhydroquinone (tBuHQ) treatment, respectively. A, C, E, G: Gcn2+/+ or −/− mouse livers were perfused for 35 min with perfusate containing complete amino acids (open bars, A and C) or deficient in Met (gray bars, A and C). B, D, F, H: Perk+/+ or Alb/Cre Perk−/− mouse livers perfused for 35 min with perfusate containing DMSO (open bars, B and D) or 40 μM tBuHQ (gray bars, B and D). A and B: insets show representative Western blots for Ser51 phosphorylated (2αP, top) and total (2αT, bottom) eIF2α. Bars represent quantitation of Western blot band density, calculated as ratio of phosphorylated over total eIF2α. Values are means ± SE. C and D: guanine nucleotide exchange activity of eIF2B. n = 9 or 10 livers/group. Statistical significance was determined by unpaired t-test: *P < 0.001, **P < 0.025. E–H: polysome profiles of GCN2 (E, G)- and PERK (F, H)-perfused mouse livers. Supernatant from liver homogenate was resolved on a 20–47% sucrose density gradient. Absorbance at 254 nm was continuously measured and recorded to generate profiles. Assignment of the 40S, 60/80S, subpolysome, and polysome peaks was as demonstrated in E. E and F: representative gradient profile for conditions of wild-type control condition. G and H: representative gradient profile for wild-type treated condition. Profiles are representative of n = 9 or 10 livers/group.

Mobilization of calcium from the ER promotes misfolding of proteins within the ER lumen, leading to activation of PERK and subsequently to phosphorylation of eIF2α. The absence of ER stress-induced eIF2α kinase activity in livers from Perk−/− mice was confirmed by perfusing livers from Perk+/+ and Perk−/− mice with tBuHQ to promote ER calcium release (51) and then measuring eIF2α phosphorylation and eIF2B activity. As shown in Fig. 1, B and D, perfusion of livers from Perk+/+, but not Perk−/−, mice with tBuHQ resulted in both increased eIF2α phosphorylation and decreased eIF2B activity, respectively. Moreover, polysome disaggregation was induced by perfusion of livers from Perk+/+ mice with tBuHQ (Fig. 1, F and H). Interestingly, both eIF2B activity and polysome aggregation were affected to a larger extent in response to perfusion with tBuHQ compared with Met deprivation.

Global alterations in mRNA expression in livers lacking GCN2 or PERK.

Initially, the expression of individual mRNAs in livers of Gcn2+/+ mice perfused with all amino acids, or without Met, was compared with those expressed in livers of Gcn2−/− mice perfused under the same condition. The time point chosen for these studies, i.e., 35 min, was selected based on previous studies (45a) showing that in cells in culture mRNAs shift from the RNP fraction to polysomes within 30 min. This duration was likely too short to allow for detection of some transcriptional changes. However, even at this relatively early time point, the expression of 574 (all amino acids) and 628 (without Met) mRNAs was greater in Gcn2−/− compared with Gcn2+/+ mice (Supplemental Table S2, tabs 1 and 2, respectively), and 141 mRNAs were common to both lists (Fig. 2 and Supplemental Table S2, tab 3). In parallel, the expression of 616 (all amino acids) and 675 (without Met) mRNAs was lower in livers of Gcn2−/− compared with Gcn2+/+ mice (Supplemental Table S2, tabs 4 and 5, respectively), and 163 mRNAs were common to both lists (Fig. 2 and Supplemental Table S2, tab 6). Those mRNAs exhibiting altered expression in livers lacking GCN2, and whose expression was changed in response to perfusion both with and without Met, were functionally grouped with GeneSpring. As shown in Fig. 3A, mRNAs exhibiting increased expression in the absence of GCN2 grouped into categories such as transport, with some mRNAs exhibiting Z score of 10 or greater (Supplemental Table S2, tab 3). In contrast, those mRNAs whose expression was reduced in Gcn2−/− compared with control livers include those in categories such as regulation of physiological processes, immune response, and apoptosis (Fig. 3A). Of particular interest is the finding that the expression of mRNAs encoding two proteins involved in amino acid synthesis, asparagine synthetase (Asns) and dopachrome tautomerase (Dct/TRP2/Tyrp-2), was dramatically less in Gcn2−/− compared with wild-type liver (Supplemental Table S2, tab 6). To verify the change in Asns mRNA expression identified by microarray analysis, Asns mRNA abundance was quantitated by qRT-PCR. As shown in Fig. 4, Asns mRNA expression was significantly lower in livers from Gcn2−/− compared with Gcn2+/+ mice (P < 0.001) as well as in Perk−/− compared with Perk+/+ mice (P < 0.05). However, the magnitude of the PERK-dependent change was smaller compared with the GCN2-dependent change. Similarly, the GCN2-dependent change in Cyp2c55 mRNA expression (P < 0.01) was also larger than the PERK-dependent change (not significant). Recent studies suggest that Asns mRNA expression is regulated in part by the transcription factor ATF5 (1). Therefore, the expression of ATF5 mRNA was measured by qRT-PCR. In agreement with the observed changes in Asns expression, the expression of ATF5 mRNA was significantly lower in livers from Gcn2−/− compared with control mice (Fig. 4; P < 0.05), suggesting that GCN2 may act through ATF5 to modulate Asns expression.

Fig. 2.

Fig. 2.

Number of mRNAs whose expression changed in eIF2α kinase−/− vs. +/+ perfused livers. GCN2 or PERK mouse livers were perfused for 35 min with perfusate ± Met or ± tBuHQ, respectively. Total RNA was extracted from the unfractionated supernatant and used in hybridization arrays experiments. A: analyses of the effect of eIF2α kinase on mRNA abundance were done by sorting of the Z score of genes in the arrays. mRNAs with Z score >1.5 when comparing expression in −/− vs. +/+ samples were considered to have increased expression in the absence of the eIF2α kinase. mRNAs with Z score less than −1.5 when comparing expression in −/− vs. +/+ samples were considered to have decreased expression in the absence of the eIF2α kinase. B: Venn diagram of mRNAs whose expression increased in the absence of GCN2 or PERK. C: Venn diagram of mRNAs whose expression decreased in the absence of GCN2 or PERK.

Fig. 3.

Fig. 3.

Functional categories of genes whose mRNA expression changed in the absence of GCN2 or PERK. Mouse livers were perfused for 35 min with or without treatment as described in Fig. 2. Total RNA was extracted from liver homogenates for use in hybridization array experiments. Analyses of eIF2α kinase-dependent increase or decrease in mRNA expression were done by sorting of genes with Z score greater than 1.5 or less than −1.5. Sublists of sorted genes were used for biological process classification in GeneSpring. Bars represent number of genes within the functional category whose mRNA expression increased (positive values) or decreased (negative values) in the absence of GCN2 (A) or PERK (B). A functional category is included only if it contains ≥3 genes and has a GeneSpring P value of <0.05.

Fig. 4.

Fig. 4.

Validation of hybridization array results of mRNAs whose expression decreased in the absence of GCN2 (left) or PERK (right). Experimental conditions and analyses used for the identification of genes whose mRNA expression decreased in the absence of GCN2 or PERK were as described in Figs. 2 and 3. Total RNA extracted from unfractionated liver samples was used in quantitative (q)RT-PCR to validate mRNAs with Z score less than −1.5 (Asns, Dct, Calr, Cyp2c55) or those reported in the literature to affect Asns transcription (ATF5) or to be transcriptionally regulated by PERK (Herpud1).

