Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Biochem Soc Trans ; 51(5): 1749-1763, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37721138

ABSTRACT

Genes encoding histone proteins are recurrently mutated in tumor samples, and these mutations may impact nucleosome stability, histone post-translational modification, or chromatin dynamics. The prevalence of histone mutations across diverse cancer types suggest that normal chromatin structure is a barrier to tumorigenesis. Oncohistone mutations disrupt chromatin structure and gene regulatory mechanisms, resulting in aberrant gene expression and the development of cancer phenotypes. Examples of oncohistones include the histone H3 K27M mutation found in pediatric brain cancers that blocks post-translational modification of the H3 N-terminal tail and the histone H2B E76K mutation found in some solid tumors that disrupts nucleosome stability. Oncohistones may comprise a limited fraction of the total histone pool yet cause global effects on chromatin structure and drive cancer phenotypes. Here, we survey histone mutations in cancer and review their function and role in tumorigenesis.


Subject(s)
Histones , Neoplasms , Humans , Child , Histones/metabolism , Nucleosomes/genetics , Mutation , Neoplasms/genetics , Neoplasms/pathology , Chromatin , Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics
2.
PLoS Genet ; 17(12): e1009934, 2021 12.
Article in English | MEDLINE | ID: mdl-34914716

ABSTRACT

MicroRNAs (miRNA) are short non-coding RNAs widely implicated in gene regulation. Most metazoan miRNAs utilize the RNase III enzymes Drosha and Dicer for biogenesis. One notable exception is the RNA polymerase II transcription start sites (TSS) miRNAs whose biogenesis does not require Drosha. The functional importance of the TSS-miRNA biogenesis is uncertain. To better understand the function of TSS-miRNAs, we applied a modified Crosslinking, Ligation, and Sequencing of Hybrids on Argonaute (AGO-qCLASH) to identify the targets for TSS-miRNAs in HCT116 colorectal cancer cells with or without DROSHA knockout. We observed that miR-320a hybrids dominate in TSS-miRNA hybrids identified by AGO-qCLASH. Targets for miR-320a are enriched for the eIF2 signaling pathway, a downstream component of the unfolded protein response. Consistently, in miR-320a mimic- and antagomir- transfected cells, differentially expressed gene products are associated with eIF2 signaling. Within the AGO-qCLASH data, we identified the endoplasmic reticulum (ER) chaperone calnexin as a direct miR-320a down-regulated target, thus connecting miR-320a to the unfolded protein response. During ER stress, but not amino acid deprivation, miR-320a up-regulates ATF4, a critical transcription factor for resolving ER stress. In summary, our study investigates the targetome of the TSS-miRNAs in colorectal cancer cells and establishes miR-320a as a regulator of unfolded protein response.


Subject(s)
Activating Transcription Factor 4/genetics , Colorectal Neoplasms/genetics , MicroRNAs/genetics , Ribonuclease III/genetics , Antagomirs/genetics , Argonaute Proteins/genetics , Calnexin/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/pathology , DEAD-box RNA Helicases/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum Stress/genetics , Eukaryotic Initiation Factor-2/genetics , Gene Knockout Techniques , HCT116 Cells , Humans , Signal Transduction/genetics , Transcription Initiation Site
3.
Cancers (Basel) ; 12(11)2020 Nov 05.
Article in English | MEDLINE | ID: mdl-33167336

