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











Publication year range
1.
Int J Biol Macromol ; 120(Pt A): 310-317, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30144543

ABSTRACT

Activating transcription factor 3 (ATF3) is a stress-responsive factor that belongs to the activator protein 1 (AP-1) family of transcription factors. ATF3 expression is stimulated by various factors such as hypoxia, cytokines, and chemotherapeutic and DNA damaging agents. Upon stimulation, ATF3 can form homodimers or heterodimers with other members of the AP-1 family to repress or activate transcription. Under physiological conditions, ATF3 expression is transient and plays a pivotal role in controlling the expression of cell-cycle regulators and tumor suppressor, DNA repair, and apoptosis genes. However, under pathological conditions such as those during breast cancer, a sustained and prolonged expression of ATF3 has been observed. In this review, the structure and function of ATF3, its posttranslational modifications (PTM), and its interacting proteins are discussed with a special emphasis on breast cancer metastasis.


Subject(s)
Activating Transcription Factor 3/metabolism , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/metabolism , Protein Multimerization , Protein Processing, Post-Translational , Activating Transcription Factor 3/chemistry , Activating Transcription Factor 3/genetics , Animals , Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle , DNA Repair , Female , Humans , Male , Neoplasm Metastasis , Neoplasm Proteins/genetics , Structure-Activity Relationship
2.
Diabetes ; 67(6): 1093-1104, 2018 06.
Article in English | MEDLINE | ID: mdl-29535089

ABSTRACT

Leptin acts via its receptor (LepRb) to modulate gene expression in hypothalamic LepRb-expressing neurons, thereby controlling energy balance and glucose homeostasis. Despite the importance of the control of gene expression in hypothalamic LepRb neurons for leptin action, the transcriptional targets of LepRb signaling have remained undefined because LepRb cells contribute a small fraction to the aggregate transcriptome of the brain regions in which they reside. We thus employed translating ribosome affinity purification followed by RNA sequencing to isolate and analyze mRNA from the hypothalamic LepRb neurons of wild-type or leptin-deficient (Lepob/ob) mice treated with vehicle or exogenous leptin. Although the expression of most of the genes encoding the neuropeptides commonly considered to represent the main targets of leptin action were altered only following chronic leptin deprivation, our analysis revealed other transcripts that were coordinately regulated by leptin under multiple treatment conditions. Among these, acute leptin treatment increased expression of the transcription factor Atf3 in LepRb neurons. Furthermore, ablation of Atf3 from LepRb neurons (Atf3LepRbKO mice) decreased leptin efficacy and promoted positive energy balance in mice. Thus, this analysis revealed the gene targets of leptin action, including Atf3, which represents a cellular mediator of leptin action.


Subject(s)
Activating Transcription Factor 3/agonists , Gene Expression Regulation , Hypothalamus/metabolism , Leptin/metabolism , Neurons/metabolism , Receptors, Leptin/agonists , Signal Transduction , Activating Transcription Factor 3/chemistry , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Animals , Crosses, Genetic , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Energy Metabolism/drug effects , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Hypothalamus/cytology , Hypothalamus/drug effects , Hypothalamus/pathology , Leptin/analogs & derivatives , Leptin/pharmacology , Leptin/therapeutic use , Lipotropic Agents/pharmacology , Lipotropic Agents/therapeutic use , Male , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/pathology , Obesity/drug therapy , Obesity/metabolism , Obesity/pathology , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Sequence Analysis, RNA , Signal Transduction/drug effects
3.
J Biol Chem ; 292(37): 15266-15276, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28765281

ABSTRACT

Persistent DNA damage induces profound alterations in gene expression that, in turn, influence tissue homeostasis, tumorigenesis, and cancer treatment outcome. However, the underlying mechanism for gene expression reprogramming induced by persistent DNA damage remains poorly understood. Here, using a highly effective bioluminescence-based reporter system and other tools, we report that persistent DNA damage inhibits nonsense-mediated RNA decay (NMD), an RNA surveillance and gene-regulatory pathway, in noncycling cells. NMD suppression by persistent DNA damage required the activity of the p38α MAPK. Activating transcription factor 3 (ATF3), an NMD target and a key stress-inducible transcription factor, was stabilized in a p38α- and NMD-dependent manner following persistent DNA damage. Our results reveal a novel p38α-dependent pathway that regulates NMD activity in response to persistent DNA damage, which, in turn, controls ATF3 expression in affected cells.


