Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cell Death Dis ; 10(12): 903, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31787756

ABSTRACT

Endoplasmic reticulum (ER) stress signaling plays a critical role in the control of cell survival or death. Persistent ER stress activates proapoptotic pathway involving the ATF4/CHOP axis. Although accumulating evidences support its important contribution to cardiovascular diseases, but its mechanism is not well characterized. Here, we demonstrate a critical role for PRMT1 in the control of ER stress in cardiomyocytes. The inhibition of PRMT1 augments tunicamycin (TN)-triggered ER stress response in cardiomyocytes while PRMT1 overexpression attenuates it. Consistently, PRMT1 null hearts show exacerbated ER stress and cell death in response to TN treatment. Interestingly, ATF4 depletion attenuates the ER stress response induced by PRMT1 inhibition. The methylation-deficient mutant of ATF4 with the switch of arginine 239 to lysine exacerbates ER stress accompanied by enhanced levels of proapoptotic cleaved Caspase3 and phosphorylated-γH2AX in response to TN. The mechanistic study shows that PRMT1 modulates the protein stability of ATF4 through methylation. Taken together, our data suggest that ATF4 methylation on arginine 239 by PRMT1 is a novel regulatory mechanism for protection of cardiomyocytes from ER stress-induced cell death.


Subject(s)
Activating Transcription Factor 4/metabolism , Endoplasmic Reticulum Stress , Myocytes, Cardiac/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Activating Transcription Factor 4/chemistry , Activating Transcription Factor 4/genetics , Amino Acid Sequence , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Line , Endoplasmic Reticulum Stress/drug effects , Humans , Methylation/drug effects , Mutation/genetics , Myocytes, Cardiac/drug effects , Organ Specificity , Protein Binding/drug effects , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Rats , Transcription Factor CHOP/metabolism , Tunicamycin/pharmacology , Up-Regulation/drug effects
2.
RNA Biol ; 14(12): 1660-1667, 2017 12 02.
Article in English | MEDLINE | ID: mdl-28745933

ABSTRACT

Reinitiation after translation of short upstream ORFs (uORFs) represents one of the means of regulation of gene expression on the mRNA-specific level in response to changing environmental conditions. Over the years it has been shown-mainly in budding yeast-that its efficiency depends on cis-acting features occurring in sequences flanking reinitiation-permissive uORFs, the nature of their coding sequences, as well as protein factors acting in trans. We earlier demonstrated that the first two uORFs from the reinitiation-regulated yeast GCN4 mRNA leader carry specific structural elements in their 5' sequences that interact with the translation initiation factor eIF3 to prevent full ribosomal recycling post their translation. Actually, this interaction turned out to be instrumental in stabilizing the mRNA·40S post-termination complex, which is thus capable to eventually resume scanning and reinitiate on the next AUG start site downstream. Recently, we also provided important in vivo evidence strongly supporting the long-standing idea that to stimulate reinitiation, eIF3 has to remain bound to ribosomes elongating these uORFs until their stop codon has been reached. Here we examined the importance of eIF3 and sequences flanking uORF1 of the human functional homolog of yeast GCN4, ATF4, in stimulation of efficient reinitiation. We revealed that the molecular basis of the reinitiation mechanism is conserved between yeasts and humans.


Subject(s)
Eukaryotic Initiation Factor-3/metabolism , Open Reading Frames , Peptide Chain Initiation, Translational , Activating Transcription Factor 4/chemistry , Activating Transcription Factor 4/metabolism , Animals , Eukaryotic Initiation Factor-3/chemistry , Humans , Mammals , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/metabolism
3.
Fish Shellfish Immunol ; 64: 155-164, 2017 May.
Article in English | MEDLINE | ID: mdl-28263879