ABSTRACT

Multiple myeloma (MM) cells consume huge amounts of glutamine and, as a consequence, the amino acid concentration is lower-than-normal in the bone marrow (BM) of MM patients. Here we show that MM-dependent glutamine depletion induces glutamine synthetase in stromal cells, as demonstrated in BM biopsies of MM patients, and reproduced in vitro by co-culturing human mesenchymal stromal cells (MSCs) with MM cells. Moreover, glutamine depletion hinders osteoblast differentiation of MSCs, which is also severely blunted by the spent, low-glutamine medium of MM cells, and rescued by glutamine restitution. Glutaminase and the concentrative glutamine transporter SNAT2 are induced during osteoblastogenesis in vivo and in vitro, and both needed for MSCs differentiation, pointing to enhanced the requirement for the amino acid. Osteoblastogenesis also triggers the induction of glutamine-dependent asparagine synthetase (ASNS), and, among non-essential amino acids, asparagine rescues differentiation of glutamine-starved MSCs, by restoring the transcriptional profiles of differentiating MSCs altered by glutamine starvation. Thus, reduced asparagine availability provides a mechanistic link between MM-dependent Gln depletion in BM and impairment of osteoblast differentiation. Inhibition of Gln metabolism in MM cells and supplementation of asparagine to stromal cells may, therefore, constitute novel approaches to prevent osteolytic lesions in MM.

4.
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
5.
Biochim Biophys Acta Gene Regul Mech ; 1861(2): 72-79, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29413899

ABSTRACT

Activating transcription factor 3 (ATF3) is a highly regulated protein that is implicated in a wide range of pathological conditions including inflammation and transformation. Transcription from the ATF3 gene is induced by several stress-induced signaling pathways, including amino acid limitation (amino acid response, AAR) and ER stress (unfolded protein response, UPR). Induction of ATF3 transcription by these pathways is mediated by ATF4 and cJUN recruitment to enhancer elements within the ATF3 gene. Although a canonical promoter (promoter A) has been studied by numerous laboratories, a second promoter activity (promoter A1), 43 kb upstream of the first, has been reported to respond to stress-induced signaling and to be critical for ATF3 expression in certain transformed cells. The results of the present study show that in normal human hepatocytes and HepG2 human hepatoma cells both basal as well as AAR- and UPR-induced transcription occurs almost exclusively from promoter A. This selectivity between the two promoters correlated with increased binding of ATF4, recruitment of RNA polymerase II, and the expected histone modifications in the promoter A region of the gene. Time course studies of ATF3 transcription activity revealed that the temporal kinetics for ATF3 induction differ between the AAR and UPR, with the former being more transient than the latter. Collectively, the results document that ATF3 expression in normal and transformed human liver originates from the canonical promoter A that responds to multiple stress signals.


Subject(s)
Activating Transcription Factor 3/genetics , Amino Acids/metabolism , Endoplasmic Reticulum Stress/genetics , Hepatocytes/metabolism , Promoter Regions, Genetic/genetics , Activating Transcription Factor 3/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cells, Cultured , Hep G2 Cells , Humans , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Response Elements/genetics , Transcription, Genetic , Unfolded Protein Response/genetics
6.
Adv Nutr ; 7(4): 780S-9S, 2016 07.
Article in English | MEDLINE | ID: mdl-27422515

ABSTRACT

Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have promise in regenerative medicine because of their ability to differentiate into all 3 primary germ layers. This review describes recent advances in the understanding of the link between the metabolism of ESCs/iPSCs and their maintenance/differentiation in the cell culture setting, with particular emphasis on amino acid (AA) metabolism. ESCs are endowed with unique metabolic features with regard to energy consumption, metabolite flux through particular pathways, and macromolecular synthesis. Therefore, nutrient availability has a strong influence on stem cell growth, self-renewal, and lineage specification, both in vivo and in vitro. Evidence from several laboratories has documented that self-renewal and differentiation of mouse ESCs are critically dependent on proline metabolism, with downstream metabolites possibly serving as signal molecules. Likewise, catabolism of either threonine (mouse) or methionine (human) is required for growth and differentiation of ESCs because these AAs serve as precursors for donor molecules used in histone methylation and acetylation. Epigenetic mechanisms are recognized as critical steps in differentiation, and AA metabolism in ESCs appears to modulate these epigenetic processes. Recent reports also document that, in vitro, the nutrient composition of the culture medium in which ESCs are differentiated into embryoid bodies can influence lineage specification, leading to enrichment of a specific cell type. Although research designed to direct tissue specification of differentiating embryoid bodies in culture is still in its infancy, early results indicate that manipulation of the nutrient milieu can promote or suppress the formation of specific cell lineages.