The studies described in the previous paragraph were repeated with liver-specific Perk-knockout mice, referred to here as Perk−/− mice, and Perk+/+ mice. Thus mRNA expression in livers of Perk−/− mice perfused in the absence or presence of tBuHQ was compared with livers of Perk+/+ mice perfused under the same conditions. In livers perfused without tBuHQ, 690 mRNAs exhibited increased abundance and 1,320 decreased abundance in livers of Perk−/− compared with Perk+/+ mice (Supplemental Table S3, tabs 1 and 4, respectively). Moreover, in livers perfused with tBuHQ, 886 mRNAs were present in greater amounts and 802 in lesser amounts, in livers of Perk−/− compared with Perk+/+ mice (Supplemental Table S3, tabs 2 and 5, respectively). In contrast to the GCN2 mice, only 59 mRNAs were commonly upregulated in control and treated Perk−/− compared with wild-type mouse liver (Fig. 2B), and the highest Z score was only 4.95 (Supplemental Table S3, tab 3). The mRNAs functionally grouped into categories including response to stimulus, positive regulation of biological process, and enzyme-linked receptor protein signaling pathway (Fig. 3B). The absence of PERK also led to decreased expression of a smaller common set of mRNAs compared with GCN2 (Fig. 2C; Supplemental Table S3, tab 6), and those that changed grouped into categories such as cell motility and cell adhesion (Fig. 3B). Only two mRNAs were upregulated in livers from both Gcn2−/− and Perk−/− mice compared with wild-type control mice (Fig. 2B), C-type lectin-related f (GenBank AK017207) and an mRNA encoding a hypothetical protein with similarity to zinc finger protein 691 (GenBank AV344364). No mRNAs were commonly downregulated in the absence of GCN2 and PERK (Fig. 2C).

GCN2 and PERK activation leads to recruitment of different functional groups of mRNAs for translation.

The effect of GCN2 or PERK activation on the distribution of individual mRNAs present in polysomes was assessed by perfusing livers in the presence or absence of Met or tBuHQ, respectively, followed by sucrose density gradient centrifugation to separate mRNAs present in polysomes from those not present (i.e., those being translated from those that are untranslated). Three criteria had to be met in order for an mRNA to be considered as translationally upregulated in a GCN2-dependent manner. First, the amount of a particular mRNA had to be the same or lower in the total fraction of wild-type mouse liver perfused in the absence compared with the presence of Met (i.e., no increase in mRNA transcription). Second, the amount of a particular mRNA had to be greater in the polysome fraction from livers of wild-type mice perfused in the absence compared with the presence of Met (i.e., increased translation by shifting into polysomes after stress). Third, the amount of that mRNA had to be the same or lower in the polysome fraction from livers of Gcn2−/− mice perfused in the absence compared with the presence of Met (i.e., increased translation following stress only in the presence of GCN2). Thus, if an mRNA exhibited increased abundance in polysomes in response to Met deprivation in livers of both Gcn2+/+ and Gcn2−/−, it was not considered to be translationally upregulated in a GCN2-dependent manner. A total of 490 mRNAs, or ∼3% of the 15,700 mRNAs identified by GeneSpring, exhibited a GCN2-dependent shift into polysomes in response to Met deprivation (Fig. 5). Grouping of the 490 mRNAs into functional classifications revealed that the proteins encoded by the mRNAs participate in a variety of cellular processes related to metabolism and energy production, with the three processes with the lowest P values being generation of precursor metabolites and energy, electron transport, and lipid metabolism (Table 1 and Supplemental Table S4, tab 1). Of mRNAs that shifted into polysomes in a GCN2-dependent manner, the greatest number encode proteins involved in generation of precursor metabolites and energy (Fig. 6).

Fig. 5.

Fig. 5.

Number of mRNAs whose abundance in the polysome fraction changed after treatment in wild-type livers. GCN2 or PERK mouse livers were perfused for 35 min with perfusate ± Met or ± tBuHQ, respectively. Total RNA was extracted from the unfractionated supernatants and polysome fractions for use in hybridization array experiments. A: analyses of the effect of treatment on mRNA abundance in the polysome fraction were done by sorting of the Z scores of genes in the arrays. mRNAs with Z score > 1.5 in polysome fraction of wild-type treated livers were considered to have shifted into polysomes after activation of the eIF2α kinase. mRNAs with Z score less than −1.5 in polysome fraction of wild-type treated livers were considered to have shifted out of polysomes after activation of the eIF2α kinase. B: Venn diagram of mRNAs that shifted into polysomes after activation of GCN2 or PERK. C: Venn diagram of mRNAs that shifted out of polysomes after activation of GCN2 or PERK.

Table 1.

Group classification based on biological function of mRNAs that exhibit GCN2-dependent or PERK-dependent shift into polysomes in response to Met deprivation or tBuHQ treatment, respectively

Biological Process P Value No. of mRNAs Affected
GCN2 dependent
Generation of precursor metabolites and energy 1.50E-06 26
Electron transport 9.39E-06 21
Lipid metabolism 1.41E-04 20
Blood coagulation 3.92E-04 6
Carbohydrate metabolism 9.94E-03 11
Death 1.21E-02 13
Proteolysis 1.52E-02 18
Endocytosis 2.32E-02 7
Steroid metabolism 3.94E-02 5
Alcohol metabolism 4.34E-02 5
PERK dependent
Response to biotic stimulus 2.15E-06 26
Electron transport 1.17E-03 12
Response to wound healing 2.81E-03 9
Response to stress 5.71E-03 20
Humoral immune response 8.45E-03 5
Protein folding 1.25E-02 5
Anion transport 2.08E-02 6
Translation 2.44E-02 7
Programmed cell death 4.37E-02 12

GCN2, general control nonderepressible 2; PERK, double-stranded RNA-activated protein kinase (PKR)-like endoplasmic reticulum-resident kinase; tBuHQ, 2,5-di-tert-butylhydroquinone. The mRNAs in each category are listed in Supplemental Table S9.

Fig. 6.

Fig. 6.

Functional categories of genes whose mRNAs shift into or out of polysomes after Met deprivation, in the presence of GCN2. Gcn2+/+ or −/− mouse livers were perfused for 35 min ± Met, and supernatant was fractioned on a 20–47% sucrose density gradient. Total RNA was extracted from the unfractionated supernatant and the polysomal gradient fraction for use in hybridization array experiments. Analyses of GCN2-dependent shift of mRNAs into polysomes were done by sorting of Z scores of genes in arrays containing unfractionated supernatant and polysomal gradient fraction. Sublists of sorted genes were used for biological process classification in GeneSpring. Bars represent number of genes within the functional category whose mRNA shifted into (positive values) or out of (negative values) polysome fraction. A functional category is included only if it contains ≥3 genes and has a GeneSpring P value of <0.05.

The criteria for designating an mRNA as translationally downregulated in a GCN2-dependent manner were as follows. First, the amount of a particular mRNA had to be the same or more in the total fraction from livers of wild-type mice perfused in the absence compared with the presence of Met (i.e., no decrease in mRNA transcription). Second, the amount of a particular mRNA had to be less in the polysome fraction from livers of Gcn2+/+ mice perfused in the absence compared with the presence of Met (i.e., decreased translation by shifting out of polysomes after stress). Third, the amount of that mRNA had to be the same or greater in the polysome fraction from livers of Gcn2−/− mice perfused in the absence compared with the presence of Met (i.e., decreased translation is dependent on Gcn2). Thus, if an mRNA exhibited decreased polysome abundance in response to Met deprivation in livers of both Gcn2+/+ and Gcn2−/−, it was not considered to be translationally downregulated in a GCN2-dependent manner. As shown in Fig. 5C, 527 mRNAs were found to fit this pattern (also see Supplemental Table S4, tab 2). When grouped by biological function, >100 mRNAs encoding proteins involved in cellular metabolism and >60 mRNAs encoding proteins involved in either cellular protein or macromolecule metabolism shifted out of the polysome fraction.

