Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
JIMD Rep ; 64(2): 167-179, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36873094

ABSTRACT

Asparagine synthetase (ASNS) catalyzes the synthesis of asparagine (Asn) from aspartate and glutamine. Biallelic mutations in the ASNS gene result in ASNS Deficiency (ASNSD). Children with ASNSD exhibit congenital microcephaly, epileptic-like seizures, and continued brain atrophy, often leading to premature mortality. This report describes a 4-year-old male with global developmental delay and seizures with two novel mutations in the ASNS gene, c.614A > C (maternal) and c.1192dupT (paternal) encoding p.H205P and p.Y398Lfs*4 variants, respectively. We employed the novel use of immortalized lymphoblastoid cell lines (LCL) to show that the proliferation of the heterozygotic parental LCL was not severely affected by culture in Asn-free medium, but growth of the child's cells was suppressed by about 50%. Asn production by the LCL from both the father and the child was significantly decreased relative to the mother's cells. mRNA and protein analysis of the paternal LCL cells for the Y398Lfs*4 variant revealed reductions in both. Attempts to ectopically express the truncated Y398Lfs*4 variant in either HEK293T or ASNS-null cells resulted in little or no detectable protein. Expression and purification of the H205P variant from HEK293T cells revealed enzymatic activity similar to wild-type ASNS. Stable expression of WT ASNS rescued the growth of ASNS-null JRS cells in Asn-free medium and the H205P variant was only slightly less effective. However, the Y398Lfs*4 variant appeared to be unstable in JRS cells. These results indicate that co-expression of the H205P and Y398Lfs*4 variants leads to a significant reduction in Asn synthesis and cellular growth.

2.
J Biol Chem ; 299(1): 102786, 2023 01.
Article in English | MEDLINE | ID: mdl-36509145

ABSTRACT

Escherichia coli YoaA aids in the resolution of DNA damage that halts DNA synthesis in vivo in conjunction with χ, an accessory subunit of DNA polymerase III. YoaA and χ form a discrete complex separate from the DNA polymerase III holoenzyme, but little is known about how YoaA and χ work together to help the replication fork overcome damage. Although YoaA is predicted to be an iron-sulfur helicase in the XPD/Rad3 helicase family based on sequence analysis, the biochemical activities of YoaA have not been described. Here, we characterize YoaA and show that purified YoaA contains iron. YoaA and χ form a complex that is stable through three chromatographic steps, including gel filtration chromatography. When overexpressed in the absence of χ, YoaA is mostly insoluble. In addition, we show the YoaA-χ complex has DNA-dependent ATPase activity. Our measurement of the YoaA-χ helicase activity illustrates for the first time YoaA-χ translocates on ssDNA in the 5' to 3' direction and requires a 5' single-stranded overhang, or ssDNA gap, for DNA/DNA unwinding. Furthermore, YoaA-χ preferentially unwinds forked duplex DNA that contains both 3' and 5' single-stranded overhangs versus duplex DNA with only a 5' overhang. Finally, we demonstrate YoaA-χ can unwind damaged DNA that contains an abasic site or damage on 3' ends that stall replication extension. These results are the first biochemical evidence demonstrating YoaA is a bona fide iron-sulfur helicase, and we further propose the physiologically relevant form of the helicase is YoaA-χ.


Subject(s)
DNA Helicases , DNA Polymerase III , Escherichia coli Proteins , Escherichia coli , DNA Helicases/metabolism , DNA Polymerase III/genetics , DNA Replication , DNA, Single-Stranded , Escherichia coli/metabolism , Iron , Escherichia coli Proteins/metabolism , DNA Repair
3.
DNA Repair (Amst) ; 100: 103006, 2021 04.
Article in English | MEDLINE | ID: mdl-33582602