Subject(s)
Amino Acids/metabolism , Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Acetylation , Animals , Cells, Cultured , Embryonic Stem Cells/cytology , Energy Metabolism , Epigenesis, Genetic , Humans , Methionine/metabolism , Methylation , Mice , Proline/metabolism , Regenerative Medicine/methods , Threonine/metabolism
7.
Nucleic Acids Res ; 44(20): 9719-9732, 2016 Nov 16.
Article in English | MEDLINE | ID: mdl-27471030

ABSTRACT

The response to amino acid (AA) limitation of the entire aminoacyl-tRNA synthetase (ARS) gene family revealed that 16/20 of the genes encoding cytoplasmic-localized enzymes are transcriptionally induced by activating transcription factor 4 (Atf4) via C/ebp-Atf-Response-Element (CARE) enhancers. In contrast, only 4/19 of the genes encoding mitochondrial-localized ARSs were weakly induced. Most of the activated genes have a functional CARE near the transcription start site (TSS), but for others the CARE is downstream. Regardless of the location of CARE enhancer, for all ARS genes there was constitutive association of RNA polymerase II (Pol II) and the general transcription machinery near the TSS. However, for those genes with a downstream CARE, Atf4, C/ebp-homology protein (Chop), Pol II and TATA-binding protein exhibited enhanced recruitment to the CARE during AA limitation. Increased Atf4 binding regulated the association of elongation factors at both the promoter and the enhancer regions, and inhibition of cyclin-dependent kinase 9 (CDK9), that regulates these elongation factors, blocked induction of the AA-responsive ARS genes. Protein pull-down assays indicated that Atf4 directly interacts with CDK9 and its associated protein cyclin T1. The results demonstrate that AA availability modulates the ARS gene family through modulation of transcription elongation.


Subject(s)
Activating Transcription Factor 4/metabolism , Amino Acids/metabolism , Amino Acyl-tRNA Synthetases/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Response Elements , Transcription, Genetic , Cell Line , Chromatin Immunoprecipitation , Enhancer Elements, Genetic , Gene Knockout Techniques , High-Throughput Nucleotide Sequencing , Humans , Promoter Regions, Genetic , RNA, Messenger/chemistry , RNA, Messenger/genetics , Transcription Elongation, Genetic , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Transcription Initiation Site
8.
Oncotarget ; 7(3): 3128-43, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26657730

ABSTRACT

Cancer cells are frequently exposed to physiological stress conditions such as hypoxia and nutrient limitation. Escape from stress-induced apoptosis is one of the mechanisms used by malignant cells to survive unfavorable conditions. B-cell Translocation Gene 1 (BTG1) is a tumor suppressor that is frequently deleted in acute lymphoblastic leukemia and recurrently mutated in diffuse large B cell lymphoma. Moreover, low BTG1 expression levels have been linked to poor outcome in several solid tumors. How loss of BTG1 function contributes to tumor progression is not well understood. Here, using Btg1 knockout mice, we demonstrate that loss of Btg1 provides a survival advantage to primary mouse embryonic fibroblasts (MEFs) under stress conditions. This pro-survival effect involves regulation of Activating Transcription Factor 4 (ATF4), a key mediator of cellular stress responses. We show that BTG1 interacts with ATF4 and positively modulates its activity by recruiting the protein arginine methyl transferase PRMT1 to methylate ATF4 on arginine residue 239. We further extend these findings to B-cell progenitors, by showing that loss of Btg1 expression enhances stress adaptation of mouse bone marrow-derived B cell progenitors. In conclusion, we have identified the BTG1/PRMT1 complex as a new modifier of ATF4 mediated stress responses.