The criteria used for designating mRNAs as being translationally up- or downregulated in a PERK-dependent manner were similar to those described above for GCN2, except that the experimental conditions utilized tBuHQ treatment rather than Met deprivation. Activation of PERK resulted in an increase in the abundance of 447 mRNAs in the polysome fraction, or ∼2.4% of the 18,700 mRNAs identified by GeneSpring (Fig. 5). Grouping of the mRNAs into functional categories (Fig. 7, Table 1, and Supplemental Table S5) revealed that a number of the biological processes associated with PERK activation were distinct from those affected by GCN2 activation. PERK activation also resulted in 967 mRNAs shifting out of polysomes (Fig. 5C). Of these mRNAs, >60 encode proteins involved in development and >40 are involved in either biosynthesis or cellular biosynthesis (Fig. 7). Notably, of those mRNAs that shifted into the polysome fraction in response to activation of GCN2 or PERK, 11 were common to both groups (Fig. 5B). In addition, 44 mRNAs shifted out of the polysome fraction in response to activation of either kinase (Fig. 5C).

Fig. 7.

Fig. 7.

Functional categories of genes whose mRNAs shift in or out of polysomes after tBuHQ treatment in the presence of PERK. Perk+/+ or −/− mouse livers were perfused for 35 min ± tBuHQ, and supernatant was fractioned on a 20–47% sucrose density gradient. Total RNA was extracted from the unfractionated supernatant and the polysomal gradient fraction for use in hybridization array experiments. Analyses of PERK-dependent shift of mRNAs into polysomes were done by sorting of Z scores of genes in arrays containing unfractionated supernatant and polysomal gradient fraction. Sublists of sorted genes were used for biological process classification in GeneSpring. Bars represent number of genes within the functional category whose mRNA shifted into (positive values) or out of (negative values) polysome fraction. A functional category is included only if it contains ≥3 genes and has a GeneSpring P value of <0.05.

Validation of microarray data.

qRT-PCR was used to validate the changes in mRNA abundance in the polysome fraction for selected mRNAs. The mRNAs selected for validation include mRNAs encoding three putative “housekeeping” proteins (β-actin, GAPDH, and Tbp), six mRNAs whose abundance in polysomes was altered in either a GCN2 [IGFBP-2 (aka BP2/IBP2), Slc3a2 (aka 4F2/4F2HC/CD98/Ly-10/Ly-m10/Mdu1/Mgp-2Hhc), and Trfr2 (aka Tfr2)]- or a PERK [Ldlr (aka Hlb301), Slc2a2 (aka Glut-2), and Stch]-dependent manner, and mRNAs previously reported to be translationally regulated [ATF3, ATF4, Bag-3 (aka Bis) (16, 60), Ddit3 (aka CHOP10/chop/gadd153) (5), and Hspa5 (aka Bip/Hsce70/Sez7/Grp78)]. The Z scores obtained by microarray analysis for the selected mRNAs are shown in Supplemental Table S6. In addition to analysis of mRNA abundance in the polysome fraction, the amount of the individual mRNAs present in the nonpolysomal and subpolysomal fractions was also quantitated. As shown in Fig. 8, A and B, in livers from Gcn2+/+ mice Met deprivation had no significant effect on the abundance of any of the housekeeping mRNAs present in the polysome fraction, except that the amount of Tbp mRNA was decreased in livers of wild-type mice. With the exception of Ldlr, all of the mRNAs identified by microarray analysis as being translationally regulated in response to activation of GCN2 and/or PERK exhibited a shift in distribution from the non- and/or subpolysomal fractions into polysomes (Fig. 8C). In contrast, in livers from Gcn2−/− mice, none of these mRNAs shifted into polysomes (Fig. 8D). Of the mRNAs shown in previous studies to be subject to translational regulation, only Hspa5 shifted into polysomes in response to Met deprivation (Fig. 8E), and the effect was dependent on GCN2 (Fig. 8F).

Fig. 8.

Fig. 8.

qRT-PCR validation of polysome distribution of selected mRNAs in Gcn2 wild-type vs. knockout perfused livers. Mouse livers were perfused for 35 min ± Met. Total RNA was extracted from the unfractionated supernatant and the nonpolysomal, subpolysomal, and polysomal gradient fractions for use in hybridization array and qRT-PCR experiments. Expression of mRNA was determined by qRT-PCR and normalized to the average expression of the housekeeping genes β-actin, Gapdh, and Tbp (see materials and methods for details on calculations). Values are means ± SD; n = 6 or 7. A and B: putative housekeeping genes. C and D: genes identified in this experiment to have shifted into polysomes after activation of GCN2. E and F: genes reported in the literature whose mRNA shifted into polysomes after activation of GCN2. Statistical analysis of the data is presented in Supplemental Table S7.

PERK activation had little effect on the polysomal distribution of the mRNAs encoding the three housekeeping proteins other than GAPDH exhibiting a PERK-dependent shift into polysomes (Fig. 9, A and B). The mRNAs encoding Slc3a2, Trfr2, Ldlr, and Slc2a2 also shifted from the non- and/or subpolysomal fractions into polysomes, and Stch exhibited a similar trend, although the change was not large enough to reach statistical significance (Fig. 9C). Interestingly, all of these mRNAs also shifted into polysomes in livers from Perk−/− mice perfused with tBuHQ (Fig. 9D). Of the five mRNAs previously shown to be subject to translational regulation, all demonstrated a dramatic shift from the non- and/or subpolysome fraction into polysomes in wild-type liver (Fig. 9E). Although there was a trend for these mRNAs to shift into polysomes in tBuHQ-treated livers from Perk−/− mice, only Hspa5 reached statistical significance, and even for that mRNA the magnitude of the shift was blunted in Perk−/− compared with Perk+/+ livers (Fig. 9F). Although the basis for the observed change is unknown, it must occur through a mechanism distinct from phosphorylation of eIF2α and inhibition of eIF2B, because neither was changed in PERK−/− livers treated with tBuHQ.

Fig. 9.

Fig. 9.

qRT-PCR validation of polysome distribution of selected mRNAs in Perk wild-type vs. knockout perfused livers. Mouse livers were perfused for 35 min ± tBuHQ. Total RNA was extracted from the unfractionated supernatant and the nonpolysomal, subpolysomal, and polysomal gradient fractions for use in hybridization array and qRT-PCR experiments. Expression of mRNA was determined by qRT-PCR and normalized to the average expression of the housekeeping genes β-actin, Gapdh, and Tbp (see materials and methods for details on calculations). Values are means ± SD; n = 6 or 7. A and B: putative housekeeping genes. C and D: genes identified in this experiment to have shifted into polysomes after activation of PERK. E and F: genes reported in the literature whose mRNA shifted into polysomes after activation of PERK. Statistical analysis of the data is presented in Supplemental Table S8.