ABSTRACT

Efficient and faithful replication of DNA is essential for all organisms. However, the replication fork frequently encounters barriers that need to be overcome to ensure cell survival and genetic stability. Cells must carefully balance and regulate replication vs. repair reactions. In Escherichia coli, the replisome consists of the DNA polymerase III holoenzyme, including DNA polymerase, proofreading exonuclease, processivity clamp and clamp loader, as well as a fork helicase, DnaB and primase, DnaG. We provide evidence here that one component of the clamp loader complex, HolC (or χ) plays a dual role via its ability to form 2 mutually exclusive complexes: one with HolD (or ψ) that recruits the clamp-loader and hence the DNA polymerase holoenzyme and another with helicase-like YoaA protein, a DNA-damage inducible repair protein. By yeast 2 hybrid analysis, we show that two residues of HolC, F64 and W57, at the interface in the structure with HolD, are required for interaction with HolD and for interaction with YoaA. Mutation of these residues does not interfere with HolC's interaction with single-strand DNA binding protein, SSB. In vivo, these mutations fail to complement the poor growth and sensitivity to azidothymidine, a chain-terminating replication inhibitor. In support of the notion that these are exclusive complexes, co-expression of HolC, HolD and YoaA, followed by pulldown of YoaA, yields a complex with HolC but not HolD. YoaA fails to pulldown HolC-F64A. We hypothesize that HolC, by binding with SSB, can recruit the DNA polymerase III holoenzyme through HolD, or an alternative repair complex with YoaA helicase.


Subject(s)
DNA Polymerase III/metabolism , DNA Repair , DNA Replication , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , DNA, Bacterial/metabolism , Escherichia coli/genetics , Protein Binding , Protein Conformation
4.
Sci Rep ; 10(1): 1691, 2020 02 03.
Article in English | MEDLINE | ID: mdl-32015389

ABSTRACT

Protein functional constraints are manifest as superfamily and functional-subgroup conserved residues, and as pairwise correlations. Deep Analysis of Residue Constraints (DARC) aids the visualization of these constraints, characterizes how they correlate with each other and with structure, and estimates statistical significance. This can identify determinants of protein functional specificity, as we illustrate for bacterial DNA clamp loader ATPases. These load ring-shaped sliding clamps onto DNA to keep polymerase attached during replication and contain one δ, three γ, and one δ' AAA+ subunits semi-circularly arranged in the order δ-γ1-γ2-γ3-δ'. Only γ is active, though both γ and δ' functionally influence an adjacent γ subunit. DARC identifies, as functionally-congruent features linking allosterically the ATP, DNA, and clamp binding sites: residues distinctive of γ and of γ/δ' that mutually interact in trans, centered on the catalytic base; several γ/δ'-residues and six γ/δ'-covariant residue pairs within the DNA binding N-termini of helices α2 and α3; and γ/δ'-residues associated with the α2 C-terminus and the clamp-binding loop. Most notable is a trans-acting γ/δ' hydroxyl group that 99% of other AAA+ proteins lack. Mutation of this hydroxyl to a methyl group impedes clamp binding and opening, DNA binding, and ATP hydrolysis-implying a remarkably clamp-loader-specific function.


Subject(s)
DNA-Binding Proteins/metabolism , Protein Subunits/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Binding Sites/physiology , DNA Polymerase III/metabolism , DNA, Bacterial/metabolism , Escherichia coli/metabolism , Hydrolysis , Protein Structure, Secondary , Sensitivity and Specificity
5.
Biochim Biophys Acta Mol Cell Res ; 1866(3): 371-381, 2019 03.
Article in English | MEDLINE | ID: mdl-30290239

ABSTRACT

Endoplasmic reticulum (ER) stress activates three principal signaling pathways, collectively known as the unfolded protein response, leading to translational and transcriptional control mechanisms that dictate the cell's response as adaptive or apoptotic. The present study illustrates that for HepG2 human hepatocellular carcinoma cells the signaling pathways triggered by ER stress extend beyond the three principal pathways to include mitogen-activated protein kinase (MAPK) signaling, leading to activation of transcription from the early growth response 1 (EGR1) gene. Analysis provided evidence for a SRC-RAS-RAF-MEK-ERK cascade mechanism that leads to enhanced phosphorylation of the transcription factor ELK1. ELK1 and serum response factor (SRF) are constitutively bound to the EGR1 promoter and are phosphorylated by nuclear localized ERK. The promoter abundance of both phospho-SRF and phopsho-ELK1 was increased by ER stress, but the SRF phosphorylation was transient. Knockdown of ELK1 had little effect on the basal EGR1 mRNA content, but completely blocked the increase in response to ER stress. Conversely, knockdown of SRF suppressed basal EGR1 mRNA content, but had only a small effect on the induction by ER stress. This research highlights the importance of MAPK signaling in response to ER stress and identifies ELK1 as a transcriptional mediator and the EGR1 gene as a target.