Subject(s)
Activating Transcription Factor 4/metabolism , Neoplasm Proteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Stress, Physiological/physiology , Animals , Apoptosis/physiology , B-Lymphocytes/cytology , Cell Line, Tumor , Fibroblasts , Humans , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
9.
J Biol Chem ; 290(25): 15878-15891, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25931127

ABSTRACT

Using an unbiased systems genetics approach, we previously predicted a role for CHAC1 in the endoplasmic reticulum stress pathway, linked functionally to activating transcription factor 4 (ATF4) following treatment with oxidized phospholipids, a model for atherosclerosis. Mouse and yeast CHAC1 homologs have been shown to degrade glutathione in yeast and a cell-free system. In this report, we further defined the ATF4-CHAC1 interaction by cloning the human CHAC1 promoter upstream of a luciferase reporter system for in vitro assays in HEK293 and U2OS cells. Mutation and deletion analyses defined two major cis DNA elements necessary and sufficient for CHAC1 promoter-driven luciferase transcription under conditions of ER stress or ATF4 coexpression: the -267 ATF/cAMP response element (CRE) site and a novel -248 ATF/CRE modifier (ACM) element. We also examined the ability of the CHAC1 ATF/CRE and ACM sequences to bind ATF4 and ATF3 using immunoblot-EMSA and confirmed ATF4, ATF3, and CCAAT/enhancer-binding protein ß binding at the human CHAC1 promoter in the proximity of the ATF/CRE and ACM using ChIP. To further validate the function of CHAC1 in a human cell model, we measured glutathione levels in HEK293 cells with enhanced CHAC1 expression. Overexpression of CHAC1 led to a robust depletion of glutathione, which was alleviated in a CHAC1 catalytic mutant. These results suggest an important role for CHAC1 in oxidative stress and apoptosis with implications for human health and disease.


Subject(s)
Activating Transcription Factor 3/metabolism , Activating Transcription Factor 4/metabolism , Gene Expression Regulation, Enzymologic/physiology , Glutathione/metabolism , RNA, Messenger/biosynthesis , Response Elements/physiology , gamma-Glutamylcyclotransferase/biosynthesis , Activating Transcription Factor 3/genetics , Activating Transcription Factor 4/genetics , Animals , Base Sequence , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Endoplasmic Reticulum Stress/physiology , Glutathione/genetics , HEK293 Cells , Humans , Mice , Oxidative Stress/physiology , RNA, Messenger/genetics , Sequence Deletion , gamma-Glutamylcyclotransferase/genetics
10.
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
11.
J Biol Chem ; 289(35): 24665-79, 2014 Aug 29.
Article in English | MEDLINE | ID: mdl-25028509

ABSTRACT

Amino acid (AA) limitation in mammalian cells triggers a collection of signaling cascades jointly referred to as the AA response (AAR). In human HepG2 hepatocellular carcinoma, the early growth response 1 (EGR1) gene was induced by either AA deprivation or endoplasmic reticulum stress. AAR-dependent EGR1 activation was discovered to be independent of the well characterized GCN2-ATF4 pathway and instead dependent on MEK-ERK signaling, one of the MAPK pathways. ChIP showed that constitutively bound ELK1 at the EGR1 proximal promoter region was phosphorylated after AAR activation. Increased p-ELK1 binding was associated with increased de novo recruitment of RNA polymerase II to the EGR1 promoter. EGR1 transcription was not induced in HEK293T cells lacking endogenous MEK activity, but overexpression of exogenous constitutively active MEK in HEK293T cells resulted in increased basal and AAR-induced EGR1 expression. ChIP analysis of the human vascular endothelial growth factor A (VEGF-A) gene, a known EGR1-responsive gene, revealed moderate increases in AAR-induced EGR1 binding within the proximal promoter and highly inducible binding to a site within the first intron. Collectively, these data document a novel AA-activated MEK-ERK-ELK1 signaling mechanism.