DISCUSSION

In the present study, perfused mouse liver preparations were used to delineate the similarities and differences between activation of GCN2 compared with PERK in the regulation of mRNA translation. Both kinases phosphorylate the same residue, Ser51, on the α-subunit of eIF2 (28, 65). However, the two kinases do not appear to contribute equally to basal eIF2α phosphorylation. Thus, in livers perfused with a complete amino acid mixture in the absence of tBuHQ, eIF2α phosphorylation was similar in wild-type and Gcn2−/− livers, whereas phosphorylation of the protein was lower in livers from Perk−/− compared with Perk+/+ mice. This result suggests that, under the conditions used here, PERK contributes more to basal eIF2α phosphorylation compared with GCN2. Interestingly, eIF2B activity was not significantly different in livers from Perk−/− compared with Perk+/+ mice perfused without tBuHQ, despite the difference in eIF2α phosphorylation. The basis for this finding may lie in the absolute amount of eIF2α present in the phosphorylated form in the control condition. Although not determined for mouse liver, we have previously shown (38) that in rat livers treated with perfusate of the same composition used the present study, <10% of eIF2α is present in the phosphorylated form. Consequently, the difference in eIF2α phosphorylation between livers of Perk−/− and Perk+/+ mice may have caused an increase in eIF2B activity that was too small to detect with the GDP exchange assay. A related observation is that, although eIF2α phosphorylation was increased to a similar extent upon activation of either PERK or GCN2, eIF2B activity was decreased to a greater extent in response to activation of PERK compared with GCN2. This finding suggests that activated PERK regulates additional proteins involved in controlling eIF2B activity. Of the kinases known to phosphorylate eIF2B, GSK-3 is the only one whose action is inhibitory (61). Recent studies (4, 34) demonstrate that activation of either PERK or PKR promotes GSK-3β phosphorylation on Ser9, an event known to inhibit its kinase activity. However, if activated PERK inhibited GSK-3 activity in perfused liver, it would be expected to increase rather than inhibit eIF2B activity. Therefore, it is unlikely that PERK-specific inhibition of GSK-3 explains the differential decrease in eIF2B activity.

Most studies examining alterations in gene expression in response to cell stress have focused on transcriptional rather than translational mechanisms. In this regard, several studies have used hybridization array analysis to document changes in mRNA expression following activation of PERK or GCN2. For example, induction of ER stress in the rat mast cell line RBL-2H3 following a 3-h treatment with tBuHQ increased the expression of mRNAs encoding proteins involved in the immune response and cell cycle regulation (including Gadd153) (54). In another study, induction of ER stress by tunicamycin led to increased expression of Asns, Ddit3, Slc3a2, and Stch mRNA expression in primary fibroblasts (45). Of these, only Asns was found to be transcriptionally regulated in the present study in Gcn2−/− compared with Gcn2+/+ mice. However, all of the mRNAs encoding these proteins, except Asns, were translationally regulated in response to activation of GCN2 and/or PERK.

Changes in mRNA expression in response to activation of either GCN2 or PERK were also observed in the present study. For example, Met deprivation led to a significantly greater increase in expression of the Cyp2c39, Clrf, Isg20, Abhd1, and Hgf mRNAs in the liver of Gcn2−/− compared with Gcn2+/+ mice. In addition to GCN2, the other major intracellular nutrient sensor is mTORC1. However, the observed changes are not likely due to alterations in signaling through mTORC1 because, as noted above, no change in phosphorylation of the mTORC1 substrates 4E-BP1 and S6K1 was observed in livers deprived of methionine. Whether or not another, as yet unidentified, amino acid sensor might exist in mammalian cells (e.g., a protein similar to the yeast Ssy1 protein) is unknown. Less surprising, but still interesting, was the relatively large number of genes that were induced by tBuHQ in the liver of Perk−/− compared with Perk+/+ mice. For example, perfusion with tBuHQ led to a greater increase in expression of the BECN1, EPPK1, CTSB, HIPK2, IGF-6, and ABCC9 mRNAs in the liver of Perk−/− compared with Perk+/+ mice. Although we did not investigate the possibility, the results could be due to induction of other ER stress signaling pathways such as ATF6 or XBP-1.

Translational derepression of ATF4 has been repeatedly documented under different stress conditions in cell models. For example, ATF4 protein expression has been shown to increase after leucine deprivation (31), treatment with thapsigargin (27), or hypoxia (6). In part, the upregulation of ATF4 protein expression is due to a shift in the distribution of the mRNA from small to larger polysomes. A similar phenomenon has been observed for GCN4 in S. cerevisiae in response to nutrient deprivation and treatment with butanol. The proposed model for the derepression event in yeast is that reduction of the met-tRNAi·eIF2·GTP ternary complex allows the ribosomes to bypass uORF and reinitiate at the GCN4 AUG. A similar model has been proposed for the ATF4 mRNA in mammalian cells by Harding et al. (27) and Wek and colleagues (31). In the present study, we observed a shift in the ATF4 mRNA from the subpolysomal to the polysomal fraction in the liver of PERK+/+, but not PERK−/−, mice treated with tBuHQ. At most, only one ribosome can be associated with a given mRNA in the subpolysomal fraction, suggesting that a fraction of the ATF4 mRNA is poorly translated under control conditions. Moreover, upon activation of the UPR, the poorly translated fraction shifts into polysomes, a result consistent with previous studies showing increased expression of the protein. However, even though the magnitude of the increase in eIF2α phosphorylation in the liver of GCN2+/+ mice deprived of Met was similar to that in Perk+/+ mice treated with tBuHQ, there was no significant change in the polysomal distribution of the ATF4 mRNA in response to Met deprivation. The basis for this observation is unknown. However, the finding may relate to the greater reduction in eIF2B activity in response to tBuHQ treatment compared with Met deprivation. The difference in eIF2B activity was mirrored by alterations in polysome aggregation, such that polysome aggregation was reduced to a greater extent in livers perfused with tBuHQ compared with livers deprived of Met. Because the proposed mechanism for upregulation of GCN4 and ATF4 mRNA translation depends on the activity of eIF2B, it is possible that the magnitude of the change induced by Met deprivation for 35 min is not sufficient to cause a significant shift in the ATF4 mRNA from the subpolysomal to the polysomal fraction. It is possible that longer periods of deprivation might promote ATF4 mRNA redistribution. However, such studies cannot be performed with the present model, because global rates of protein synthesis recover over longer periods of amino acid deprivation (e.g., 1–2 h) because of an increase in hepatic proteolysis (23).

Translational upregulation of ATF4 protein has been commonly reported to precede transcriptional upregulation of Ddit3 mRNA (47); however, there is evidence to suggest that the latter can also be translationally regulated, although little is yet known about the mechanism. As shown by the results of the present study, translation of the Ddit3 mRNA is also derepressed in response to activation of PERK, but not GCN2. Derepressed translation of the Ddit3 mRNA subsequently leads to decreased expression of Asns (69). Results from the present study show that the basal mRNA expression of two proteins involved in amino acid metabolism, Asns and Dct, depends on the presence of GCN2. Asns, whose expression was previously shown to depend on the presence of GCN2 (3), encodes asparagine synthetase, an enzyme that catalyzes the hydrolysis of the amido-N group of glutamine, which acts as a donor to convert aspartate to asparagine. Dct, shown to be regulated in response to GCN2 activation for the first time in this study, is involved in tyrosine metabolism and melanin synthesis. Recent reports have placed ATF5 upstream of Asns, proposing that in opposition to Ddit3 (aka CHOP), ATF5 binds to the C/EBP-ATF regulatory sequence in the Asns promoter, thus upregulating Asns gene transcription (1). The decrease in ATF5 and Asns mRNA expression in Gcn2−/− mice observed in this study provides further support for this idea. Although only a small number of transcripts, and none of those identified through the array analysis as being transcriptionally dependent on PERK, have been analyzed by qRT-PCR, their Z score value typically showed smaller changes than those identified in the GCN2 study.