ABSTRACT

PRKRA (interferon-inducible double-stranded RNA-dependent protein kinase activator A) is a protective protein which regulates the adaptation of cells to ER stress and virus-stimulated signaling pathways by activating PKR. In the present study, a grass carp (Ctenopharyngodon idella) PRKRA full-length cDNA (named CiPRKRA, KT891991) was cloned and identified. The full-length cDNA is comprised of a 5' UTR (36 bp), a 3' UTR (350 bp) and the longest ORF (882 bp) encoding a polypeptide of 293 amino acids. The deduced amino acid sequence of CiPRKRA contains three typical dsRNA binding motifs (dsRBM). Phylogenetic tree analysis revealed a closer evolutionary relationship of CiPRKRA with other fish PRKRA, especially with Danio rerio PRKRA. qRT-PCR showed that CiPRKRA was significantly up-regulated after stimulation with tunicamycin (Tm) and Poly I:C in C. idella kidney (CIK) cells. To further study its transcriptional regulation, the partial promoter sequence of CiPRKRA (1463 bp) containing one ISRE and one CARE was cloned by Tail-PCR. Subsequently, grass carp IRF2 (CiIRF2) and ATF4 (CiATF4) were expressed in Escherichia coli BL21 and purified by affinity chromatography with the Ni-NTA His-Bind Resin. In vitro, both CiIRF2 and CiATF4 bound to CiPRKRA promoter with high affinity by gel mobility shift assays, revealing that IRF2 and ATF4 might be potential transcriptional regulatory factors for CiPRKRA. Dual-luciferase reporter assays were applied to further investigate the transcriptional regulation of CiPRKRA in vivo. Recombinant plasmid of pGL3-PRKRAPro was constructed and transiently co-transfected into CIK cells with pcDNA3.1-CiIRF2 and pcDNA3.1-CiATF4, respectively. The results showed that both CiIRF2 and CiATF4 significantly decreased the luciferase activity of pGL3-PRKRAPro, suggesting that they play a negative role in CiPRKRA transcription.


Subject(s)
Activating Transcription Factor 4/genetics , Carps/physiology , Fish Proteins/genetics , Gene Expression Regulation/genetics , Interferon Regulatory Factor-2/genetics , Activating Transcription Factor 4/chemistry , Activating Transcription Factor 4/metabolism , Amino Acid Sequence , Animals , Antiviral Agents/pharmacology , Base Sequence , Carps/genetics , Carps/immunology , Cloning, Molecular , DNA, Complementary/genetics , DNA, Complementary/metabolism , Fish Proteins/chemistry , Fish Proteins/metabolism , Gene Expression Regulation/drug effects , Interferon Regulatory Factor-2/chemistry , Interferon Regulatory Factor-2/metabolism , Phylogeny , Poly I-C/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tunicamycin/pharmacology
4.
Amino Acids ; 48(12): 2831-2842, 2016 12.
Article in English | MEDLINE | ID: mdl-27613409

ABSTRACT

Amino-acid deprivation is sensed by the eIF2α kinase GCN2. Under conditions of essential amino-acid limitation, GCN2 phosphorylates eIF2α, inhibiting the formation of a new ternary complex and hence mRNA translation initiation. While decreasing global mRNA translation, eIF2α phosphorylation also increases the translation of the integrated stress response (ISR) transcription factor ATF4, which increases the expression of many stress response genes that contain a C/EBP-ATF response element (CARE), including Atf4, 4Ebp1, Asns, and Chop. Using wild-type as well as Gcn2 knockout and unphosphorylatable eIF2α mutant MEFs, we characterized a novel GCN2/eIF2α phosphorylation-independent, but ATF4-dependent, pathway that upregulates the expression of CARE-containing genes in MEFs lacking GCN2 or phosphorylatable eIF2α when these cells are exposed to methionine-deficient, and to a lesser extent arginine- or histidine-deficient, medium. Thus, we demonstrate a GCN2/eIF2α phosphorylation-independent pathway that converges with the GCN2/eIF2α kinase-dependent pathway at the level of ATF4 and similarly results in the upregulation of CARE-containing genes. We hypothesize that the essential role of methionine-charged initiator tRNA in forming ternary complex is responsible for the robust ability of methionine deficiency to induce ATF4 and the ISR even in the absence of GCN2 or eIF2α kinase activity.