Subject(s)
Activating Transcription Factor 3/metabolism , DNA Damage , Gene Expression Regulation , Mitogen-Activated Protein Kinase 14/metabolism , Nonsense Mediated mRNA Decay , RNA, Messenger/metabolism , Activating Transcription Factor 3/chemistry , Activating Transcription Factor 3/genetics , Biomarkers/metabolism , Bleomycin/toxicity , Cells, Cultured , Cellular Senescence , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Gamma Rays/adverse effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Genes, Reporter/drug effects , Genes, Reporter/radiation effects , HEK293 Cells , Humans , Luminescent Measurements , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/genetics , Mutagens/toxicity , Nonsense Mediated mRNA Decay/drug effects , Nonsense Mediated mRNA Decay/radiation effects , Oxidative Stress , Protein Stability/drug effects , Protein Stability/radiation effects , RNA Interference , RNA Stability/drug effects , RNA Stability/radiation effects , RNA, Messenger/chemistry
4.
J Biol Chem ; 292(31): 12885-12894, 2017 08 04.
Article in English | MEDLINE | ID: mdl-28607151

ABSTRACT

Satellite cells are skeletal muscle stem cells that provide myonuclei for postnatal muscle growth, maintenance, and repair/regeneration in adults. Normally, satellite cells are mitotically quiescent, but they are activated in response to muscle injury, in which case they proliferate extensively and exhibit up-regulated expression of the transcription factor MyoD, a master regulator of myogenesis. MyoD forms a heterodimer with E proteins through their basic helix-loop-helix domain, binds to E boxes in the genome and thereby activates transcription at muscle-specific promoters. The central role of MyoD in muscle differentiation has increased interest in finding potential MyoD regulators. Here we identified transducin-like enhancer of split (TLE3), one of the Groucho/TLE family members, as a regulator of MyoD function during myogenesis. TLE3 was expressed in activated and proliferative satellite cells in which increased TLE3 levels suppressed myogenic differentiation, and, conversely, reduced TLE3 levels promoted myogenesis with a concomitant increase in proliferation. We found that, via its glutamine- and serine/proline-rich domains, TLE3 interferes with MyoD function by disrupting the association between the basic helix-loop-helix domain of MyoD and E proteins. Our findings indicate that TLE3 participates in skeletal muscle homeostasis by dampening satellite cell differentiation via repression of MyoD transcriptional activity.


Subject(s)
Co-Repressor Proteins/metabolism , Gene Expression Regulation, Developmental , Muscle Development , Muscle Fibers, Skeletal/metabolism , MyoD Protein/antagonists & inhibitors , Myoblasts/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Activating Transcription Factor 3/chemistry , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Animals , Cell Proliferation , Cells, Cultured , Co-Repressor Proteins/antagonists & inhibitors , Co-Repressor Proteins/chemistry , Co-Repressor Proteins/genetics , Gene Deletion , Helix-Loop-Helix Motifs , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/cytology , MyoD Protein/chemistry , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts/cytology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Multimerization , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Satellite Cells, Skeletal Muscle/cytology
5.
BMC Genomics ; 17: 335, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27146783