Surprisingly, Hspa5 (GRP78/BiP), an mRNA whose expression (46) and translational state are most closely linked to the UPR, was found in this study also to be regulated after amino acid deprivation. Hspa5 mRNA was shown by Stephens and colleagues (67) to shift into polysomes after UPR, in both the cytosolic and ER-bound mRNA populations. In the present study, the Hspa5 mRNA also shifted into polysomes after activation of either GCN2 or PERK. Interestingly, the mRNA encoding Hspa5 contains an IRES in its 5′-UTR. Even more surprising is the finding that the Stch mRNA shifted into polysomes after Met deprivation but not tBuHQ treatment. Stch mRNA, whose expression is induced by ER stress associated with treatment with the calcium ionophore A-23187, codes for a constitutively expressed, microsome-associated chaperone protein that has a cellular distribution similar to BiP (57). Together, these data would suggest that translation of the Stch mRNA would be expected to be regulated by PERK, but not GCN2.

Regulation of cellular transporter expression appears to be a common response to stress at both the transcriptional (53) and translational levels. A well-studied example of the latter is the amino acid transporter cationic amino acid transporter 1 (Cat-1) mRNA, which contains both an uORF and an IRES (78). Its translation after glucose deprivation is proposed to be PERK dependent and to occur through the cooperative action of the two 5′-UTR structures (22, 78). In the present study, the finding that mRNAs encoding three transporters (Slc2a2, Slc3a2, and Trfr2) shifted into polysomes in response to activation of both PERK and GCN2 suggests a common pool of cellular mRNAs whose translation is derepressed under any type of stress that increases eIF2α phosphorylation. Under either amino acid deprivation or ER stress, increased expression of the protein encoded by these mRNAs would allow cells to increase nutrient uptake (glucose, amino acids, and iron, respectively) that provides building blocks for cellular molecules. Of the three transporters, most is known about the regulation of the Trfr2 mRNA via its iron-regulatory element.

The observation that the mRNAs encoding Igfbp-2, Ldlr, and Bag3 shift into polysomes in response only to activation of either PERK or GCN2 provides an example of unique regulation by each eIF2α kinase. The IGFBP protein family has been strongly implicated in metabolic processes, and transcription of the genes encoding the proteins is modulated after various stresses (3, 40). For example, Igfbp-1 mRNA expression is increased in Leu-deprived HepG2 human hepatoma cells after small interfering RNA (siRNA) knockdown of ATF4 as well as in hepatocytes from Gcn2−/− mice, suggesting that Igfbp-1 expression is regulated by amino acids independently of the GCN2/ATF4 pathway (3). Igfbp-2 expression has been reported to specifically increase after protein or caloric restriction and is involved in regulating liver growth (50). Although Ldlr mRNA expression has been linked to UPR caused by treatment with protease inhibitors (83), and recent work has shown that retention of mutated forms of Ldlr in the ER can lead to UPR and PERK activation (66), little is known about the role of PERK in regulating the translation of mRNAs involved in lipid metabolism. Similarly, besides studies showing that the Bag1 mRNA is translationally regulated through an IRES in the 5′-UTR of its mRNA (11), evidence for regulation of other family members is sparse.

An important question is how activation of one eIF2α kinase might result in differential translation of specific mRNA subsets. Although the answer to this question is unknown, it is tempting to speculate that, in part, the effect may be a result of differences in the subcellular localization of the kinases, leading to a gradient of eIF2α phosphorylation within the cell. Evidence in support of this idea is provided by a recent study (42) showing that activation of PERK in response to ER stress results in the accumulation of eIF2 phosphorylated on Ser51 of the α-subunit in the cytosol adjacent to the ER. The concentration of phosphorylated eIF2 near the ER could preferentially repress the translation of mRNAs destined for that subcellular compartment.

Sequence analyses and experiments initiated in the 1980s by Kozak (43, 44) have popularized the concept of upstream, 5′ noncoding region sequences positively influencing translation initiation. Studies done in yeast and mammalian model systems since then have provided evidence for this theory, and two working models involving leaky scanning or reinitiation in mRNAs containing uORF or cap-independent translation initiation in those containing IRES have evolved (9, 2426, 75, 78, 82). Despite the continuing prolific amount of new data and new mRNA candidates whose translation is being regulated in this manner, the effort continues to be piecemeal. A comprehensive survey of the mammalian genome for genes coding for mRNAs that contain uORF/s or IRES has only been done in silico (in the case of uORF) or not at all (in the case of IRES). Computer analyses of coding sequences have shown that 10–50% of the mammalian mRNAs contain uAUGs (12, 30, 49, 62, 79). Of these, only one-quarter have complete ORF (12, 30, 49, 62, 79). These uORF along with out-of-frame uAUGs are more conserved across species than in-frame uAUGs (12, 30, 49, 62, 79). The uORF code for proteins ranging from 20 to 100 amino acids in length, with some being synthesized in vivo (58). In addition to reconfirming Kozak's rules of conserved bases surrounding the coding sequence AUG and translation initiation at the first AUG, they also presented a population of potential mRNA candidates with uORF that can be regulated by leaky scanning, reinitiation, or ribosome shunting during translation. The percentage of mRNAs identified in the present microarray study that were potentially regulated through these alternative translation initiation pathways is on the low end of the in silico data. Two caveats must be considered: 1) the applied experimental techniques may not favor detection of many mRNAs whose expression is low, and 2) this experimental percentage is a sum of mRNA groups containing uORF or IRES, whereas that from the in silico study may include mainly just those mRNAs containing uORF. A future application for this microarray analysis is to identify biological groups of mRNAs whose translation initiation is similarly regulated after a stressful stimulus.

In summary, the results obtained in the present study show that, despite sharing a common downstream target, GCN2 and PERK regulate the transcription and translation of distinct, as well as overlapping, mRNA subsets. In addition, the percentage of mRNAs translationally regulated after activation of either GCN2 or PERK is in the same range as that identified through sequence analyses to contain uORF. The hybridization array data presented here provide important selective targets for further comparative study concerning GCN2, PERK, and their activating stressors.

GRANTS

This study was supported by National Institute of Diabetes and Digestive and Kidney Diseases Grant DK-13499.

Acknowledgments

We thank Sharon L. Rannels and Jeffrey O'Neil for technical help; Anne Stanley for synthesizing the qRT-PCR primers; Dr. Craig Praul for performing the array hybridization experiments; Daniel Krissinger, Wei Zhao, and Drs. Willard Freeman, Wenlei Liu, and the late Gary Chase for advice and assistance on microarray experimental design and analyses; and Robert Brucklacher and Daniel Krissinger for help with qRT-PCR. We especially thank Daniel Krissinger for bringing the method of Z-score analysis to our attention.

Address for reprint requests and other correspondence: S. R. Kimball, Dept. of Cellular and Molecular Physiology, Pennsylvania State Univ. College of Medicine, PO Box 850, Hershey, PA 17033 (e-mail: skimball@psu.edu).

Footnotes

1

The online version of this article contains supplemental material.