Subject(s)
Activating Transcription Factor 4/genetics , Glial Cell Line-Derived Neurotrophic Factor/genetics , Methionine/metabolism , Protein Biosynthesis , Protein Serine-Threonine Kinases/genetics , Activating Transcription Factor 4/chemistry , Activating Transcription Factor 4/metabolism , Amino Acids/metabolism , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Fibroblasts , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Methionine/deficiency , Methionine/genetics , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/biosynthesis , Signal Transduction , Ternary Complex Factors/chemistry , Ternary Complex Factors/genetics , Transcriptional Activation/genetics
5.
Fish Shellfish Immunol ; 58: 59-66, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27623341

ABSTRACT

Drosophila Toll and mammalian Toll-like receptors (TLRs) are a family of evolutionarily conserved immune receptors that play a crucial role in the first-line defense against intruded pathogens. Activating transcription factor 4 (ATF4), a member of the ATF/CREB transcription factor family, is an important factor that participates in TLR signaling and other physiological processes. However, in crustaceans, whether ATF4 homologs were involved in TLR signaling remains unclear. In the current study, we identified a Toll homolog PcToll2 and a novel ATF4 homolog PcATF4 from Procambarus clarkii, and analyzed the likely regulatory activity of PcATF4 in PcToll2 signaling. The complete cDNA sequence of PcToll2 was 4175 bp long containing an open reading frame of 2820 bp encoding a 939-amino acid protein, and the cDNA sequence of PcATF4 was 2027 bp long with an open reading frame of 1296 bp encoding a 431-amino acid protein. PcToll2 and human TLR4 shared the high identity and they were grouped into a cluster. Furthermore, PcToll2 had a close relationship with other shrimp TLRs that possessed potential antibacterial activity. PcToll2 was highly expressed in the hemocytes, heart and gills, while PcATF4 mainly distributed in gills. Upon challenge with Vibrio parahemolyticus, PcToll2 and PcATF4 together with the antimicrobial peptides of ALF1 and ALF2 were significantly up-regulated in the hemocytes, and the PcATF4 was translocated into the nucleus. After PcToll2 silencing and challenge with Vibrio, the translocation of PcATF4 into the nucleus was inhibited and the expression of ALF1 and ALF2 was reduced, but the expression of PcDorsal and PcSTAT was not affected. Furthermore, after PcATF4 knockdown and challenge with or without Vibrio, the expression of ALF1 and ALF2 was also decreased while the expression of PcToll2 was upregulated. These results suggested that PcToll2 might regulate the expression of ALF1 and ALF2 by promoting the import of PcATF4, instead of the routine transcription factor PcDorsal, into the nucleus participating in the immune defense against Gram-negative bacteria.


Subject(s)
Antimicrobial Cationic Peptides/genetics , Arthropod Proteins/metabolism , Astacoidea/immunology , Astacoidea/microbiology , Toll-Like Receptors/metabolism , Vibrio parahaemolyticus/physiology , Activating Transcription Factor 4/chemistry , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Arthropod Proteins/chemistry , Arthropod Proteins/genetics , Arthropod Proteins/immunology , Astacoidea/classification , Astacoidea/genetics , DNA, Complementary/genetics , DNA, Complementary/metabolism , Gene Expression Regulation , Phylogeny , Protein Transport/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sequence Analysis, Protein , Tissue Distribution , Toll-Like Receptors/chemistry , Toll-Like Receptors/genetics
6.
PLoS One ; 10(6): e0130635, 2015.
Article in English | MEDLINE | ID: mdl-26086088

ABSTRACT

Perturbation of endoplasmic reticulum (ER) homeostasis triggers the ER stress response (also known as Unfolded Protein Response), a hallmark of many pathological disorders. However the connection between ER stress and inflammation remains largely unexplored. Recent data suggest that ER stress controls the activity of inflammasomes, key signaling platforms that mediate innate immune responses. Here we report that expression of NLRP1, a core inflammasome component, is specifically up-regulated during severe ER stress conditions in human cell lines. Both IRE1α and PERK, but not the ATF6 pathway, modulate NLRP1 gene expression. Furthermore, using mutagenesis, chromatin immunoprecipitation and CRISPR-Cas9-mediated genome editing technology, we demonstrate that ATF4 transcription factor directly binds to NLRP1 promoter during ER stress. Although involved in different types of inflammatory responses, XBP-1 splicing was not required for NLRP1 induction. This study provides further evidence that links ER stress with innate.