Subject(s)
Early Growth Response Protein 1/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Carcinoma, Hepatocellular/pathology , DNA-Binding Proteins/metabolism , Early Growth Response Protein 1/biosynthesis , Early Growth Response Protein 1/genetics , Endoplasmic Reticulum Stress/physiology , Gene Expression Regulation/genetics , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Phosphorylation , Signal Transduction , Transcription Factors/metabolism , ets-Domain Protein Elk-1/genetics , ets-Domain Protein Elk-1/metabolism
6.
Mol Genet Metab ; 123(3): 317-325, 2018 03.
Article in English | MEDLINE | ID: mdl-29279279

ABSTRACT

Asparagine Synthetase Deficiency (ASD) is a recently described inborn error of metabolism caused by bi-allelic pathogenic variants in the asparagine synthetase (ASNS) gene. ASD typically presents congenitally with microcephaly and severe, often medically refractory, epilepsy. Development is generally severely affected at birth. Tone is abnormal with axial hypotonia and progressive appendicular spasticity. Hyperekplexia has been reported. Neuroimaging typically demonstrates gyral simplification, abnormal myelination, and progressive cerebral atrophy. The present report describes two siblings from consanguineous parents with a homozygous Arg49Gln variant associated with a milder form of ASD that is characterized by later onset of symptoms. Both siblings had a period of normal development before onset of seizures, and development regression. Primary fibroblast studies of the siblings and their parents document that homozygosity for Arg49Gln blocks cell growth in the absence of extracellular asparagine. Functional studies with these cells suggest no impact of the Arg49Gln variant on basal ASNS mRNA or protein levels, nor on regulation of the gene itself. Molecular modelling of the ASNS protein structure indicates that the Arg49Gln variant lies near the substrate binding site for glutamine. Collectively, the results suggest that the Arg49Gln variant affects the enzymatic function of ASNS. The clinical, cellular, and molecular observations from these siblings expand the known phenotypic spectrum of ASD.


Subject(s)
Amino Acid Metabolism, Inborn Errors/genetics , Aspartate-Ammonia Ligase/genetics , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/genetics , Intellectual Disability/genetics , Seizures/genetics , Arginine/genetics , Asparagine/biosynthesis , Aspartate-Ammonia Ligase/deficiency , Binding Sites/genetics , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/metabolism , Child , Child, Preschool , Consanguinity , DNA Mutational Analysis , Female , Fibroblasts/metabolism , Glutamine/genetics , Glutamine/metabolism , Homozygote , Humans , Male , Models, Molecular , Mutation , Siblings
7.
Biochim Biophys Acta ; 1853(3): 539-48, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25523140

ABSTRACT

Amino acid (AA) deprivation in mammalian cells activates a collection of signaling cascades known as the AA response (AAR), which is characterized by transcriptional induction of stress-related genes, including FBJ murine osteosarcoma viral oncogene homolog (cFOS). The present study established that the signaling mechanism underlying the AA-dependent transcriptional regulation of the cFOS gene in HepG2 human hepatocellular carcinoma cells is independent of the classic GCN2-eIF2-ATF4 pathway. Instead, a RAS-RAF-MEK-ERK cascade mediates AAR signaling to the cFOS gene. Increased cFOS transcription is observed from 4-24 h after AAR-activation, exhibiting little or no overlap with the rapid and transient increase triggered by the well-known serum response. Furthermore, serum is not required for the AA-responsiveness of the cFOS gene and no phosphorylation of promoter-bound serum response factor (SRF) is observed. The ERK-phosphorylated transcription factor E-twenty six-like (p-ELK1) is increased in its association with the cFOS promoter after activation of the AAR. This research identified cFOS as a target of the AAR and further highlights the importance of AA-responsive MAPK signaling in HepG2 cells.