REFERENCES

  • 1.Al Sarraj J, Vinson C, Thiel G. Regulation of asparagine synthetase gene transcription by the basic region leucine zipper transcription factors ATF5 and CHOP. J Biol Chem 386: 873–879, 2005. [DOI] [PubMed] [Google Scholar]
  • 2.Anthony TG, Reiter AK, Anthony JC, Kimball SR, Jefferson LS. Deficiency of dietary EAA preferentially inhibits mRNA translation of ribosomal proteins in liver of meal-fed rats. Am J Physiol Endocrinol Metab 281: E430–E439, 2001. [DOI] [PubMed] [Google Scholar]
  • 3.Averous J, Maurin AC, Bruhat A, Jousse C, Arliguie C, Fafournoux P. Induction of IGFBP-1 expression by amino acid deprivation of HepG2 human hepatoma cells involves both a transcriptional activation and an mRNA stabilization due to its 3′UTR. FEBS Lett 579: 2609–2614, 2005. [DOI] [PubMed] [Google Scholar]
  • 4.Baltzis D, Pluquet O, Papadakis AI, Kazemi S, Qu LK, Koromilas AE. The eIF2α kinases PERK and PKR activate glycogen synthase kinase 3 to promote the proteasomal degradation of p53. J Biol Chem 282: 31675–31687, 2007. [DOI] [PubMed] [Google Scholar]
  • 5.Bi M, Naczki C, Koritzinsky M, Fels D, Blais J, Hu N, Harding H, Novoa I, Varia M, Raleigh J, Scheuner D, Kaufman RJ, Bell J, Ron D, Wouters BG, Koumenis C, Bi M, Naczki C, Koritzinsky M, Fels D, Blais J, Hu N, Harding H, Novoa I, Varia M, Raleigh J, Scheuner D, Kaufman RJ, Bell J, Ron D, Wouters BG, Koumenis C. ER stress-regulated translation increases tolerance to extreme hypoxia and promotes tumor growth. EMBO J 24: 3470–3481, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Blais JD, Filipenko V, Bi M, Harding HP, Ron D, Koumenis C, Wouters BG, Bell JC. Activating transcription factor 4 is translationally regulated by hypoxic stress. Mol Cell Biol 24: 7469–7482, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Brazma A, Hingamp P, Quackenbush J, Sherlock G, Spellman P, Stoeckert C, Aach J, Ansorge W, Ball CA, Causton HC, Gaasterland T, Glenisson P, Holstege FC, Kim IF, Markowitz V, Matese JC, Parkinson H, Robinson A, Sarkans U, Schulze-Kremer S, Stewart J, Taylor R, Vilo J, Vingron M. Minimum information about a microarray experiment (MIAME)—toward standards for microarray data. Nat Genet 29: 365–371, 2001. [DOI] [PubMed] [Google Scholar]
  • 8.Bustin SA Absolute quantification of mRNA using real-time reverse transcription polymerase chain reaction assays. J Mol Endocrinol 25: 169–193, 2000. [DOI] [PubMed] [Google Scholar]
  • 9.Calkhoven CF, Muller C, Martin R, Krosl G, Pietsch H, Hoang T, Leutz A. Translational control of SCL-isoform expression in hematopoietic lineage choice. Genes Dev 17: 959–964, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Cheadle C, Vawter MP, Freed WJ, Becker KG. Analysis of microarray data using Z score transformation. J Mol Diagn 5: 73–81, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Coldwell MJ, deSchoolmeester ML, Fraser GA, Pickering BM, Packham G, Willis AE. The p36 isoform of BAG-1 is translated by internal ribosome entry following heat shock. Oncogene 20: 4095–4100, 2001. [DOI] [PubMed] [Google Scholar]
  • 12.Crowe ML, Wang XQ, Rothnagel JA. Evidence for conservation and selection of upstream open reading frames suggests probable encoding of bioactive peptides. BMC Genomics 7: 16, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.de Haro C, Ochoa S. Further studies on the mode of action of the heme-controlled translational inhibitor. Proc Natl Acad Sci USA 76: 1741–1745, 1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Deng J, Harding HP, Raught B, Gingras AC, Berlanga JJ, Scheuner D, Kaufman RJ, Ron D, Sonenberg N. Activation of GCN2 in UV-irradiated cells inhibits translation. Curr Biol 12: 1279–1286, 2002. [DOI] [PubMed] [Google Scholar]
  • 15.Dever TE, Feng L, Wek RC, Cigan AM, Donahue TF, Hinnebusch AG. Phosphorylation of initiation factor 2alpha by protein kinase GCN2 mediates gene-specific translational control of GCN4 in yeast. Cell 68: 585–596, 1992. [DOI] [PubMed] [Google Scholar]
  • 16.Dobbyn HC, Hill K, Hamilton TL, Spriggs KA, Pickering BM, Coldwell MJ, de Moor CH, Bushell M, Willis AE. Regulation of BAG-1 IRES-mediated translation following chemotoxic stress. Oncogene 27: 1167–1174, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Ehrenfeld E, Manis S. Inhibition of 80S initiation complex formation by infection with poliovirus. J Gen Virol 43: 441–445, 1979. [DOI] [PubMed] [Google Scholar]
  • 18.Everson WV, Flaim KE, Susco DM, Kimball SR, Jefferson LS. Effect of amino acid deprivation on initiation of protein synthesis in rat hepatocytes. Am J Physiol Cell Physiol 256: C18–C27, 1989. [DOI] [PubMed] [Google Scholar]
  • 20.Feldhoff RC, Taylor JM, Jefferson LS. Synthesis and secretion of rat albumin in vivo, in perfused liver, and in isolated hepatocytes. Effects of hypophysectomy and growth hormone treatment. J Biol Chem 252: 3611–3616, 1977. [PubMed] [Google Scholar]
  • 21.Fenwick ML, Walker MJ. Suppression of the synthesis of cellular macromolecules by herpes simplex virus. J Gen Virol 41: 37–51, 1978. [DOI] [PubMed] [Google Scholar]
  • 22.Fernandez J, Bode B, Koromilas A, Diehl JA, Krukovets I, Snider MD, Hatzoglou M. Translation mediated by the internal ribosome entry site of the cat-1 mRNA is regulated by glucose availability in a PERK kinase-dependent manner. J Biol Chem 277: 11780–11787, 2002. [DOI] [PubMed] [Google Scholar]
  • 23.Flaim KE, Peavy DE, Everson WE, Jefferson LS. The role of amino acids in the regulation of protein synthesis in perfused rat liver. I. Reduction in rates of synthesis resulting from amino acid deprivation and recovery during flow-through perfusion. J Biol Chem 257: 2932–2938, 1982. [PubMed] [Google Scholar]
  • 24.Gaba A, Wang Z, Krishnamoorthy T, Hinnebusch AG, Sachs MS. Physical evidence for distinct mechanisms of translational control by upstream open reading frames. EMBO J 20: 6453–6463, 2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Gaccioli F, Huang CC, Wang C, Bevilacqua E, Franchi-Gazzola R, Gazzola GC, Bussolati O, Snider MD, Hatzoglou M. Amino acid starvation induces the SNAT2 neutral amino acid transporter by a mechanism that involves eukaryotic initiation factor 2alpha phosphorylation and cap-independent translation. J Biol Chem 281: 17929–17940, 2006. [DOI] [PubMed] [Google Scholar]
  • 26.Gerlitz G, Jagus R, Elroy-Stein O. Phosphorylation of initiation factor-2alpha is required for activation of internal translation initiation during cell differentiation. Eur J Biochem 269: 2810–2819, 2002. [DOI] [PubMed] [Google Scholar]
  • 27.Harding HP, Novoa I, Zhang Y, Zeng H, Wek R, Schapira M, Ron D. Regulated translation initiation controls stress-induced gene expression in mammalian cells. Mol Cell 6: 1099–1108, 2000. [DOI] [PubMed] [Google Scholar]
  • 28.Harding HP, Zhang Y, Ron D. Protein translation and folding are coupled by an endoplasmic-reticulum-resident kinase. Nature 397: 271–274, 1999. [DOI] [PubMed] [Google Scholar]
  • 29.Hinnebusch AG Mechanism and regulation of initiator methionyl-tRNA binding to ribosome. In: Translational Control of Gene Expression, edited by Sonenberg N, Hershey JWB, Mathews MB. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press, 2000, p. 185–243.
  • 30.Iacono M, Mignone F, Pesole G. uAUG and uORFs in human and rodent 5′untranslated mRNAs. Gene 349: 97–105, 2005. [DOI] [PubMed] [Google Scholar]
  • 31.Jiang HY, Wek SA, McGrath BC, Lu D, Hai T, Harding HP, Wang X, Ron D, Cavener DR, Wek RC. Activating transcription factor 3 is integral to the eukaryotic initiation factor 2 kinase stress response. Mol Cell Biol 24: 1365–1377, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kass GE, Duddy SK, Moore GA, Orrenius S. 2,5-Di-(tert-butyl)-1,4-benzohydroquinone rapidly elevates cytosolic Ca2+ concentration by mobilizing the inositol 1,4,5-trisphosphate-sensitive Ca2+ pool. J Biol Chem 264: 15192–15198, 1989. [PubMed] [Google Scholar]
  • 33.Katze MG, Detjen BM, Safer B, Krug RM. Translational control by influenza virus: suppression of the kinase that phosphorylates the alpha subunit of initiation factor eIF-2 and selective translation of influenza viral mRNAs. Mol Cell Biol 6: 1741–1750, 1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Kazemi S, Mounir Z, Baltzis D, Raven JF, Wang S, Krishnamoorthy JL, Pluquet O, Pelletier J, Koromilas AE. A novel function of eIF2α kinases as inducers of the phosphoinositide-3 kinase signaling pathway. Mol Biol Cell 18: 3635–3644, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kimball SR Regulation of global and specific mRNA translation by amino acids. J Nutr 132: 883–886, 2002. [DOI] [PubMed] [Google Scholar]
  • 36.Kimball SR, Antonetti DA, Brawley RM, Jefferson LS. Mechanism of inhibition of peptide chain initiation by amino acid deprivation in perfused rat liver. Regulation involving inhibition of eukaryotic initiation factor 2alpha phosphatase activity. J Biol Chem 266: 1969–1976, 1991. [PubMed] [Google Scholar]
  • 37.Kimball SR, Everson WV, Myers LM, Jefferson LS. Purification and characterization of eukaryotic initiation factor 2 and a guanine nucleotide exchange factor from rat liver. J Biol Chem 262: 2220–2227, 1987. [PubMed] [Google Scholar]
  • 38.Kimball SR, Jefferson LS. Inhibition of microsomal calcium sequestration causes an impairment of initiation of protein synthesis in perfused rat liver. Biochem Biophys Res Commun 177: 1082–1086, 1991. [DOI] [PubMed] [Google Scholar]
  • 39.Kimball SR, Anthony TG, Cavener DR, Jefferson LS. Nutrient signaling through mammalian GCN2. Top Curr Genet 7: 113–130, 2004. [Google Scholar]
  • 40.Kokkinakis DM, Liu X, Chada S, Ahmed MM, Shareef MM, Singha UK, Yang S, Luo J. Modulation of gene expression in human central nervous system tumors under methionine deprivation-induced stress. Cancer Res 64: 7513–7525, 2004. [DOI] [PubMed] [Google Scholar]
  • 41.Komar AA, Hatzoglou M. Internal ribosome entry sites in cellular mRNAs: mystery of their existence. J Biol Chem 280: 23425–23428, 2005. [DOI] [PubMed] [Google Scholar]
  • 42.Kondratyev M, Avezov E, Shenkman M, Groisman B, Lederkremer GZ. PERK-dependent compartmentalization of ERAD and unfolded protein response machineries during ER stress. Exp Cell Res 313: 3395–3407, 2007. [DOI] [PubMed] [Google Scholar]
  • 43.Kozak M Compilation and analysis of sequences upstream from the translational start site in eukaryotic mRNAs. Nucleic Acids Res 12: 857–872, 1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Kozak M Effects of intercistronic length on the efficiency of reinitiation by eucaryotic ribosomes. Mol Cell Biol 7: 3438–3445, 1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Lecca MR, Wagner U, Patrignani A, Berger EG, Hennet T. Genome-wide analysis of the unfolded protein response in fibroblasts from congenital disorders of glycosylation type-I patients. FASEB J 19: 240–242, 2005. [DOI] [PubMed] [Google Scholar]
  • 45a.Loreni F, Amaldi F. Translational regulation of ribosomal protein synthesis in Xenopus cultured cells: mRNA relocation between polysomes and RNP during nutritional shifts. Eur J Biochem 205: 1027–1032, 1992. [DOI] [PubMed] [Google Scholar]
  • 46.Luo S, Baumeister P, Yang S, Abcouwer SF, Lee AS. Induction of Grp78/BiP by translational block: activation of the Grp78 promoter by ATF4 through and upstream ATF/CRE site independent of the endoplasmic reticulum stress elements. J Biol Chem 278: 37375–37385, 2003. [DOI] [PubMed] [Google Scholar]
  • 47.Ma Y, Brewer JW, Diehl JA, Hendershot LM. Two distinct stress signaling pathways converge upon the CHOP promoter during the mammalian unfolded protein response. J Mol Biol 318: 1351–1365, 2002. [DOI] [PubMed] [Google Scholar]
  • 48.Mazan-Mamczarz K, Kawai T, Martindale JL, Gorospe M. En masse analysis of nascent translation using microarrays. Biotechniques 39: 61–67, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Meijer HA, Thomas AA. Control of eukaryotic protein synthesis by upstream open reading frames in the 5′-untranslated region of an mRNA. Biochem J 367: 1–11, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Miller RA, Chang Y, Galecki AT, Al-Regaiey K, Kopchick JJ, Bartke A. Gene expression patterns in calorically restricted mice: partial overlap with long-lived mutant mice. Mol Endocrinol 16: 2657–2666, 2002. [DOI] [PubMed] [Google Scholar]
  • 51.Moore GA, McConkey DJ, Kass GE, O'Brien PJ, Orrenius S. 2,5-Di(tert-butyl)-1,4-benzohydroquinone—a novel inhibitor of liver microsomal Ca2+ sequestration. FEBS Lett 224: 331–336, 1987. [DOI] [PubMed] [Google Scholar]
  • 53.Murray JI, Whitfield ML, Trinklein ND, Myers RM, Brown PO, Botstein D. Diverse and specific gene expression responses to stresses in cultured human cells. Mol Biol Cell 15: 2361–2374, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Nakamura R, Ishida S, Ozawa S, Saito Y, Okunuki H, Teshima R, Sawada J. Gene expression profiling of Ca2+-ATPase inhibitor DTBHQ and antigen-stimulated RBL-2H3 mast cells. Inflamm Res 51: 611–618, 2002. [DOI] [PubMed] [Google Scholar]
  • 55.Novoa I, Zeng H, Harding HP, Ron D. Feedback inhibition of the unfolded protein response by GADD34-mediated dephosphorylation of eIF2alpha. J Cell Biol 153: 1011–1022, 2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.O'Neill RE, Racaniello VR. Inhibition of translation in cells infected with a poliovirus 2Apro mutant correlates with phosphorylation of the alpha subunit of eucaryotic initiation factor 2. J Virol 63: 5069–5075, 1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Otterson GA, Flynn GC, Kratzke RA, Coxon A, Johnston PG, Kaye FJ. Stch encodes the “ATPase core” of a microsomal stress 70 protein. EMBO J 13: 1216–1225, 1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Oyama M, Itagaki C, Hata H, Suzuki Y, Izumi T, Natsume T, Isobe T, Sugano S. Analysis of small human proteins reveals the translation of upstream open reading frames of mRNAs. Genome Res 14: 2048–2052, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Pavio N, Romano PR, Graczyk TM, Feinstone SM, Taylor DR. Protein synthesis and endoplasmic reticulum stress can be modulated by the hepatitis C virus envelope protein E2 through the eukaryotic initiation factor 2alpha kinase PERK. J Virol 77: 3578–3585, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Pickering BM, Mitchell SA, Spriggs KA, Stoneley M, Willis AE. Bag-1 internal ribosome entry segment activity is promoted by structural changes mediated by poly(rC) binding protein 1 and recruitment of polypyrimidine tract binding protein 1. Mol Cell Biol 24: 5595–5605, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Proud CG Regulation of eukaryotic initiation factor eIF2B. Prog Mol Subcell Biol 26: 95–114, 2001. [DOI] [PubMed] [Google Scholar]
  • 62.Rogozin IB, Kochetov AV, Kondrashov FA, Koonin EV, Milanesi L. Presence of ATG triplets in 5′ untranslated regions of eukaryotic cDNAs correlates with a “weak” context of the start codon. Bioinformatics 17: 890–900, 2001. [DOI] [PubMed] [Google Scholar]
  • 63.Scorsone KA, Panniers R, Rowlands AG, Henshaw EC. Phosphorylation of eukaryotic initiation factor 2 during physiological stresses which affect protein synthesis. J Biol Chem 262: 14538–14543, 1987. [PubMed] [Google Scholar]
  • 64.Siekierka J, Mariano TM, Reichel PA, Mathews MB. Translational control by adenovirus: lack of virus-associated RNAI during adenovirus infection results in phosphorylation of initiation factor eIF-2 and inhibition of protein synthesis. Proc Natl Acad Sci USA 82: 1959–1963, 1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Sood R, Porter AC, Olsen DA, Cavener DR, Wek RC. A mammalian homologue of GCN2 protein kinase important for translational control by phosphorylation of eukaryotic initiation factor-2alpha. Genetics 154: 787–801, 2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Sorensen S, Ranheim T, Bakken KS, Leren TP, Kulseth MA. Retention of mutant low density lipoprotein receptor in endoplasmic reticulum (ER) leads to ER stress. J Biol Chem 281: 468–476, 2006. [DOI] [PubMed] [Google Scholar]
  • 67.Stephens SB, Dodd RD, Brewer JW, Lager PJ, Keene JD, Nicchitta CV. Stable ribosome binding to the endoplasmic reticulum enables compartment-specific regulation of mRNA translation. Mol Biol Cell 16: 5819–5831, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Stoneley M, Willis AE. Cellular internal ribosome entry segments: structures, trans-acting factors and regulation of gene expression. Oncogene 23: 3200–3207, 2004. [DOI] [PubMed] [Google Scholar]
  • 69.Su N, Kilberg MS. C/EBP homology protein (CHOP) interacts with activating transcription factor 4 (ATF4) and negatively regulates the stress-dependent induction of the asparagine synthetase gene. J Biol Chem 283: 35106–35117, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Sudhakar A, Ramachandran A, Ghosh S, Hasnain SE, Kaufman RJ, Ramaiah KV. Phosphorylation of serine 51 in initiation factor 2alpha (eIF2alpha) promotes complex formation between eIF2alpha(P) and eIF2B and causes inhibition in the guanine nucleotide exchange activity of eIF2B. Biochemistry 39: 12929–12938, 2000. [DOI] [PubMed] [Google Scholar]
  • 71.Svitkin YV, Gradi A, Imataka H, Morino S, Sonenberg N. Eukaryotic initiation factor 4GII (eIF4GII), but not eIF4GI, cleavage correlates with inhibition of host cell protein synthesis after human rhinovirus infection. J Virol 73: 3467–3472, 1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Tolman EL, Schworer CM, Jefferson LS. Effects of hypophysectomy on amino acid metabolism and gluconeogenesis in the perfused rat liver. J Biol Chem 248: 4552–4560, 1973. [PubMed] [Google Scholar]
  • 73.Tzamarias D, Roussou I, Thireos G. Coupling of GCN4 mRNA translational activation with decreased rates of polypeptide chain initiation. Cell 57: 947–954, 1989. [DOI] [PubMed] [Google Scholar]
  • 74.Vattem KM, Wek RC. Reinitiation involving upstream ORFs regulates ATF4 mRNA translation in mammalian cells. Proc Natl Acad Sci USA 101: 11269–11274, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Wang XQ, Rothnagel JA. 5′-Untranslated regions with multiple upstream AUG codons can support low-level translation via leaky scanning and reinitiation. Nucleic Acids Res 32: 1382–1391, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Wek RC, Jiang HY, Anthony TG. Coping with stress: eIF2 kinases and translational control. Biochem Soc Trans 34: 7–11, 2006. [DOI] [PubMed] [Google Scholar]
  • 77.Willcocks MM, Carter MJ, Roberts LO. Cleavage of eukaryotic initiation factor eIF4G and inhibition of host-cell protein synthesis during feline calicivirus infection. J Gen Virol 85: 1125–1130, 2004. [DOI] [PubMed] [Google Scholar]
  • 78.Yaman I, Fernandez J, Liu H, Caprara M, Komar AA, Koromilas AE, Zhou L, Snider MD, Scheuner D, Kaufman RJ, Hatzoglou M. The zipper model of translational control: a small upstream ORF is the switch that controls structural remodeling of an mRNA leader. Cell 113: 519–531, 2003. [DOI] [PubMed] [Google Scholar]
  • 79.Yamashita R, Suzuki Y, Nakai K, Sugano S. Small open reading frames in 5′ untranslated regions of mRNAs. C R Biol 326: 987–991, 2003. [DOI] [PubMed] [Google Scholar]
  • 80.Zhang P, McGrath B, Li S, Frank A, Zambito F, Reinert J, Gannon M, Ma K, McNaughton K, Cavener DR. The PERK eukaryotic initiation factor 2alpha kinase is required for the development of the skeletal system, postnatal growth, and the function and viability of the pancreas. Mol Cell Biol 22: 3864–3874, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Zhang P, McGrath BC, Reinert J, Olsen DS, Lei L, Gill S, Wek SA, Vattem KM, Wek RC, Kimball SR, Jefferson LS, Cavener DR. The GCN2 eIF2alpha kinase is required for adaptation to amino acid deprivation in mice. Mol Cell Biol 22: 6681–6688, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Zhang Y, Li W, Vore M. Translational regulation of rat multidrug resistance-associated protein 2 expression is mediated by upstream open reading frames in the 5′ untranslated region. Mol Pharmacol 71: 377–383, 2007. [DOI] [PubMed] [Google Scholar]
  • 83.Zhou H, Pandak WM Jr, Lyall V, Natarajan R, Hylemon PB. HIV protease inhibitors activate the unfolded protein response in macrophages: implication for atherosclerosis and cardiovascular disease. Mol Pharmacol 68: 690–700, 2005. [DOI] [PubMed] [Google Scholar]

Articles from Physiological Genomics are provided here courtesy of American Physiological Society

RESOURCES