Subject(s)
Activating Transcription Factor 4/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Inflammasomes/metabolism , Activating Transcription Factor 4/chemistry , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , CRISPR-Cas Systems/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endoribonucleases/metabolism , HCT116 Cells , HeLa Cells , Humans , Jurkat Cells , K562 Cells , Mutagenesis , NLR Proteins , Promoter Regions, Genetic , Protein Binding , Protein Serine-Threonine Kinases/metabolism , RNA Splicing , Real-Time Polymerase Chain Reaction , Regulatory Factor X Transcription Factors , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation , X-Box Binding Protein 1 , eIF-2 Kinase/metabolism
7.
J Biol Chem ; 290(18): 11749-61, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25795775

ABSTRACT

The RET proto-oncogene, a tyrosine kinase receptor, is widely known for its essential role in cell survival. Germ line missense mutations, which give rise to constitutively active oncogenic RET, were found to cause multiple endocrine neoplasia type 2, a dominant inherited cancer syndrome that affects neuroendocrine organs. However, the mechanisms by which RET promotes cell survival and prevents cell death remain elusive. We demonstrate that in addition to cytoplasmic localization, RET is localized in the nucleus and functions as a tyrosine-threonine dual specificity kinase. Knockdown of RET by shRNA in medullary thyroid cancer-derived cells stimulated expression of activating transcription factor 4 (ATF4), a master transcription factor for stress-induced apoptosis, through activation of its target proapoptotic genes NOXA and PUMA. RET knockdown also increased sensitivity to cisplatin-induced apoptosis. We observed that RET physically interacted with and phosphorylated ATF4 at tyrosine and threonine residues. Indeed, RET kinase activity was required to inhibit the ATF4-dependent activation of the NOXA gene because the site-specific substitution mutations that block threonine phosphorylation increased ATF4 stability and activated its targets NOXA and PUMA. Moreover, chromatin immunoprecipitation assays revealed that ATF4 occupancy increased at the NOXA promoter in TT cells treated with tyrosine kinase inhibitors or the ATF4 inducer eeyarestatin as well as in RET-depleted TT cells. Together these findings reveal RET as a novel dual kinase with nuclear localization and provide mechanisms by which RET represses the proapoptotic genes through direct interaction with and phosphorylation-dependent inactivation of ATF4 during the pathogenesis of medullary thyroid cancer.


Subject(s)
Activating Transcription Factor 4/metabolism , Apoptosis , Proto-Oncogene Proteins c-ret/metabolism , Activating Transcription Factor 4/chemistry , Active Transport, Cell Nucleus/drug effects , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cisplatin/pharmacology , Gene Expression Regulation/drug effects , Humans , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein Kinase Inhibitors/pharmacology , Proteolysis/drug effects , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Threonine/metabolism , Transcription, Genetic/drug effects
8.
Genome Res ; 23(6): 988-97, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23590861

ABSTRACT

To evaluate the effect of CG methylation on DNA binding of sequence-specific B-ZIP transcription factors (TFs) in a high-throughput manner, we enzymatically methylated the cytosine in the CG dinucleotide on protein binding microarrays. Two Agilent DNA array designs were used. One contained 40,000 features using de Bruijn sequences where each 8-mer occurs 32 times in various positions in the DNA sequence. The second contained 180,000 features with each CG containing 8-mer occurring three times. The first design was better for identification of binding motifs, while the second was better for quantification. Using this novel technology, we show that CG methylation enhanced binding for CEBPA and CEBPB and inhibited binding for CREB, ATF4, JUN, JUND, CEBPD, and CEBPG. The CEBPB|ATF4 heterodimer bound a novel motif CGAT|GCAA 10-fold better when methylated. The electrophoretic mobility shift assay (EMSA) confirmed these results. CEBPB ChIP-seq data using primary female mouse dermal fibroblasts with 50× methylome coverage for each strand indicate that the methylated sequences well-bound on the arrays are also bound in vivo. CEBPB bound 39% of the methylated canonical 10-mers ATTGC|GCAAT in the mouse genome. After ATF4 protein induction by thapsigargin which results in ER stress, CEBPB binds methylated CGAT|GCAA in vivo, recapitulating what was observed on the arrays. This methodology can be used to identify new methylated DNA sequences preferentially bound by TFs, which may be functional in vivo.


Subject(s)
Activating Transcription Factor 4/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , CpG Islands , DNA Methylation , Activating Transcription Factor 4/chemistry , Animals , Base Sequence , Binding Sites , CCAAT-Enhancer-Binding Protein-beta/chemistry , Female , Fibroblasts , Mice , Nucleotide Motifs , Position-Specific Scoring Matrices , Protein Binding/drug effects , Protein Multimerization , Thapsigargin/immunology , Transcription Factors/metabolism
9.
BMC Genomics ; 13: 665, 2012 Nov 24.
Article in English | MEDLINE | ID: mdl-23176727

ABSTRACT

BACKGROUND: The present study coupled expression profiling with chromatin immunoprecipitation sequencing (ChIP-seq) to examine peroxisome proliferator-activated receptor-ß/δ (PPARß/δ)-dependent regulation of gene expression in mouse keratinocytes, a cell type that expresses PPARß/δ in high concentration. RESULTS: Microarray analysis elucidated eight different types of regulation that modulated PPARß/δ-dependent gene expression of 612 genes ranging from repression or activation without an exogenous ligand, repression or activation with an exogenous ligand, or a combination of these effects. Bioinformatic analysis of ChIP-seq data demonstrated promoter occupancy of PPARß/δ for some of these genes, and also identified the presence of other transcription factor binding sites in close proximity to PPARß/δ bound to chromatin. For some types of regulation, ATF4 is required for ligand-dependent induction of PPARß/δ target genes. CONCLUSIONS: PPARß/δ regulates constitutive expression of genes in keratinocytes, thus suggesting the presence of one or more endogenous ligands. The diversity in the types of gene regulation carried out by PPARß/δ is consistent with dynamic binding and interactions with chromatin and indicates the presence of complex regulatory networks in cells expressing high levels of this nuclear receptor such as keratinocytes. Results from these studies are the first to demonstrate that differences in DNA binding of other transcription factors can directly influence the transcriptional activity of PPARß/δ.


Subject(s)
Activating Transcription Factor 4/metabolism , Gene Expression Profiling , Gene Expression Regulation , PPAR delta/genetics , PPAR delta/metabolism , PPAR-beta/genetics , Regulatory Sequences, Nucleic Acid/genetics , Activating Transcription Factor 4/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Chromatin/metabolism , DNA/metabolism , Genomics , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Transcription, Genetic
10.
Gene Expr Patterns ; 9(5): 335-40, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19232401

ABSTRACT

FIAT represses osteocalcin gene transcription by heterodimerizing with ATF4 and preventing it from binding to DNA. We report here the expression profiles of FIAT and ATF4 during osteoblastogenesis. Messenger RNA levels for the osteoblast transcriptional regulators Satb2, Runx2, Fiat, and Atf4 were quantified using real-time reverse-transcription PCR (RT-qPCR) and respective protein levels monitored by immunodetection in differentiating primary osteoblast cultures. Satb2, Fiat, and Atf4 mRNA levels remained constant throughout the differentiation sequence, whereas Runx2 transcript levels were significantly increased by 12 days post-confluency. Using immunofluorescence, the SATB2, RUNX2, and ATF4 signals appeared to increase as a function of time in culture. FIAT protein expression was readily detected in early cultures, but signal intensity decreased thereafter. When immunoblotting was used to quantify the relative amounts of FIAT and ATF4 proteins, the expression levels of the two proteins were found to be inversely correlated. The decrease in FIAT protein levels coincided with increased binding of ATF4 to the osteocalcin gene promoter, and with increased osteocalcin expression measured by RT-qPCR or immunoblotting. Immunohistochemistry of long bones from mice at E16.5 and 2 days post-natal revealed that both proteins are initially expressed in osteoblasts. In adult bone, FIAT was detected in osteocytes, while ATF4 expression was observed in active osteoblasts and lining cells, but not in osteocytes. Taken together, these data support the idea that a stoichiometric excess of ATF4 over FIAT in mature osteoblasts releases ATF4 from sequestration by FIAT, thereby allowing ATF4 homodimerization and subsequent transactivation of the osteocalcin gene.


Subject(s)
Activating Transcription Factor 4/genetics , Carrier Proteins/genetics , Gene Expression Profiling , Nuclear Proteins/genetics , Osteoblasts/metabolism , Activating Transcription Factor 4/chemistry , Activating Transcription Factor 4/metabolism , Animals , Blotting, Western , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Co-Repressor Proteins , Femur/growth & development , Femur/metabolism , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Immunohistochemistry , Mice , Mice, Inbred C57BL , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Osteoblasts/cytology , Osteocalcin/genetics , Osteocytes/cytology , Osteocytes/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Protein Multimerization , Reverse Transcriptase Polymerase Chain Reaction , Tibia/growth & development , Tibia/metabolism , Time Factors
11.
J Immunol ; 182(1): 466-76, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19109178

ABSTRACT

To understand pathways mediating the inflammatory responses of human aortic endothelial cells to oxidized phospholipids, we previously used a combination of genetics and genomics to model a coexpression network encompassing >1000 genes. CHAC1 (cation transport regulator-like protein 1), a novel gene regulated by ox-PAPC (oxidized 1-palmitoyl-2-arachidonyl-sn-3-glycero-phosphorylcholine), was identified in a co-regulated group of genes enriched for components of the ATF4 (activating transcription factor 4) arm of the unfolded protein response pathway. Herein, we characterize the role of CHAC1 and validate the network model. We first define the activation of CHAC1 mRNA by chemical unfolded protein response-inducers, but not other cell stressors. We then define activation of CHAC1 by the ATF4-ATF3-CHOP (C/EBP homologous protein), and not parallel XBP1 (X box-binding protein 1) or ATF6 pathways, using siRNA and/or overexpression plasmids. To examine the subset of genes downstream of CHAC1, we used expression microarray analysis to identify a list of 227 differentially regulated genes. We validated the activation of TNFRSF6B (tumor necrosis factor receptor superfamily, member 6b), a FASL decoy receptor, in cells treated with CHAC1 small interfering RNA. Finally, we showed that CHAC1 overexpression enhanced apoptosis, while CHAC1 small interfering RNA suppressed apoptosis, as determined by TUNEL, PARP (poly(ADP-ribose) polymerase) cleavage, and AIF (apoptosis-inducing factor) nuclear translocation.


Subject(s)
Activating Transcription Factor 3/chemistry , Activating Transcription Factor 4/chemistry , Apoptosis Regulatory Proteins/chemistry , Protein Folding , Signal Transduction/drug effects , Signal Transduction/immunology , Transcription Factor CHOP/chemistry , Vesicular Transport Proteins/chemistry , Activating Transcription Factor 3/genetics , Apoptosis Inducing Factor/physiology , Apoptosis Regulatory Proteins/physiology , Dithiothreitol/pharmacology , Gene Expression Profiling , HeLa Cells , Humans , Oligonucleotide Array Sequence Analysis , Phosphatidylcholines/pharmacology , Poly(ADP-ribose) Polymerases/physiology , Transcription Factor CHOP/genetics , Tunicamycin/pharmacology , Vesicular Transport Proteins/biosynthesis , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/physiology
13.
Int J Biochem Cell Biol ; 40(1): 14-21, 2008.
Article in English | MEDLINE | ID: mdl-17466566

ABSTRACT

Activating transcription factor 4 (ATF4) belongs to the ATF/CREB (activating transcription factor/cyclic AMP response element binding protein) family of basic region-leucine zipper (bZip) transcription factors, which have the consensus binding site cAMP responsive element (CRE). ATF4 has numerous dimerization partners. ATF4 is induced by stress signals including anoxia/hypoxia, endoplasmic reticulum stress, amino acid deprivation, and oxidative stress. ATF4 expression is regulated transcriptionally, translationally via the PERK pathway of eIF2alpha phosphorylation, and posttranslationally by phosphorylation, which targets ATF4 to proteasomal degradation. ATF4 regulates the expression of genes involved in oxidative stress, amino acid synthesis, differentiation, metastasis and angiogenesis. Transgenic studies have demonstrated ATF4 to be involved in hematopoiesis, lens and skeletal development, fertility, proliferation, differentiation, and long-term memory. ATF4 expression is upregulated in cancer. Since ATF4 is induced by tumour microenvironmental factors, and regulates processes relevant to cancer progression, it might serve as a potential therapeutic target in cancer.


Subject(s)
Activating Transcription Factor 4 , Activating Transcription Factor 4/chemistry , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Animals, Genetically Modified , Cell Differentiation , Gene Expression Regulation , Hematopoiesis/physiology , Humans , Hypoxia/metabolism , Mice , Oxidative Stress/physiology , Transcription, Genetic
14.
Biochemistry ; 47(1): 14-29, 2008 Jan 08.
Article in English | MEDLINE | ID: mdl-18052253

ABSTRACT

ATF4 plays a crucial role in the cellular response to stress. The E3 ubiquitin ligase, SCF beta-TrCP protein responsible for ATF4 degradation by the proteasome, binds to ATF4 through a DpSGXXXpS phosphorylation motif, which is similar but not identical to the DpSGXXpS motif found in most other substrates of beta-TrCP. NMR studies were performed on the free and bound forms of a peptide derived from this ATF4 motif that enabled the elucidation of the conformation of the ligand complexed to the beta-TrCP protein and its binding mode. Saturation transfer difference (STD) NMR allowed the study of competition for binding to beta-TrCP, between the phosphorylation motifs of ATF4 and beta-catenin, to characterize the ATF4 binding epitope. Docking protocols were performed using the crystal structure of the beta-catenin-beta-TrCP complex as a template and NMR results of the ATF4-beta-TrCP complex. In agreement with the STD results, in order to bind to beta-TrCP, the ATF4 DpSGIXXpSXE motif required the association of two negatively charged areas, in addition to the hydrophobic interaction in the beta-TrCP central channel. Docking studies showed that the ATF4 DpSGIXXpSXE motif fits the binding pocket of beta-TrCP through an S-turning conformation. The distance between the two phosphate groups is 17.8 A, which matched the corresponding distance 17.1 A for the other extended DpSGXXpS motif in the beta-TrCP receptor model. This study identifies the residues of the beta-TrCP receptor involved in ligand recognition. Using a new concept of STD competition experiment, we show that ATF4 competes and inhibits binding of beta-catenin to beta-TrCP.


Subject(s)
Activating Transcription Factor 4/chemistry , Magnetic Resonance Spectroscopy/methods , Peptides/chemistry , beta Catenin/chemistry , beta-Transducin Repeat-Containing Proteins/chemistry , Amino Acid Motifs , Amino Acid Sequence , Humans , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Peptides/chemical synthesis , Peptides/metabolism , Phosphorylation , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , beta Catenin/metabolism , beta-Transducin Repeat-Containing Proteins/genetics , beta-Transducin Repeat-Containing Proteins/metabolism
15.
Peptides ; 28(12): 2253-67, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17996332

ABSTRACT

ATF4 plays a crucial role in the cellular response to stress and the F-box protein beta-TrCP, the receptor component of the SCF E3 ubiquitin ligase responsible for ATF4 degradation by the proteasome, binds to ATF4, and controls its stability. Association between the two proteins depends on ATF4 phosphorylation of serine residues 219 and 224 present in the context of DpSGXXXpS, which is similar but not identical to the DpSGXXpS motif found in most other substrates of beta-TrCP. We used NMR spectroscopy to analyze the structure of the 23P-ATF4 peptide. The 3D structure of the ligand was determined on the basis of NOESY restraints that provide an hairpin loop structure. In contrast, no ordered structure was observed in the NMR experiments for the nonphosphorylated 23-ATF4 in solution. This structural study provides information, which could be used to study the beta-TrCP receptor-ligand interaction in docking procedure. Docking studies showed that the binding epitope of the ligand, is represented by the DpSGIXXpSXE motif. 23P-ATF4 peptide fits the binding pocket of protein beta-TrCP very well, considering that the DpSGIXXpSXE motif adopts an S-turning conformation contrary to the extended DpSGXXpS motif in the other known beta-TrCP ligands.


Subject(s)
Activating Transcription Factor 4/chemistry , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Humans , Hydrogen Bonding , Ligands , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphorylation , Protein Conformation , beta-Transducin Repeat-Containing Proteins/chemistry , beta-Transducin Repeat-Containing Proteins/metabolism
16.
Blood ; 110(10): 3610-7, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17684156

ABSTRACT

The activating transcription factor-4 (ATF-4) is translationally induced under anoxic conditions, mediates part of the unfolded protein response following endoplasmic reticulum (ER) stress, and is a critical regulator of cell fate. Here, we identified the zipper II domain of ATF-4 to interact with the oxygen sensor prolyl-4-hydroxylase domain 3 (PHD3). The PHD inhibitors dimethyloxalylglycine (DMOG) and hypoxia, or proteasomal inhibition, all induced ATF-4 protein levels. Hypoxic induction of ATF-4 was due to increased protein stability, but was independent of the ubiquitin ligase von Hippel-Lindau protein (pVHL). A novel oxygen-dependent degradation (ODD) domain was identified adjacent to the zipper II domain. Mutations of 5 prolyl residues within this ODD domain or siRNA-mediated down-regulation of PHD3, but not of PHD2, was sufficient to stabilize ATF-4 under normoxic conditions. These data demonstrate that PHD-dependent oxygen-sensing recruits both the hypoxia-inducible factor (HIF) and ATF-4 systems, and hence not only confers adaptive responses but also cell fate decisions.