Subject(s)
Amino Acids/deficiency , Carcinoma, Hepatocellular/genetics , Genes, fos/genetics , Liver Neoplasms/genetics , MAP Kinase Signaling System/physiology , Activating Transcription Factor 4/physiology , Amino Acids/pharmacology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cells, Cultured , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Protein Serine-Threonine Kinases/physiology , Transcriptional Activation/drug effects
8.
J Biol Chem ; 286(42): 36724-38, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-21862593

ABSTRACT

Mammalian cells respond to protein or amino acid (AA) limitation by activating a number of signaling pathways, collectively referred to as the AA response (AAR), that modulate a range of cellular functions, including transcriptional induction of target genes. This study demonstrates that in hepatocellular carcinoma cells, expression of c-JUN, JUN-B, c-FOS, and FOS-B was induced by the AAR, whereas JUN-D, FRA-1, and FRA-2 were not. Of the four activated FOS/JUN members, c-JUN made the largest contribution to the induction of several known AAR target genes. For several human liver, prostate, and ovarian cell lines, the AAR-induced increase in c-JUN expression was greater in transformed cells compared with nontransformed counterparts, an effect independent of cell growth rate. Thus far, the best characterized AA-responsive genes are all transcriptionally activated by ATF4, but the AAR-dependent induction of c-JUN transcription was ATF4-independent. The increased expression of c-JUN was dependent on ATF2 and on activation of the MEK-ERK and JNK arms of the MAPK signaling pathways. Formation of c-JUN-ATF2-activated heterodimers was increased after AA limitation, and c-JUN or ATF2 knockdown suppressed the induction of c-JUN and other AAR target genes. AA deprivation triggers a feed-forward process that involves phosphorylation of existing c-JUN protein by JNK and subsequent auto-activation of the c-JUN gene by recruitment of c-JUN and ATF2 to two AP-1 sites within the proximal promoter. The results document the novel observation that AP-1 sequences within the c-JUN gene can function as transcriptional amino acid-response elements.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Gene Expression Regulation, Neoplastic , Genes, jun , Liver Neoplasms/metabolism , MAP Kinase Signaling System , Oncogene Protein p65(gag-jun)/biosynthesis , Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Amino Acids/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Gene Knockdown Techniques , Genes, fos/genetics , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Oncogene Protein p65(gag-jun)/genetics , Phosphorylation/genetics , Response Elements/genetics , Transcription, Genetic/genetics
9.
Nucleic Acids Res ; 36(17): 5571-80, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18757893

ABSTRACT

It is unclear whether Mediator complex in yeast is necessary for all RNA polymerase II (Pol II) transcription or if it is limited to genes activated by environmental stress. In mammals, amino acid limitation induces SNAT2 transcription through ATF4 binding at an amino acid response element. ATF4 is the functional counterpart to the yeast amino acid-dependent regulator GCN4 and GCN4 recruits Mediator during transcriptional activation. Consistent with enhanced SNAT2 transcription activity, the present data demonstrate that amino acid limitation increased SNAT2 promoter association of the general transcription factors that make up the preinitiation complex, including Pol II, but there was no increase in Mediator recruitment. Furthermore, siRNA knockdown of eight Mediator subunits caused no significant decrease in SNAT2 transcription. The estrogen-dependent pS2 gene was used as a positive control for both the ChIP and the siRNA approaches and the data demonstrated the requirement for Mediator recruitment. These results document that activation of the SNAT2 gene by the mammalian amino acid response pathway occurs independently of enhanced Mediator recruitment.


Subject(s)
Amino Acid Transport System A/genetics , Amino Acids/metabolism , Response Elements , Transcription Factors, General/metabolism , Transcriptional Activation , Cell Line, Tumor , Histones/metabolism , Humans , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , RNA Interference , Transcription Factors, General/antagonists & inhibitors , Transcription Factors, General/genetics
10.
Biochem J ; 410(3): 473-84, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18052938