Subject(s)
Amino Acids/metabolism , Early Growth Response Protein 1/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Transcription, Genetic , Base Sequence , DNA Primers , HEK293 Cells , Humans , Real-Time Polymerase Chain Reaction
12.
Am J Physiol Endocrinol Metab ; 305(3): E325-35, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23736538

ABSTRACT

In somatic cells, a collection of signaling pathways activated by amino acid limitation have been identified and referred to as the amino acid response (AAR). Despite the importance of possible detrimental effects of nutrient limitation during in vitro culture, the AAR has not been investigated in embryonic stem cells (ESC). AAR activation caused the expected increase in transcription factors that mediate specific AAR pathways, as well as the induction of asparagine synthetase, a terminal AAR target gene. Neither AAR activation nor stable knockdown of activating transcription factor (Atf) 4, a transcriptional mediator of the AAR, adversely affected ESC self-renewal or pluripotency. Low-level induction of the AAR over a 12-day period of embryoid body differentiation did alter lineage specification such that the primitive endodermal, visceral endodermal, and endodermal lineages were favored, whereas mesodermal and certain ectodermal lineages were suppressed. Knockdown of Atf4 further enhanced the AAR-induced increase in endodermal formation, suggesting that this phenomenon is mediated by an Atf4-independent mechanism. Collectively, the results indicate that, during differentiation of mouse embryoid bodies in culture, the availability of nutrients, such as amino acids, can influence the formation of specific cell lineages.


Subject(s)
Amino Acids/metabolism , Cell Differentiation/physiology , Cell Lineage/physiology , Embryonic Stem Cells/metabolism , Activating Transcription Factor 4/biosynthesis , Activating Transcription Factor 4/genetics , Animals , Aspartate-Ammonia Ligase/metabolism , Blotting, Western , Cell Count , Cells, Cultured , Flow Cytometry , Mice , Protein Biosynthesis , RNA/biosynthesis , RNA/isolation & purification , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Transcription, Genetic
13.
Mol Biol Cell ; 24(15): 2477-90, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23761072

ABSTRACT

Environmental stresses that disrupt protein homeostasis induce phosphorylation of eIF2, triggering repression of global protein synthesis coincident with preferential translation of ATF4, a transcriptional activator of the integrated stress response (ISR). Depending on the extent of protein disruption, ATF4 may not be able to restore proteostatic control and instead switches to a terminal outcome that features elevated expression of the transcription factor CHOP (GADD153/DDIT3). The focus of this study is to define the mechanisms by which CHOP directs gene regulatory networks that determine cell fate. We find that in response to proteasome inhibition, CHOP enhances the expression of a collection of genes encoding transcription regulators, including ATF5, which is preferentially translated during eIF2 phosphorylation. Transcriptional expression of ATF5 is directly induced by both CHOP and ATF4. Knockdown of ATF5 increases cell survival in response to proteasome inhibition, supporting the idea that both ATF5 and CHOP have proapoptotic functions. Transcriptome analysis of ATF5-dependent genes reveals targets involved in apoptosis, including NOXA, which is important for inducing cell death during proteasome inhibition. This study suggests that the ISR features a feedforward loop of stress-induced transcriptional regulators, each subject to transcriptional and translational control, which can switch cell fate toward apoptosis.