ABSTRACT

BACKGROUND: Dysregulation of the common stress responsive transcription factor ATF3 has been causally linked to many important human diseases such as cancer, atherosclerosis, infections, and hypospadias. Although it is believed that the ATF3 transcription activity is central to its cellular functions, how ATF3 regulates gene expression remains largely unknown. Here, we employed ATF3 wild-type and knockout isogenic cell lines to carry out the first comprehensive analysis of global ATF3-binding profiles in the human genome under basal and stressed (DNA damage) conditions. RESULTS: Although expressed at a low basal level, ATF3 was found to bind a large number of genomic sites that are often associated with genes involved in cellular stress responses. Interestingly, ATF3 appears to bind a large portion of genomic sites distal to transcription start sites and enriched with p300 and H3K27ac. Global gene expression profiling analysis indicates that genes proximal to these genomic sites were often regulated by ATF3. While DNA damage elicited by camptothecin dramatically altered the ATF3 binding profile, most of the genes regulated by ATF3 upon DNA damage were pre-bound by ATF3 before the stress. Moreover, we demonstrated that ATF3 was co-localized with the major stress responder p53 at genomic sites, thereby collaborating with p53 to regulate p53 target gene expression upon DNA damage. CONCLUSIONS: These results suggest that ATF3 likely bookmarks genomic sites and interacts with other transcription regulators to control gene expression.


Subject(s)
Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , DNA/metabolism , Histones/metabolism , p300-CBP Transcription Factors/metabolism , Activating Transcription Factor 3/chemistry , Binding Sites/drug effects , Camptothecin/pharmacology , Cell Line, Tumor , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Knockout Techniques , HCT116 Cells , HEK293 Cells , Humans , Tumor Suppressor Protein p53/metabolism
6.
Transcription ; 5(4): e964559, 2014.
Article in English | MEDLINE | ID: mdl-25483406
7.
Int J Mol Sci ; 14(4): 8367-80, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23591848

ABSTRACT

SUMOylation plays an important role in regulating a wide range of cellular processes. Previously, we showed that ATF3, a stress response mediator, can be SUMOylated and lysine 42 is the major SUMO site. However, the significance of ATF3 SUMOylation in biological processes is still poorly understood. In the present study, we investigated the role of ATF3 SUMOylation on CCND activity and cellular proliferation in human prostate cancer cells. First, we showed that ATF3 can be SUMOylated endogenously in the overexpression system, and lysine 42 is the major SUMO site. Unlike normal prostate tissue and androgen-responsive LNCaP cancer cells, androgen-independent PC3 and DU145 cancer cells did not express ATF3 endogenously. Overexpression of ATF3 increased CCND1/2 expression in PC3 and DU145 cancer cells. Interestingly, we observed that SUMOylation is essential for ATF3-mediated CCND1/2 activation. Finally, we observed that SUMOylation plays a functional role in ATF3-mediated cellular proliferation in PC3 and DU145 cells. Taken together, our results demonstrate that SUMO modification of ATF3 influences CCND1/2 activity and cellular proliferation of prostate cancer PC3 and DU145 cells and explains at least in part how ATF3 functions to regulate cancer development.


Subject(s)
Activating Transcription Factor 3/metabolism , Cyclin D1/metabolism , Cyclin D2/metabolism , Prostatic Neoplasms/metabolism , Activating Transcription Factor 3/chemistry , Activating Transcription Factor 3/genetics , Amino Acid Substitution , Androgens/metabolism , Cell Line, Tumor , Cell Proliferation , Cyclin D1/genetics , Cyclin D2/genetics , Humans , Lysine/chemistry , Male , Mutagenesis, Site-Directed , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Prostate/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sumoylation , Transfection
8.
Anal Biochem ; 408(2): 321-7, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-20868646

ABSTRACT

The utility of antibody reagents for the detection of specific cellular targets for both research and diagnostic applications is widespread and continually expanding. Often it is useful to develop specific antibodies as reagent pairs that distinguish different epitopes of the target such that sandwich enzyme-linked immunosorbent assay can be used for selective and specific detection. However, the identification of pairing antibodies is often cumbersome and labor-intensive even with the use of designed peptide-specific epitopes as antigens. We have developed a robust and high-throughput method for identifying pairing complementary antibodies derived either from commercial sources or during a rabbit hybridoma monoclonal screening and selection process using protein A capture with the AlphaScreen bead-based assay format. We demonstrate the value and effectiveness of this assay with three protein targets: Akt2, ATF3, and NAEß (the ß-subunit of the neddylation activation enzyme).