ABSTRACT

A nutrient stress signalling pathway is triggered in response to protein or amino acid deprivation, namely the AAR (amino acid response), and previous studies have shown that C/EBPbeta (CCAAT/enhancer-binding protein beta) expression is up-regulated following activation of the AAR. DNA-binding studies, both in vitro and in vivo, have revealed increased C/EBPbeta association with AARE (AAR element) sequences in AAR target genes, but its role is still unresolved. The present results show that in HepG2 human hepatoma cells, the total amount of C/EBPbeta protein, both the activating [LAP* and LAP (liver-enriched activating protein)] and inhibitory [LIP (liver-enriched inhibitory)] isoforms, was increased in histidine-deprived cells. Immunoblotting of subcellular fractions and immunostaining revealed that most of the C/EBPbeta was located in the nucleus. Consistent with these observations, amino acid limitation caused an increase in C/EBPbeta DNA-binding activity in nuclear extracts and chromatin immunoprecipitation revealed an increase in C/EBPbeta binding to the AARE region in vivo, but at a time when transcription from the target gene was declining. A constant fraction of the basal and increased C/EBPbeta protein was phosphorylated on Thr(235) and the phospho-C/EBPbeta did bind to an AARE. Induction of AARE-enhanced transcription was slightly greater in C/EBPbeta-deficient MEFs (mouse embryonic fibroblasts) or C/EBPbeta siRNA (small interfering RNA)-treated HepG2 cells compared with the corresponding control cells. Transient expression of LAP*, LAP or LIP in C/EBPbeta-deficient fibroblasts caused suppression of increased transcription from an AARE-driven reporter gene. Collectively, the results demonstrate that C/EBPbeta is not required for transcriptional activation by the AAR pathway but, when present, acts in concert with ATF3 (activating transcription factor 3) to suppress transcription during the latter stages of the response.


Subject(s)
Amino Acids/administration & dosage , CCAAT-Enhancer-Binding Protein-beta/biosynthesis , Dietary Proteins/administration & dosage , Repressor Proteins/biosynthesis , Transcription, Genetic , Animals , Base Sequence , Blotting, Western , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , DNA Primers , Electrophoretic Mobility Shift Assay , Immunohistochemistry , Liver/metabolism , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Repressor Proteins/metabolism
11.
Biochem J ; 391(Pt 3): 649-58, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16026328

ABSTRACT

For animals, dietary protein is critical for the nutrition of the organism and, at the cellular level, protein nutrition translates into amino acid availability. Amino acid deprivation triggers the AAR (amino acid response) pathway, which causes enhanced transcription from specific target genes. The present results show that C/EBPbeta (CCAAT/enhancer-binding protein beta) mRNA and protein content were increased following the deprivation of HepG2 human hepatoma cells of a single amino acid. Although there was a modest increase in mRNA half-life following histidine limitation, the primary mechanism for the elevated steady-state mRNA was increased transcription. Transient transfection documented that C/EBPbeta genomic fragments containing the 8451 bp 5' upstream of the transcription start site did not contain amino-acid-responsive elements. However, deletion analysis of the genomic region located 3' downstream of the protein coding sequence revealed that a 93 bp fragment contained an amino-acid-responsive activity that functioned as an enhancer. Exogenous expression of ATF4 (activating transcription factor 4), known to activate other genes through amino acid response elements, caused increased transcription from reporter constructs containing the C/EBPbeta enhancer in cells maintained in complete amino acid medium. Chromatin immunoprecipitation demonstrated that RNA polymerase II is bound at the C/EBPbeta promoter and at the 93 bp regulatory region in vivo, whereas ATF4 binds to the enhancer region only. Immediately following amino acid removal, the kinetics of binding for ATF4, ATF3, and C/EBPbeta itself to the 93 bp regulatory region were similar to those observed for the amino-acid-responsive asparagine synthetase gene. Collectively the findings show that expression of C/EBPbeta, which contributes to the regulation of amino-acid-responsive genes, is itself controlled by amino acid availability through transcription.


Subject(s)
Amino Acids/deficiency , CCAAT-Enhancer-Binding Protein-beta/genetics , Enhancer Elements, Genetic/genetics , Transcription, Genetic/genetics , Activating Transcription Factor 4/genetics , Amino Acids/metabolism , Amino Acids/pharmacology , Cell Line, Tumor , Gene Deletion , Humans , Open Reading Frames , Protein Binding , RNA, Messenger/metabolism , Transcription, Genetic/drug effects , Up-Regulation
12.
J Biol Chem ; 279(49): 50829-39, 2004 Dec 03.
Article in English | MEDLINE | ID: mdl-15385533

ABSTRACT

Expression of human asparagine synthetase (ASNS), which catalyzes asparagine and glutamate biosynthesis, is transcriptionally induced following amino acid deprivation. Previous overexpression and electrophoresis mobility shift analysis showed the involvement of the transcription factors ATF4, C/EBPbeta, and ATF3-FL through the nutrient-sensing response element-1 (NSRE-1) within the ASNS promoter. Amino acid deprivation caused an elevated mRNA level for ATF4, C/EBPbeta, and ATF3-FL, and the present study established that the nuclear protein content for ATF4 and ATF3-FL were increased during amino acid limitation, whereas C/EBPbeta-LIP declined slightly. The total amount of C/EBPbeta-LAP protein was unchanged, but changes in the distribution among multiple C/EBPbeta-LAP forms were observed. Overexpression studies established that ATF4, ATF3-FL, and C/EBPbeta-LAP could coordinately modulate the transcription from the human ASNS promoter. Chromatin immunoprecipitation demonstrated that amino acid deprivation increased ATF3-FL, ATF4, and C/EBPbeta binding to the ASNS promoter and enhanced promoter association of RNA polymerase II, TATA-binding protein, and TFIIB of the general transcription machinery. A time course revealed a markedly different temporal order of interaction between these transcription factors and the ASNS promoter. During the initial 2 h, there was a 20-fold increase in ATF4 binding and a rapid increase in histone H3 and H4 acetylation, which closely paralleled the increased transcription rate of the ASNS gene, whereas the increase in ATF3-FL and C/EBPbeta binding was considerably slower and more closely correlated with the decline in transcription rate between 2 and 6 h. The data suggest that ATF3-FL and C/EBPbeta act as transcriptional suppressors for the ASNS gene to counterbalance the transcription rate activated by ATF4 following amino acid deprivation.


Subject(s)
Aspartate-Ammonia Ligase/biosynthesis , Aspartate-Ammonia Ligase/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Histones/metabolism , Promoter Regions, Genetic , Trans-Activators/metabolism , Transcription, Genetic , Acetylation , Activating Transcription Factor 3 , Amino Acids/chemistry , Cell Line , Cell Line, Tumor , Chromatin/chemistry , Chromatin/metabolism , Chromatin Immunoprecipitation , Histones/chemistry , Humans , Immunoblotting , Kinetics , Models, Biological , Plasmids/metabolism , Protein Binding , RNA/chemistry , RNA Polymerase II/chemistry , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , TATA-Box Binding Protein/chemistry , Time Factors , Transcription Factor TFIIB/chemistry , Transfection
13.
J Biol Chem ; 279(27): 27948-56, 2004 Jul 02.
Article in English | MEDLINE | ID: mdl-15102854

ABSTRACT

CCAAT/enhancer-binding protein beta (C/EBPbeta) is a member of the bZIP family of transcription factors that contribute to the regulation of a wide range of important cellular processes. The data in the present study document that transcription from the human C/EBPbeta gene is induced in response to endoplasmic reticulum stress, such as glucose deprivation, or treatment of cells with tunicamycin or thapsigargin. Transient transfection of C/EBPbeta genomic fragments linked to a luciferase reporter gene demonstrated that the C/EBPbeta promoter plays no major regulatory role. Instead, by deletion analysis it was discovered that a 46-bp region, located at a genomic site that corresponds to the 3'-untranslated region of the C/EBPbeta mRNA, harbored an element that was required for the stress response. Mutagenesis demonstrated that a cis-regulatory element located at nt +1614-1621 (5'-TGACGCAA-3') is responsible for activation of the C/EBPbeta gene. Electrophoresis mobility shift analysis revealed that proteins are bound to this element and that the amount of binding is increased following glucose deprivation. This element is homologous to a previously reported mammalian unfolded protein response element that binds XBP-1. Consistent with those data, overexpression of XBP-1 caused an increase in transcription that was mediated by the C/EBPbeta mammalian unfolded protein response element.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/biosynthesis , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Response Elements , Base Sequence , Binding Sites , Blotting, Northern , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Gene Deletion , Genes, Reporter , Glucose/metabolism , Humans , Immunoblotting , Luciferases/metabolism , Models, Genetic , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Folding , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thapsigargin/pharmacology , Time Factors , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Activation , Transfection , Tunicamycin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...