Subject(s)
Activating Transcription Factors/metabolism , Apoptosis , Homeostasis , Transcription Factor CHOP/physiology , Activating Transcription Factors/genetics , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Survival , Cells, Cultured , Eukaryotic Initiation Factor-2/metabolism , Feedback, Physiological , Gene Expression Regulation , Gene Knockout Techniques , Gene Regulatory Networks , Leupeptins/pharmacology , Mice , Phosphorylation , Promoter Regions, Genetic , Proteasome Inhibitors/pharmacology , Protein Biosynthesis , Protein Processing, Post-Translational , Proteolysis , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Response Elements , Stress, Physiological , Thapsigargin/pharmacology , Transcriptional Activation , Transcriptome
14.
Nat Cell Biol ; 15(5): 481-90, 2013 May.
Article in English | MEDLINE | ID: mdl-23624402

ABSTRACT

Protein misfolding in the endoplasmic reticulum (ER) leads to cell death through PERK-mediated phosphorylation of eIF2α, although the mechanism is not understood. ChIP-seq and mRNA-seq of activating transcription factor 4 (ATF4) and C/EBP homologous protein (CHOP), key transcription factors downstream of p-eIF2α, demonstrated that they interact to directly induce genes encoding protein synthesis and the unfolded protein response, but not apoptosis. Forced expression of ATF4 and CHOP increased protein synthesis and caused ATP depletion, oxidative stress and cell death. The increased protein synthesis and oxidative stress were necessary signals for cell death. We show that eIF2α-phosphorylation-attenuated protein synthesis, and not Atf4 mRNA translation, promotes cell survival. These results show that transcriptional induction through ATF4 and CHOP increases protein synthesis leading to oxidative stress and cell death. The findings suggest that limiting protein synthesis will be therapeutic for diseases caused by protein misfolding in the ER.


Subject(s)
Activating Transcription Factor 4/metabolism , Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-2/metabolism , Protein Biosynthesis , Transcription Factor CHOP/metabolism , Transcription, Genetic , Activating Transcription Factor 4/genetics , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Cell Death , Cell Survival , Chromatin Immunoprecipitation , Eukaryotic Initiation Factor-2/genetics , Gene Expression Regulation , Mice , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Protein Folding , Protein Interaction Mapping , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Transcription Factor CHOP/genetics , Unfolded Protein Response
15.
PLoS One ; 8(2): e52390, 2013.
Article in English | MEDLINE | ID: mdl-23390487

ABSTRACT

The Angelman/Prader-Willi syndrome (AS/PWS) domain contains at least 8 imprinted genes regulated by a bipartite imprinting center (IC) associated with the SNRPN gene. One component of the IC, the PWS-IC, governs the paternal epigenotype and expression of paternal genes. The mechanisms by which imprinting and expression of paternal genes within the AS/PWS domain - such as MKRN3 and NDN - are regulated by the PWS-IC are unclear. The syntenic region in the mouse is organized and imprinted similarly to the human domain with the murine PWS-IC defined by a 6 kb interval within the Snrpn locus that includes the promoter. To identify regulatory elements that may mediate PWS-IC function, we mapped the location and allele-specificity of DNase I hypersensitive (DH) sites within the PWS-IC in brain cells, then identified transcription factor binding sites within a subset of these DH sites. Six major paternal-specific DH sites were detected in the Snrpn gene, five of which map within the 6 kb PWS-IC. We postulate these five DH sites represent functional components of the murine PWS-IC. Analysis of transcription factor binding within multiple DH sites detected nuclear respiratory factors (NRF's) and YY1 specifically on the paternal allele. NRF's and YY1 were also detected in the paternal promoter region of the murine Mrkn3 and Ndn genes. These results suggest that NRF's and YY1 may facilitate PWS-IC function and coordinately regulate expression of paternal genes. The presence of NRF's also suggests a link between transcriptional regulation within the AS/PWS domain and regulation of respiration. 3C analyses indicated Mkrn3 lies in close proximity to the PWS-IC on the paternal chromosome, evidence that the PWS-IC functions by allele-specific interaction with its distal target genes. This could occur by allele-specific co-localization of the PWS-IC and its target genes to transcription factories containing NRF's and YY1.