Subject(s)
Antibodies/chemistry , Immunoassay/methods , Staphylococcal Protein A/chemistry , Activating Transcription Factor 3/chemistry , Activating Transcription Factor 3/immunology , Animals , Antibodies/immunology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Epitopes/immunology , Hybridomas/metabolism , Peptides/immunology , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/immunology , Rabbits
9.
Gene Expr ; 15(1): 1-11, 2010.
Article in English | MEDLINE | ID: mdl-21061913

ABSTRACT

Activating transcription factor 3 (ATF3) gene encodes a member of the ATF family of transcription factors and is induced by various stress signals. All members of this family share the basic region-leucine zipper (bZip) DNA binding motif and bind to the consensus sequence TGACGTCA in vitro. Previous reviews and an Internet source have covered the following topics: the nomenclature of ATF proteins, the history of their discovery, the potential interplays between ATFs and other bZip proteins, ATF3-interacting proteins, ATF3 target genes, and the emerging roles of ATF3 in cancer and immunity (see footnote 1). In this review, we present evidence and clues that prompted us to put forth the idea that ATF3 functions as a "hub" of the cellular adaptive-response network. We will then focus on the roles of ATF3 in modulating inflammatory response. Inflammation is increasingly recognized to play an important role for the development of many diseases. Putting this in the context of the hub idea, we propose that modulation of inflammation by ATF3 is a unifying theme for the potential involvement of ATF3 in various diseases.


Subject(s)
Activating Transcription Factor 3/metabolism , Adaptation, Physiological , Inflammation/etiology , Inflammation/metabolism , Signal Transduction , Activating Transcription Factor 3/chemistry , Activating Transcription Factor 3/genetics , Amino Acid Sequence , Animals , Humans , Inflammation/genetics , Molecular Sequence Data , Protein Processing, Post-Translational
10.
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
11.
Eur J Endocrinol ; 158(5): 729-39, 2008 May.
Article in English | MEDLINE | ID: mdl-18426833

ABSTRACT

INTRODUCTION: Hypospadias is a common inborn error of the genital development, whose complex etiology remains elusive. Defects of the androgen metabolism and activity have been found in a subset of boys with hypospadias. Moreover, the balance between androgens and estrogens seems to be important to the proper male genital development. Activating transcription factor 3 (ATF3), an estrogen responsive gene, has been reported to be expressed during sexual development and up-regulated in hypospadic genital skin. We investigated ATF3 as a candidate gene for hypospadias. MATERIAL AND METHODS: Genotyping of eight-tagged single nucleotide polymorphisms (SNP)s was performed in 330 boys with hypospadias and in 380 healthy controls. Screening for mutations in ATF3 was conducted in a subset of boys with hypospadias. ATF3 expression was evaluated in the foreskin of boys with hypospadias and in healthy controls and in the human fetal genitalia by immunohistochemistry. RESULTS: Three common SNPs, spanning a region of about 16 kb in intron 1 of ATF3, are associated with hypospadias. These SNPs are not linked and their effects are independent. The combination of the three risk SNPs yields the highest significance. Mutation screening identified the gene variant c536A>G in one patient and c817C>T in the 3'-UTR in two other patients. ATF3 expression was evidenced in the developing male urethra. CONCLUSIONS: ATF3 gene variants influence the risk of hypospadias. Its hormonal responsiveness may underlie this risk effect. But also other ATF3-dependent biological aspects, such as cell survival and death, response to stress stimuli, or the control of epithelial-mesenchymal interactions, may be of importance.