Subject(s)
Activating Transcription Factor 4/metabolism , Dioxygenases/physiology , Oxygen/pharmacology , Protein Processing, Post-Translational/drug effects , Activating Transcription Factor 4/chemistry , Amino Acid Sequence , Cell Hypoxia/physiology , Dioxygenases/chemistry , HeLa Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid
17.
Nucleic Acids Res ; 35(17): 5954-65, 2007.
Article in English | MEDLINE | ID: mdl-17726049

ABSTRACT

When an essential amino acid is limited, a signaling cascade is triggered that leads to increased translation of the 'master regulator', activating transcription factor 4 (ATF4), and resulting in the induction of specific target genes. Binding of ATF4 to the amino acid response element (AARE) is an essential step in the transcriptional activation of CHOP (a CCAAT/enhancer-binding protein-related gene) by amino acid deprivation. We set out to identify proteins that interact with ATF4 and that play a role in the transcriptional activation of CHOP. Using a tandem affinity purification (TAP) tag approach, we identified p300/CBP-associated factor (PCAF) as a novel interaction partner of ATF4 in leucine-starved cells. We show that the N-terminal region of ATF4 is required for a direct interaction with PCAF and demonstrate that PCAF is involved in the full transcriptional response of CHOP by amino acid starvation. Chromatin immunoprecipitation analysis revealed that PCAF is engaged on the CHOP AARE in response to amino acid starvation and that ATF4 is essential for its recruitment. We also show that PCAF stimulates ATF4-driven transcription via its histone acetyltransferase domain. Thus PCAF acts as a coactivator of ATF4 and is involved in the enhancement of CHOP transcription following amino acid starvation.


Subject(s)
Activating Transcription Factor 4/metabolism , Amino Acids/physiology , Transcription Factor CHOP/genetics , Transcriptional Activation , p300-CBP Transcription Factors/metabolism , Activating Transcription Factor 4/chemistry , Activating Transcription Factor 4/physiology , Animals , Binding Sites , Cell Line , Cells, Cultured , HeLa Cells , Humans , Leucine/physiology , Mice , Protein Structure, Tertiary , Response Elements , p300-CBP Transcription Factors/chemistry , p300-CBP Transcription Factors/genetics
18.
J Biol Chem ; 280(50): 41537-45, 2005 Dec 16.
Article in English | MEDLINE | ID: mdl-16219772

ABSTRACT

ATF4 plays a crucial role in the cellular response to stress and multiple stress responses pathways converge to the translational up-regulation of ATF4. ATF4 is a substrate of the SCF(betaTrCP) ubiquitin ligase that binds to betaTrCP through phosphorylation on a DSGXXXS motif. We show here that ATF4 stability is also modulated by the histone acetyltransferase p300, which induces ATF4 stabilization by inhibiting its ubiquitination. Despite p300 acetylates ATF4, we found that p300-mediated ATF4 stabilization is independent of p300 catalytic activity, using either the inactive form of p300 or the acetylation mutant ATF4-K311R. ATF4 deleted of its p300 binding domain is no more stabilized by p300 nor recruited into nuclear speckles. In consequence of ATF4 stabilization, both p300 and the catalytically inactive enzyme increase ATF4 transcriptional activity.


Subject(s)
Activating Transcription Factor 4/chemistry , CREB-Binding Protein/physiology , Transcription, Genetic , p300-CBP Transcription Factors/chemistry , Amino Acid Motifs , Blotting, Western , CREB-Binding Protein/chemistry , Catalysis , Cell Line , Cell Nucleus/metabolism , Electrophoresis, Polyacrylamide Gel , Gene Silencing , Genetic Vectors , Glutathione Transferase/metabolism , HeLa Cells , Histone Acetyltransferases/chemistry , Humans , Immunoprecipitation , Microscopy, Fluorescence , Mutation , Phosphorylation , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Transfection , Ubiquitin/chemistry , Ubiquitin/metabolism , p300-CBP Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...