Subject(s)
Angelman Syndrome/genetics , Gene Expression Regulation , Nuclear Respiratory Factors/genetics , Prader-Willi Syndrome/genetics , Regulatory Elements, Transcriptional , YY1 Transcription Factor/genetics , snRNP Core Proteins/genetics , Alleles , Angelman Syndrome/metabolism , Angelman Syndrome/pathology , Animals , Base Sequence , Binding Sites , Deoxyribonuclease I/metabolism , Genetic Loci , Genomic Imprinting , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Nuclear Respiratory Factors/metabolism , Prader-Willi Syndrome/metabolism , Prader-Willi Syndrome/pathology , Protein Binding , Synteny , Transcription, Genetic , YY1 Transcription Factor/metabolism , snRNP Core Proteins/metabolism
16.
Biochem J ; 449(1): 219-29, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-22978410

ABSTRACT

Amino acid deprivation of mammalian cells triggers several signalling pathways, the AAR (amino acid response), that results in transcriptional activation. For the ASNS (asparagine synthetase) and ATF3 (activating transcription factor 3) genes, increased transcription occurs in conjunction with recruitment of ATF4 to the gene. In HepG2 cells, analysis of the ASNS and ATF3 genes during AAR activation revealed increases in histone H3K4me3 (histone 3 trimethylated Lys4) and H4Ac (acetylated histone 4) levels, marks associated with active transcription, but a concurrent loss of total H3 protein near the promoter. The dynamic nature of AAR-regulated transcription was illustrated by a decline in ASNS transcription activity within minutes after removal of the AAR stress and a return to basal levels by 2 h. Reversal of ASNS transcription occurred in parallel with decreased promoter-associated H4Ac and ATF4 binding. However, the reduction in histone H3 and increase in H3K4me3 were not reversed. In yeast, persistence of H3K4me3 has been proposed to be a 'memory' mark of gene activity that alters the responsiveness of the gene, but the time course and magnitude of ASNS induction was unaffected when cells were challenged with a second round of AAR activation. The results of the present study document changes in gene-associated nucleosome abundance and histone modifications in response to amino-acid-dependent transcription.


Subject(s)
Activating Transcription Factor 3/genetics , Amino Acids/genetics , Aspartate-Ammonia Ligase/genetics , Histones/genetics , Transcriptional Activation/genetics , Activating Transcription Factor 3/metabolism , Amino Acids/chemistry , Amino Acids/metabolism , Aspartate-Ammonia Ligase/metabolism , Hep G2 Cells , Histones/chemistry , Histones/metabolism , Humans , Signal Transduction/physiology
17.
J Biol Chem ; 287(43): 36393-403, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-22955275

ABSTRACT

Following amino acid deprivation, the amino acid response (AAR) induces transcription from specific genes through a collection of signaling mechanisms, including the GCN2-eIF2-ATF4 pathway. The present report documents that the histone demethylase JMJD3 is an activating transcription factor 4 (ATF4)-dependent target gene. The JMJD3 gene contains two AAR-induced promoter activities and chromatin immunoprecipitation (ChIP) analysis showed that the AAR leads to enhanced ATF4 recruitment to the C/EBP-ATF response element (CARE) upstream of Promoter-1. AAR-induced histone modifications across the JMJD3 gene locus occur upon ATF4 binding. Jmjd3 transcription is not induced in Atf4-knock-out cells, but the AAR-dependent activation was rescued by inhibition of histone deacetylation with trichostatin A (TSA). The TSA rescue of AAR activation in the absence of Atf4 also occurred for the Atf3 and C/EBP homology protein (Chop) genes, but not for the asparagine synthetase gene. ChIP analysis of the Jmjd3, Atf3, and Chop genes in Atf4 knock-out cells documented that activation of the AAR in the presence of TSA led to specific changes in acetylation of histone H4. The results suggest that a primary function of ATF4 is to recruit histone acetyltransferase activity to a sub-set of AAR target genes. Thus, absolute binding of ATF4 to these particular genes is not required and no ATF4 interaction with the general transcription machinery is necessary. The data are consistent with the hypothesis that ATF4 functions as a pioneer factor to alter chromatin structure and thus, enhance transcription in a gene-specific manner.


Subject(s)
Activating Transcription Factor 4/metabolism , Histone Deacetylases/metabolism , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Response Elements , Acetylation/drug effects , Activating Transcription Factor 4/genetics , Animals , Chromatin/genetics , Chromatin/metabolism , HEK293 Cells , Hep G2 Cells , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Histones/genetics , Humans , Hydroxamic Acids/pharmacology , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Mice, Knockout , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
18.
Adv Nutr ; 3(3): 295-306, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22585903

ABSTRACT

Mammals exhibit multiple adaptive mechanisms that sense and respond to fluctuations in dietary nutrients. Consumption of reduced total dietary protein or a protein diet that is deficient in 1 or more of the essential amino acids triggers wide-ranging changes in feeding behavior and gene expression. At the level of individual cells, dietary protein deficiency is manifested as amino acid (AA) deprivation, which activates the AA response (AAR). The AAR is composed of a collection of signal transduction pathways that terminate in specific transcriptional programs designed to catalyze adaptation to the nutrient stress or, ultimately, undergo apoptosis. Independently of the AAR, endoplasmic reticulum stress activates 3 signaling pathways, collectively referred to as the unfolded protein response. The transcription factor activating transcription factor 4 is one of the terminal transcriptional mediators for both the AAR and the unfolded protein response, leading to a significant degree of overlap with regard to the target genes for these stress pathways. Over the past 5 y, research has revealed that the basic leucine zipper superfamily of transcription factors plays the central role in the AAR. Formation of both homo- and heterodimers among the activating transcription factor, CCAAT enhancer-binding protein, and FOS/JUN families of basic leucine zipper proteins forms the nucleus of a highly integrated transcription factor network that determines the initiation, magnitude, and duration of the cellular response to dietary protein or AA limitation.


Subject(s)
Activating Transcription Factors/metabolism , Amino Acids/metabolism , Dietary Proteins/administration & dosage , Activating Transcription Factors/genetics , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line , Cell Nucleus/genetics , Cell Nucleus/metabolism , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation , Humans , Signal Transduction
19.
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
20.
Physiol Genomics ; 41(3): 315-27, 2010 May.
Article in English | MEDLINE | ID: mdl-20215415

ABSTRACT

Dietary protein malnutrition is manifested as amino acid deprivation of individual cells, which activates an amino acid response (AAR) that alters cellular functions, in part, by regulating transcriptional and posttranscriptional mechanisms. The AAR was activated in HepG2 human hepatoma cells, and the changes in mRNA content were analyzed by microarray expression profiling. The results documented that 1,507 genes were differentially regulated by P < 0.001 and by more than twofold in response to the AAR, 250 downregulated and 1,257 upregulated. The spectrum of altered genes reveals that amino acid deprivation has far-reaching implications for gene expression and cellular function. Among those cellular functions with the largest numbers of altered genes were cell growth and proliferation, cell cycle, gene expression, cell death, and development. Potential biological relationships between the differentially expressed genes were analyzed by computer software that generates gene networks. Proteins that were central to the most significant of these networks included c-myc, polycomb group proteins, transforming growth factor ß1, nuclear factor (erythroid-derived 2)-like 2-related factor 2, FOS/JUN family members, and many members of the basic leucine zipper superfamily of transcription factors. Although most of these networks contained some genes that were known to be amino acid responsive, many new relationships were identified that underscored the broad impact that amino acid stress has on cellular function.


Subject(s)
Amino Acids/deficiency , Carcinoma, Hepatocellular/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Amino Acids/metabolism , Down-Regulation/genetics , Gene Regulatory Networks/genetics , Hep G2 Cells , Humans , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Signal Transduction/genetics , Transcription Factors/metabolism , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...