Subject(s)
Activating Transcription Factor 3/genetics , Genitalia, Male/abnormalities , Genitalia, Male/physiology , Hypospadias/etiology , Hypospadias/genetics , Activating Transcription Factor 3/chemistry , Activating Transcription Factor 3/metabolism , Base Sequence , Child , DNA Mutational Analysis , Female , Fetus , Genetic Predisposition to Disease/epidemiology , Genitalia, Female/physiology , Haplotypes , Humans , Hypospadias/epidemiology , Immunohistochemistry , Introns , Logistic Models , Male , Molecular Sequence Data , Polymorphism, Single Nucleotide , Protein Structure, Quaternary , Risk Factors
12.
J Biol Chem ; 281(15): 10473-81, 2006 Apr 14.
Article in English | MEDLINE | ID: mdl-16469745

ABSTRACT

ATF3 is a stress-inducible gene that encodes a member of the ATF/CREB family of transcription factors. Current literature indicates that ATF3 affects cell death and cell cycle progression. However, controversies exist, because it has been demonstrated to be a negative or positive regulator of these processes. We sought to study the roles of ATF3 in both cell death and cell cycle regulation in the same cell type using mouse fibroblasts. We show that ATF3 promotes apoptosis and cell cycle arrest. Fibroblasts deficient in ATF3 (ATF3(-/-)) were partially protected from UV-induced apoptosis, and fibroblasts ectopically expressing ATF3(-/-) under the tet-off system exhibited features characteristic of apoptosis upon ATF3 induction. Furthermore, ATF3(-/-) fibroblasts transitioned from G(2) to S phase more efficiently than the ATF3(+/+) fibroblasts, suggesting a growth arrest role of ATF3. Consistent with the growth arrest and pro-apoptotic roles of ATF3, ATF3(-) fibroblasts upon Ras transformation exhibited higher growth rate, produced more colonies in soft agar, and formed larger tumor upon xenograft injection than the ATF3(+/+) counterparts. ATF3(-/-) cells, either with or without Ras transformation, had increased Rb phosphorylation and higher levels of various cyclins. Significantly, ATF3 bound to the cyclin D1 promoter as shown by chromatin immunoprecipitation (ChIP) assay and repressed its transcription by a transcription assay. Taken together, our results indicate that ATF3 promotes cell death and cell arrest, and suppresses Ras-mediated tumorigenesis. Potential explanations for the controversy about the roles of ATF3 in cell cycle and cell death are discussed.


Subject(s)
Activating Transcription Factor 3/physiology , Neoplasms/metabolism , ras Proteins/metabolism , Activating Transcription Factor 3/chemistry , Animals , Apoptosis , Bromodeoxyuridine/pharmacology , Cell Cycle , Cell Death , Cell Proliferation , Cell Survival , Cell Transformation, Neoplastic , Chromatin Immunoprecipitation , Cyclin D1/metabolism , Dose-Response Relationship, Drug , Fibroblasts/metabolism , G1 Phase , Gene Expression Regulation, Neoplastic , Humans , Immunoblotting , Immunohistochemistry , MAP Kinase Kinase 4/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Phosphorylation , Plasmids/metabolism , Retinoblastoma Protein/metabolism , Retroviridae/genetics , S Phase , Time Factors , Transcription, Genetic , Ultraviolet Rays
13.
Acta Biochim Biophys Sin (Shanghai) ; 38(1): 58-62, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16395528

ABSTRACT

Activating transcription factor 3 (ATF3), a member of the activating transcription factor/cAMP responsive element binding protein (ATF/CREB) family of transcription factors, is induced by many physiological stresses. To investigate the activity of ATF/CREB in cells with physiological stresses, we developed a practical reporter vector, the plasmid pATF/CRE-luc, bearing activating transcription factor/cAMP responsive element (ATF/CRE) binding sites. This plasmid was constructed by inserting three repeats of the ATF/CRE binding element into the plasmid pG5luc, replacing the GAL-4 binding sites. The plasmids pACT/ATF3 and pATF/CRE-luc were transfected into HeLa and NIH3T3 cells, respectively, and the results showed that the expression of luciferase was increased in a dose-dependent manner on plasmid pACT/ATF3. The data suggested that the plasmid pATF/CRE-luc could be used as a sensitive and convenient reporter system of ATF3 activity.


Subject(s)
Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Protein Engineering/methods , Transfection/methods , Activating Transcription Factor 3/chemistry , Animals , Genes, Reporter/genetics , Genetic Vectors/genetics , HeLa Cells , Humans , Mice , NIH 3T3 Cells , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL