101
|
Wei S, Chu PC, Chuang HC, Hung WC, Kulp SK, Chen CS. Targeting the oncogenic E3 ligase Skp2 in prostate and breast cancer cells with a novel energy restriction-mimetic agent. PLoS One 2012; 7:e47298. [PMID: 23071779 PMCID: PMC3470570 DOI: 10.1371/journal.pone.0047298] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 09/12/2012] [Indexed: 11/18/2022] Open
Abstract
Substantial evidence supports the oncogenic role of the E3 ubiquitin ligase S-phase kinase-associated protein 2 (Skp2) in many types of cancers through its ability to target a broad range of signaling effectors for ubiquitination. Thus, this oncogenic E3 ligase represents an important target for cancer drug discovery. In this study, we report a novel mechanism by which CG-12, a novel energy restriction-mimetic agent (ERMA), down-regulates the expression of Skp2 in prostate cancer cells. Pursuant to our previous finding that upregulation of β-transducin repeat-containing protein (β-TrCP) expression represents a cellular response in cancer cells to ERMAs, including CG-12 and 2-deoxyglucose, we demonstrated that this β-TrCP accumulation resulted from decreased Skp2 expression. Evidence indicates that Skp2 targets β-TrCP for degradation via the cyclin-dependent kinase 2-facilitated recognition of the proline-directed phosphorylation motif 412SP. This Skp2 downregulation was attributable to Sirt1-dependent suppression of COP9 signalosome (Csn)5 expression in response to CG-12, leading to increased cullin 1 neddylation in the Skp1-cullin1-F-box protein complex and consequent Skp2 destabilization. Moreover, we determined that Skp2 and β-TrCP are mutually regulated, providing a feedback mechanism that amplifies the suppressive effect of ERMAs on Skp2. Specifically, cellular accumulation of β-TrCP reduced the expression of Sp1, a β-TrCP substrate, which, in turn, reduced Skp2 gene expression. This Skp2-β-TrCP-Sp1 feedback loop represents a novel crosstalk mechanism between these two important F-box proteins in cancer cells with aberrant Skp2 expression under energy restriction, which provides a proof-of-concept that the oncogenic Csn5/Skp2 signaling axis represents a “druggable” target for this novel ERMA.
Collapse
Affiliation(s)
- Shuo Wei
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America
| | - Po-Chen Chu
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America
| | - Hsiao-Ching Chuang
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America
| | - Wen-Chun Hung
- National Institute of Cancer Research, National Health Research Institute, Zhunan, Miaoli County, Taiwan
| | - Samuel K. Kulp
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America
| | - Ching-Shih Chen
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America
- Institute of Basic Medical Sciences, National Cheng-Kung University, Tainan, Taiwan
- * E-mail:
| |
Collapse
|
102
|
|
103
|
Chowdhry S, Zhang Y, McMahon M, Sutherland C, Cuadrado A, Hayes JD. Nrf2 is controlled by two distinct β-TrCP recognition motifs in its Neh6 domain, one of which can be modulated by GSK-3 activity. Oncogene 2012; 32:3765-81. [PMID: 22964642 PMCID: PMC3522573 DOI: 10.1038/onc.2012.388] [Citation(s) in RCA: 528] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 07/11/2012] [Accepted: 07/13/2012] [Indexed: 02/07/2023]
Abstract
Identification of regulatable mechanisms by which transcription factor NF-E2 p45-related factor 2 (Nrf2) is repressed will allow strategies to be designed that counter drug resistance associated with its up-regulation in tumours that harbour somatic mutations in Kelch-like ECH-associated protein-1 (Keap1), a gene that encodes a joint adaptor and substrate receptor for the Cul3-Rbx1/Roc1 ubiquitin ligase. We now show that mouse Nrf2 contains two binding sites for β-transducin repeat-containing protein (β-TrCP), which acts as a substrate receptor for the Skp1-Cul1-Rbx1/Roc1 ubiquitin ligase complex. Deletion of either binding site in Nrf2 decreased β-TrCP-mediated ubiquitylation of the transcription factor. The ability of one of the two β-TrCP-binding sites to serve as a degron could be both increased and decreased by manipulation of glycogen synthase kinase-3 (GSK-3) activity. Biotinylated-peptide pull-down assays identified DSGIS338 and DSAPGS378 as the two β-TrCP-binding motifs in Nrf2. Significantly, our pull-down assays indicated that β-TrCP binds a phosphorylated version of DSGIS more tightly than its non-phosphorylated counterpart, whereas this was not the case for DSAPGS. These data suggest that DSGIS, but not DSAPGS, contains a functional GSK-3 phosphorylation site. Activation of GSK-3 in Keap1-null mouse embryonic fibroblasts (MEFs), or in human lung A549 cells that contain mutant Keap1, by inhibition of the phosphoinositide 3-kinase (PI3K) – protein kinase B (PKB)/Akt pathway markedly reduced endogenous Nrf2 protein and decreased to 10-50% of normal the levels of mRNA for prototypic Nrf2-regulated enzymes, including the glutamate-cysteine ligase catalytic and modifier subunits, glutathione S-transferases Alpha-1 and Mu-1, heme oxygenase-1 and NAD(P)H:quinone oxidoreductase-1. Pre-treatment of Keap1−/− MEFs or A549 cells with the LY294002 PI3K inhibitor or the MK-2206 PKB/Akt inhibitor increased their sensitivity to acrolein, chlorambucil and cisplatin between 1.9-fold and 3.1-fold, and this was substantially attenuated by simultaneous pre-treatment with the GSK-3 inhibitor CT99021.
Collapse
Affiliation(s)
- S Chowdhry
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, UK
| | | | | | | | | | | |
Collapse
|
104
|
ATF4 interacts with Abro1/KIAA0157 scaffold protein and participates in a cytoprotective pathway. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1823:2149-56. [PMID: 22974638 DOI: 10.1016/j.bbamcr.2012.08.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 08/13/2012] [Accepted: 08/28/2012] [Indexed: 11/22/2022]
Abstract
Abro1 (Abraxas brother 1), also known as KIAA0157, is a scaffold protein that recruits various polypeptides to assemble the BRISC (BRCC36 isopeptide) deubiquitinating enzyme (DUB) complex. The BRISC enzyme has a Lys63-linked deubiquitinating activity and is comprised of four known subunits: MERIT40 (mediator of Rap80 interactions and targeting 40kDa), BRE (brain and reproductive organ-expressed), BRCC36 (BRCA1/BRCA2-containing complex, subunit 3) and Abro1. We have previously shown that Abro1 has a cytoprotective role that involves the BRISC DUB complex acting on specific Lys63-linked polyubiquitinated substrates. In this report we identify three members of the AP-1 (activating protein-1) family, the ATF4, ATF5 (activating transcription factor) and JunD proteins, as specific interactors of Abro1. The function of ATF4-Abro1 interaction was investigated under normal conditions as well as under cellular stress. Abro1 is predominantly cytoplasmic, but during cellular stress it enters the nucleus and co-localizes with ATF4. Furthermore, this interaction with ATF4 is necessary and essential for the cytoprotective function of Abro1 following oxidative stress. The ability of Abro1 to specifically interact with a number of transcription factors suggests a new mechanism of regulation of the BRISC DUB complex. This regulation involves the participation of at least three known members of the AP-1 family of transcription factors.
Collapse
|
105
|
Padrissa-Altés S, Zaouali MA, Boncompagni E, Bonaccorsi-Riani E, Carbonell T, Bardag-Gorce F, Oliva J, French SW, Bartrons R, Roselló-Catafau J. The use of a reversible proteasome inhibitor in a model of Reduced-Size Orthotopic Liver transplantation in rats. Exp Mol Pathol 2012; 93:99-110. [PMID: 22475623 DOI: 10.1016/j.yexmp.2012.03.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 03/19/2012] [Indexed: 12/18/2022]
Abstract
Ischemia/reperfusion injury (IRI), inherent in liver transplantation (LT), is the main cause of initial deficiencies and primary non-function of liver allografts. Living-related LT was developed to alleviate the mortality resulting from the scarcity of suitable deceased grafts. The main problem in using living-related LT for adults is graft size disparity. In this study we propose for the first time that the use of a proteasome inhibitor (Bortezomib) treatment could improve liver regeneration and reduce IRI after Reduced-Size Orthotopic Liver transplantation (ROLT). Rat liver grafts were reduced by removing the left lateral lobe and the two caudate lobes and preserved in UW or IGL-1 preservation solution for 1h liver and then subjected to ROLT with or without Bortezomib treatment. Our results show that Bortezomib reduces IRI after LT and is correlated with a reduction in mitochondrial damage, oxidative stress and endoplasmic reticulum stress. Furthermore, Bortezomib also increased liver regeneration after reduced-size LT and increased the expression of well-known ischemia/reperfusion protective proteins such as nitric oxide synthase, heme oxigenase 1 (HO-1) and Heat Shock Protein 70. Our results open new possibilities for the study of alternative therapeutic strategies aimed at reducing IRI and increasing liver regeneration after LT. It is hoped that the results of our study will contribute towards improving the understanding of the molecular processes involved in IRI and liver regeneration, and therefore help to improve the outcome of this type of LT in the future.
Collapse
Affiliation(s)
- Susagna Padrissa-Altés
- Experimental Hepatic Ischemia-Reperfusion Unit, Institut d´Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
106
|
Lee SH, Min KW, Zhang X, Baek SJ. 3,3'-diindolylmethane induces activating transcription factor 3 (ATF3) via ATF4 in human colorectal cancer cells. J Nutr Biochem 2012; 24:664-71. [PMID: 22819556 DOI: 10.1016/j.jnutbio.2012.03.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 02/27/2012] [Accepted: 03/22/2012] [Indexed: 12/12/2022]
Abstract
3,3'-Diindolylmethane (DIM) is a major in vivo condensation product of indole-3-carbinol, which is present in cruciferous vegetables. Although these compounds have been widely implicated in antitumorigenic and proapoptotic properties in animal as well as in vitro models of cancer, the underlying cellular mechanisms regulated by DIM are only partially understood. Activating transcription factor 3 (ATF3) is a member of the ATF/c-AMP response element-binding (CREB) subfamily of the basic-region leucine zipper family and has been known to induce apoptosis in human colorectal cancer (CRC) cells. The present study was performed to elucidate the molecular mechanism of ATF3 induction by DIM in human CRC cells. The DIM treatment induced apoptosis and induced ATF3 gene expression at protein and messenger RNA levels. DIM increased ATF3 promoter activity, and the region of -84 to +34 within ATF3 promoter was responsible for promoter activation by DIM. This region contained an ATF binding site. Deletion and point mutation of the ATF binding site (-23 to -16) abolished ATF3 promoter activation by DIM, and overexpression of ATF4 enhanced ATF3 transactivation. Chromatin immunoprecipitation assay confirmed the binding of ATF4 in the ATF3 promoter. Inhibition of ATF4 expression by small interference RNA results in repression of DIM-induced ATF3 expression. The current study demonstrates that DIM stimulates ATF3 expression through ATF4-mediated pathway and subsequently induces apoptosis in human CRC cells.
Collapse
Affiliation(s)
- Seong-Ho Lee
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996-4542, USA
| | | | | | | |
Collapse
|
107
|
Uyama M, Sato MM, Kawanami M, Tamura M. Regulation of osteoblastic differentiation by the proteasome inhibitor bortezomib. Genes Cells 2012; 17:548-58. [DOI: 10.1111/j.1365-2443.2012.01611.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 03/25/2012] [Indexed: 12/20/2022]
Affiliation(s)
| | - Mari M. Sato
- Biochemistry and Molecular Biology; Graduate School of Dental Medicine; Hokkaido University; Sapporo; 060-8586; Japan
| | - Masamitsu Kawanami
- Periodontology and Endodontology; Graduate School of Dental Medicine; Hokkaido University; Sapporo; 060-8586; Japan
| | - Masato Tamura
- Biochemistry and Molecular Biology; Graduate School of Dental Medicine; Hokkaido University; Sapporo; 060-8586; Japan
| |
Collapse
|
108
|
CK2 regulates ATF4 and CHOP transcription within the cellular stress response signalling pathway. Cell Signal 2012; 24:1797-802. [PMID: 22609407 DOI: 10.1016/j.cellsig.2012.05.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Accepted: 05/09/2012] [Indexed: 11/22/2022]
Abstract
Protein kinase CK2 is an ubiquitously expressed serine/threonine kinase. The protein levels along with CK2 activity are highly elevated in tumour cells where it protects cells from apoptosis. Accordingly, inhibition of CK2 is known to induce programmed cell death, making it a promising target for cancer therapy. Analysis of the different behaviour of hormone sensitive LNCaP cells and hormone refractory PC-3 cells after CK2 inhibition revealed CHOP ((C/EBP)-homologous protein) induction and therefore probably ER stress as crucial for apoptosis in the LNCaP cells. In the present study we investigated which promoter element of the CHOP promoter is responsible for its induction. ER stress can be generated by the accumulation of unfolded proteins, by depletion of amino acids or by oxidative stress. ER stress induces specific signalling pathways. In order to analyse which pathway might be activated by CK2 inhibition we started to analyse the activation of the different CHOP promoter elements. By using mutated reporter constructs of the CHOP promoter, it turned out that the amino acid response element (AARE) is the most prominent element for CHOP induction after CK2 inhibition. The ER stress element, however, proves to be less crucial, and along with the AP-1 binding site, they do not seem to play any role. Further we found an up-regulation of the transcription factor ATF4 after CK2 inhibition. ATF4 is involved in ER stress signalling through the AARE, which further supports our finding that CK2 inhibition provokes an amino acid induced response pathway.
Collapse
|
109
|
Malavasi ELV, Ogawa F, Porteous DJ, Millar JK. DISC1 variants 37W and 607F disrupt its nuclear targeting and regulatory role in ATF4-mediated transcription. Hum Mol Genet 2012; 21:2779-92. [PMID: 22422769 PMCID: PMC3363331 DOI: 10.1093/hmg/dds106] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Disrupted-In-Schizophrenia 1 (DISC1), a strong genetic candidate for psychiatric illness, encodes a multicompartmentalized molecular scaffold that regulates interacting proteins with key roles in neurodevelopment and plasticity. Missense DISC1 variants are associated with the risk of mental illness and with brain abnormalities in healthy carriers, but the underlying mechanisms are unclear. We examined the effect of rare and common DISC1 amino acid substitutions on subcellular targeting. We report that both the rare putatively causal variant 37W and the common variant 607F independently disrupt DISC1 nuclear targeting in a dominant-negative fashion, predicting that DISC1 nuclear expression is impaired in 37W and 607F carriers. In the nucleus, DISC1 interacts with the transcription factor Activating Transcription Factor 4 (ATF4), which is involved in the regulation of cellular stress responses, emotional behaviour and memory consolidation. At basal cAMP levels, wild-type DISC1 inhibits the transcriptional activity of ATF4, an effect that is weakened by both 37W and 607F independently, most likely as a consequence of their defective nuclear targeting. The common variant 607F additionally reduces DISC1/ATF4 interaction, which likely contributes to its weakened inhibitory effect. We also demonstrate that DISC1 modulates transcriptional responses to endoplasmic reticulum stress, and that this modulatory effect is ablated by 37W and 607F. By showing that DISC1 amino acid substitutions associated with psychiatric illness affect its regulatory function in ATF4-mediated transcription, our study highlights a potential mechanism by which these variants may impact on transcriptional events mediating cognition, emotional reactivity and stress responses, all processes of direct relevance to psychiatric illness.
Collapse
Affiliation(s)
- Elise L V Malavasi
- The Centre for Molecular Medicine at the Medical Research Council Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | | | | | | |
Collapse
|
110
|
Abstract
The noncanonical nuclear factor-κB (NF-κB) signaling pathway mediates activation of the p52/RelB NF-κB complex and, thereby, regulates specific immunological processes. This NF-κB pathway relies on the inducible processing of NF-κB2 precursor protein, p100, as opposed to the degradation of IκBα in the canonical NF-κB pathway. A central signaling component of the noncanonical NF-κB pathway is NF-κB-inducing kinase (NIK), which functions together with a downstream kinase, IKKα (inhibitor of NF-κB kinase α), to induce phosphorylation-dependent ubiquitination and processing of p100. Under normal conditions, NIK is targeted for continuous degradation by a tumor necrosis factor (TNF) receptor-associated factor-3 (TRAF3)-dependent E3 ubiquitin ligase. In response to signals mediated by a subset of TNF receptor superfamily members, NIK becomes stabilized as a result of TRAF3 degradation, leading to the activation of noncanonical NF-κB. This review discusses both the historical perspectives and the recent progress in the regulation and biological function of the noncanonical NF-κB pathway.
Collapse
Affiliation(s)
- Shao-Cong Sun
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
111
|
St-Arnaud R, Hekmatnejad B. Combinatorial control of ATF4-dependent gene transcription in osteoblasts. Ann N Y Acad Sci 2012; 1237:11-8. [PMID: 22082360 DOI: 10.1111/j.1749-6632.2011.06197.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Osteoblast-specific gene transcription requires interaction between bone cell-specific transcription factors and more widely expressed transcriptional regulators. This is particularly evident for the basic domain-leucine zipper factor activating transcription factor 4 (ATF4), whose activity can be enhanced or inhibited through interaction with other leucine zipper proteins, intermediate filament proteins, components of the basic transcriptional machinery, nuclear matrix attachment molecules, or ubiquitously expressed transcription factors. We discuss the results supporting the relevance of these interactions and present the first evidence of a functional interaction between ATF4, FIAT (factor-inhibiting ATF4-mediated transcription), and αNAC (nascent polypeptide-associated complex and coactivator alpha), three proteins that have been previously shown to associate using various protein-protein interaction assays.
Collapse
Affiliation(s)
- René St-Arnaud
- Genetics Unit, Shriners Hospital for Children, Montreal, Canada.
| | | |
Collapse
|
112
|
Mutation of ATF4 mediates resistance of neuronal cell lines against oxidative stress by inducing xCT expression. Cell Death Differ 2011; 19:847-58. [PMID: 22095285 DOI: 10.1038/cdd.2011.165] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Selecting neuronal cell lines for resistance against oxidative stress might recapitulate some adaptive processes in neurodegenerative diseases where oxidative stress is involved like Parkinson's disease. We recently reported that in hippocampal HT22 cells selected for resistance against oxidative glutamate toxicity, the cystine/glutamate antiporter system x(c)(-), which imports cystine for synthesis of the antioxidant glutathione, and its specific subunit, xCT, are upregulated. (Lewerenz et al., J Neurochem 98(3):916-25). Here, we show that in these glutamate-resistant HT22 cells upregulation of xCT mediates glutamate resistance, and xCT expression is induced by upregulation of the transcription factor ATF4. The mechanism of ATF4 upregulation consists of a 13 bp deletion in the upstream open reading frame (uORF2) overlapping the ATF4 open reading frame. The resulting uORF2-ATF4 fusion protein is efficiently translated even at a low phosphorylation levels of the translation initiation factor eIF2α, a condition under which ATF4 translation is normally suppressed. A similar ATF4 mutation associated with prominent upregulation of xCT expression was identified in PC12 cells selected for resistance against amyloid β-peptide. Our data indicate that ATF4 has a central role in regulating xCT expression and resistance against oxidative stress. ATF4 mutations might have broader significance as upregulation of xCT is found in tumor cells and associated with anticancer drug resistance.
Collapse
|
113
|
PHD1 interacts with ATF4 and negatively regulates its transcriptional activity without prolyl hydroxylation. Exp Cell Res 2011; 317:2789-99. [PMID: 21951999 DOI: 10.1016/j.yexcr.2011.09.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2010] [Revised: 09/04/2011] [Accepted: 09/09/2011] [Indexed: 12/21/2022]
Abstract
Cellular response to hypoxia plays an important role in both circulatory and pulmonary diseases and cancer. Hypoxia-inducible factors (HIFs) are major transcription factors regulating the response to hypoxia. The α-subunits of HIFs are hydroxylated by members of the prolyl-4-hydroxylase domain (PHD) family, PHD1, PHD2, and PHD3, in an oxygen-dependent manner. Here, we report on the identification of ATF4 as a protein interacting with PHD1 as well as PHD3, but not with PHD2. The central region of ATF4 including the Zipper II domain, ODD domain and β-TrCP recognition motif were involved in the interaction with PHD1. Coexistence of PHD1 stabilized ATF4, as opposed to the destabilization of ATF4 by PHD3. Moreover, coexpression of ATF4 destabilized PHD3, whereas PHD1 stability was not affected by the presence of ATF4. Mutations to alanine of proline residues in ATF4 that satisfied hydroxylation consensus by PHDs did not affect binding activity of ATF4 to PHD1 and PHD3. Furthermore, in vitro prolyl hydroxylation assay clearly indicated that ATF4 did not serve as a substrate of both PHD1 and PHD3. Coexpression of PHD1 or PHD3 with ATF4 repressed the transcriptional activity of ATF4. These results suggest that PHD1 and PHD3 control the transactivation activity of ATF4.
Collapse
|
114
|
Teske BF, Wek SA, Bunpo P, Cundiff JK, McClintick JN, Anthony TG, Wek RC. The eIF2 kinase PERK and the integrated stress response facilitate activation of ATF6 during endoplasmic reticulum stress. Mol Biol Cell 2011; 22:4390-405. [PMID: 21917591 PMCID: PMC3216664 DOI: 10.1091/mbc.e11-06-0510] [Citation(s) in RCA: 308] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
This study shows that the eIF2 kinase PERK is required not only for translational control but also for activation of ATF6 and its target genes in the unfolded protein response. The PERK pathway facilitates both the synthesis of ATF6 and trafficking of ATF6 from the endoplasmic reticulum to the Golgi for intramembrane proteolysis and activation of ATF6. Disruptions of the endoplasmic reticulum (ER) that perturb protein folding cause ER stress and elicit an unfolded protein response (UPR) that involves translational and transcriptional changes in gene expression aimed at expanding the ER processing capacity and alleviating cellular injury. Three ER stress sensors (PERK, ATF6, and IRE1) implement the UPR. PERK phosphorylation of the α subunit of eIF2 during ER stress represses protein synthesis, which prevents further influx of ER client proteins. Phosphorylation of eIF2α (eIF2α∼P) also induces preferential translation of ATF4, a transcription activator of the integrated stress response. In this study we show that the PERK/eIF2α∼P/ATF4 pathway is required not only for translational control, but also for activation of ATF6 and its target genes. The PERK pathway facilitates both the synthesis of ATF6 and trafficking of ATF6 from the ER to the Golgi for intramembrane proteolysis and activation of ATF6. As a consequence, liver-specific depletion of PERK significantly reduces both the translational and transcriptional phases of the UPR, leading to reduced protein chaperone expression, disruptions of lipid metabolism, and enhanced apoptosis. These findings show that the regulatory networks of the UPR are fully integrated and help explain the diverse biological defects associated with loss of PERK.
Collapse
Affiliation(s)
- Brian F Teske
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | |
Collapse
|
115
|
Koyanagi S, Hamdan AM, Horiguchi M, Kusunose N, Okamoto A, Matsunaga N, Ohdo S. cAMP-response element (CRE)-mediated transcription by activating transcription factor-4 (ATF4) is essential for circadian expression of the Period2 gene. J Biol Chem 2011; 286:32416-23. [PMID: 21768648 DOI: 10.1074/jbc.m111.258970] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activating transcription factor (ATF)/cAMP-response element (CRE)-binding (CREB) proteins induce the CRE-mediated gene transcription depending on the cAMP stimulation. cAMP-dependent signaling oscillates in a circadian manner, which in turn also sustains core oscillation machinery of the circadian clock. Here, we show that among the ATF/CREB family proteins, ATF4 is essential for the circadian expression of the Period2 (Per2) gene, a key component of the circadian clock. Transcription of the Atf4 gene was regulated by core components of the circadian clock, and its expression exhibited circadian oscillation in mouse tissues as well as embryonic fibroblasts. ATF4 bound to the CRE of the Per2 promoter in a circadian time-dependent manner and periodically activated the transcription of the Per2 gene. Consequently, the oscillation of the Per2 expression was attenuated in embryonic cells prepared from Atf4-null mice. Furthermore, the loss of ATF4 also disrupted the rhythms in the expression of other clock genes. These results suggest that ATF4 is a component responsible for sustaining circadian oscillation of CRE-mediated gene expression and also constitute a molecular link connecting cAMP-dependent signaling to the circadian clock.
Collapse
Affiliation(s)
- Satoru Koyanagi
- Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | |
Collapse
|
116
|
Wang A, Xu S, Zhang X, He J, Yan D, Yang Z, Xiao S. Ribosomal protein RPL41 induces rapid degradation of ATF4, a transcription factor critical for tumour cell survival in stress. J Pathol 2011; 225:285-92. [DOI: 10.1002/path.2918] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Revised: 04/04/2011] [Accepted: 04/09/2011] [Indexed: 12/28/2022]
|
117
|
Warfel NA, Niederst M, Stevens MW, Brennan PM, Frame MC, Newton AC. Mislocalization of the E3 ligase, β-transducin repeat-containing protein 1 (β-TrCP1), in glioblastoma uncouples negative feedback between the pleckstrin homology domain leucine-rich repeat protein phosphatase 1 (PHLPP1) and Akt. J Biol Chem 2011; 286:19777-88. [PMID: 21454620 PMCID: PMC3103356 DOI: 10.1074/jbc.m111.237081] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Revised: 03/25/2011] [Indexed: 12/16/2022] Open
Abstract
The PH domain leucine-rich repeat protein phosphatase, PHLPP, plays a central role in controlling the amplitude of growth factor signaling by directly dephosphorylating and thereby inactivating Akt. The cellular levels of PHLPP1 have recently been shown to be enhanced by its substrate, activated Akt, via modulation of a phosphodegron recognized by the E3 ligase β-TrCP1, thus providing a negative feedback loop to tightly control cellular Akt output. Here we show that this feedback loop is lost in aggressive glioblastoma but not less aggressive astrocytoma. Overexpression and pharmacological studies reveal that loss of the feedback loop does not result from a defect in PHLPP1 protein or in the upstream kinases that control its phosphodegron. Rather, the defect arises from altered localization of β-TrCP1; in astrocytoma cell lines and in normal brain tissue the E3 ligase is predominantly cytoplasmic, whereas in glioblastoma cell lines and patient-derived tumor neurospheres, the E3 ligase is confined to the nucleus and thus spatially separated from PHLPP1, which is cytoplasmic. Restoring the localization of β-TrCP1 to the cytosol of glioblastoma cells rescues the ability of Akt to regulate PHLPP1 stability. Additionally, we show that the degradation of another β-TrCP1 substrate, β-catenin, is impaired and accumulates in the cytosol of glioblastoma cell lines. Our findings reveal that the cellular localization of β-TrCP1 is altered in glioblastoma, resulting in dysregulation of PHLPP1 and other substrates such as β-catenin.
Collapse
Affiliation(s)
- Noel A. Warfel
- From the Department of Pharmacology and
- Biomedical Sciences Graduate Program, University of California at San Diego, La Jolla, California 92093 and
| | - Matt Niederst
- From the Department of Pharmacology and
- Biomedical Sciences Graduate Program, University of California at San Diego, La Jolla, California 92093 and
| | | | - Paul M. Brennan
- the Edinburgh Cancer Research Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| | - Margaret C. Frame
- the Edinburgh Cancer Research Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| | | |
Collapse
|
118
|
Kuo CL, Oyler GA, Shoemaker CB. Accelerated neuronal cell recovery from Botulinum neurotoxin intoxication by targeted ubiquitination. PLoS One 2011; 6:e20352. [PMID: 21629663 PMCID: PMC3101245 DOI: 10.1371/journal.pone.0020352] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 04/19/2011] [Indexed: 11/19/2022] Open
Abstract
Botulinum neurotoxin (BoNT), a Category A biodefense agent, delivers a protease to motor neuron cytosol that cleaves one or more soluble NSF attachment protein receptors (SNARE) proteins involved in neurotransmission to cause a flaccid paralysis. No antidotes exist to reverse symptoms of BoNT intoxication so severely affected patients require artificial respiration with prolonged intensive care. Time to recovery depends on toxin serotype because the intraneuronal persistence of the seven known BoNT serotypes varies widely from days to many months. Our therapeutic antidote strategy is to develop 'targeted F-box' (TFB) agents that target the different intraneuronal BoNT proteases for accelerated degradation by the ubiquitin proteasome system (UPS), thus promoting rapid recovery from all serotypes. These agents consist of a camelid heavy chain-only V(H) (VHH) domain specific for a BoNT protease fused to an F-box domain recognized by an intraneuronal E3-ligase. A fusion protein containing the 14 kDa anti-BoNT/A protease VHH, ALcB8, joined to a 15 kDa F-box domain region of TrCP (D5) was sufficient to cause increased ubiquitination and accelerate turnover of the targeted BoNT/A protease within neurons. Neuronal cells expressing this TFB, called D5-B8, were also substantially resistant to BoNT/A intoxication and recovered from intoxication at least 2.5 fold quicker than control neurons. Fusion of D5 to a VHH specific for BoNT/B protease (BLcB10) led to accelerated turnover of the targeted protease within neurons, thus demonstrating the modular nature of these therapeutic agents and suggesting that development of similar therapeutic agents specific to all botulinum serotypes should be readily achievable.
Collapse
Affiliation(s)
- Chueh-Ling Kuo
- Department of Biomedical Sciences, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - George A. Oyler
- Synaptic Research LLC, Baltimore, Maryland, United States of America
| | - Charles B. Shoemaker
- Department of Biomedical Sciences, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| |
Collapse
|
119
|
Lewerenz J, Maher P. Control of redox state and redox signaling by neural antioxidant systems. Antioxid Redox Signal 2011; 14:1449-65. [PMID: 20812872 DOI: 10.1089/ars.2010.3600] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The glutathione/glutathione disulfide (GSH/GSSG) redox pair forms the major redox couple in cells and as such plays a critical role in regulating redox-dependent cellular functions. Not only does GSH act as an antioxidant but it can also modulate the activity of a variety of different proteins. An impairment in GSH status is thought to be the precipitating event in a wide range of neurological disorders. Therefore, understanding how to maintain GSH in the CNS could provide a valuable therapeutic approach. Intracellular GSH levels are regulated by a complex series of pathways that include substrate transport and availability, rates of synthesis and regeneration, GSH utilization, and GSH efflux. To date, the most effective approaches for maintaining GSH levels in the CNS include enhancing cyst(e)ine uptake both directly and indirectly via transcriptional upregulation of system x(c)(-), increasing GSH synthesis via transcriptional upregulation of the rate limiting enzyme in GSH biosynthesis, and decreasing GSH utilization. Among the transcription factors that play critical roles in GSH metabolism are NF-E2-related factor 2 (Nrf2) and activating transcription factor 4 (ATF4). Thus, compounds that can upregulate these transcription factors may be particularly useful in promoting the functional maintenance of the CNS through their effects on GSH metabolism.
Collapse
Affiliation(s)
- Jan Lewerenz
- Department for Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | |
Collapse
|
120
|
Magne L, Blanc E, Legrand B, Lucas D, Barouki R, Rouach H, Garlatti M. ATF4 and the integrated stress response are induced by ethanol and cytochrome P450 2E1 in human hepatocytes. J Hepatol 2011; 54:729-37. [PMID: 21146245 DOI: 10.1016/j.jhep.2010.07.023] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2010] [Revised: 07/19/2010] [Accepted: 07/23/2010] [Indexed: 12/29/2022]
Abstract
BACKGROUND & AIMS Molecular mechanisms underlying alcoholic liver disease (ALD) are still not fully understood. Activating transcription factor-4 (ATF4) is the master coordinator of the integrated stress response (ISR), an adaptive pathway triggered by multiple stressors. which can promote cell death and induce metabolic dysregulation if the stress is intense or prolonged. The aim of this study was to assess the effect of alcohol on the ISR signaling pathway in human liver cells and to define the role of cytochrome P450 2E1 (CYP2E1) in this response. METHODS Primary cultured human hepatocytes and human HepG2 cells over-expressing CYP2E1 by adenoviral infection were exposed to ethanol (25-100mM) for 8-48h. RESULTS Ethanol treatment of both liver cells up-regulated ATF4 as well as the pro-survival and the pro-apoptotic transcriptional program of the ISR. Indeed, in CYP2E1-expressing HepG2 cells exposed to ethanol, the expression of ISR target genes (HMOX-1, GCLC, AsnS, IGFBP-1, GADD34,CHOP, ATF3, CHAC1) was induced. Up-regulation of ATF4 and the ISR transcriptional program was decreased by addition of the anti-oxidant glutathione. Several mechanisms mediated ATF4 protein induction, including, at early times, the phosphorylation of eIF2α which controls ATF4 translation, and, at later times, increased mRNA level and increased stability of the protein. A decrease in cell survival was also observed. CONCLUSIONS This study demonstrates that both CYP2E1 and ethanol induce ATF4 and the integrated stress response, a pathway which coordinates signals from multiple stresses, as well as established risk factors for ALD, and can display detrimental cellular effects upon prolonged activation.
Collapse
|
121
|
Putters J, Slotman JA, Gerlach JP, Strous GJ. Specificity, location and function of βTrCP isoforms and their splice variants. Cell Signal 2011; 23:641-7. [DOI: 10.1016/j.cellsig.2010.11.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Accepted: 11/25/2010] [Indexed: 11/25/2022]
|
122
|
Marion V, Sankaranarayanan S, de Theije C, van Dijk P, Lindsey P, Lamers MC, Harding HP, Ron D, Lamers WH, Köhler SE. Arginine deficiency causes runting in the suckling period by selectively activating the stress kinase GCN2. J Biol Chem 2011; 286:8866-74. [PMID: 21239484 PMCID: PMC3058991 DOI: 10.1074/jbc.m110.216119] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 01/13/2011] [Indexed: 12/18/2022] Open
Abstract
Suckling "F/A2" mice, which overexpress arginase-I in their enterocytes, develop a syndrome (hypoargininemia, reduced hair and muscle growth, impaired B-cell maturation) that resembles IGF1 deficiency. The syndrome may result from an impaired function of the GH-IGF1 axis, activation of the stress-kinase GCN2, and/or blocking of the mTORC1-signaling pathway. Arginine deficiency inhibited GH secretion and decreased liver Igf1 mRNA and plasma IGF1 concentration, but did not change muscle IGF1 concentration. GH supplementation induced Igf1 mRNA synthesis, but did not restore growth, ruling out direct involvement of the GH-IGF1 axis. In C2C12 muscle cells, arginine withdrawal activated GCN2 signaling, without impacting mTORC1 signaling. In F/A2 mice, the reduction of plasma and tissue arginine concentrations to ∼25% of wild-type values activated GCN2 signaling, but mTORC1-mediated signaling remained unaffected. Gcn2-deficient F/A2 mice suffered from hypoglycemia and died shortly after birth. Because common targets of all stress kinases (eIF2α phosphorylation, Chop mRNA expression) were not increased in these mice, the effects of arginine deficiency were solely mediated by GCN2.
Collapse
Affiliation(s)
- Vincent Marion
- From the Dept of Anatomy & Embryology and NUTRIM School for Nutrition, Toxicology, and Metabolism, and
| | | | - Chiel de Theije
- From the Dept of Anatomy & Embryology and NUTRIM School for Nutrition, Toxicology, and Metabolism, and
| | - Paul van Dijk
- From the Dept of Anatomy & Embryology and NUTRIM School for Nutrition, Toxicology, and Metabolism, and
| | - Patrick Lindsey
- the Department of Population Genetics, Genomics & Bioinformatics, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Marinus C. Lamers
- the Max-Planck Institute of Immunobiology, P.O. Box 1169, D-79011 Freiburg, Germany
| | - Heather P. Harding
- the Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, United Kingdom, and
| | - David Ron
- the Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, United Kingdom, and
| | - Wouter H. Lamers
- From the Dept of Anatomy & Embryology and NUTRIM School for Nutrition, Toxicology, and Metabolism, and
- the AMC Liver Center, Academic Medical Center, University of Amsterdam, P.O. Box 22660, 1100 DD, Amsterdam, The Netherlands
| | - S. Eleonore Köhler
- From the Dept of Anatomy & Embryology and NUTRIM School for Nutrition, Toxicology, and Metabolism, and
| |
Collapse
|
123
|
Neuronal apoptosis induced by endoplasmic reticulum stress is regulated by ATF4-CHOP-mediated induction of the Bcl-2 homology 3-only member PUMA. J Neurosci 2011; 30:16938-48. [PMID: 21159964 DOI: 10.1523/jneurosci.1598-10.2010] [Citation(s) in RCA: 278] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
An increasing body of evidence points to a key role of endoplasmic reticulum (ER) stress in acute and chronic neurodegenerative conditions. Extensive ER stress can trigger neuronal apoptosis, but the signaling pathways that regulate this cell death remain unclear. In the present study, we demonstrate that PUMA, a Bcl-2 homology 3 (BH3)-only member of the Bcl-2 family, is transcriptionally activated in cortical neurons by ER stress and is essential for ER-stress-induced cell death. PUMA is known to be a key transcriptional target of p53, but we have found that ER stress triggers PUMA induction and cell death through a p53-independent mechanism mediated by the ER-stress-inducible transcription factor ATF4 (activating transcription factor 4). Specifically, we demonstrate that ectopic expression of ATF4 sensitizes mouse cortical neurons to ER-stress-induced apoptosis and that ATF4-deficient neurons exhibit markedly reduced levels of PUMA expression and cell death. However, chromatin immunoprecipitation experiments suggest that ATF4 does not directly regulate the PUMA promoter. Rather, we found that ATF4 induces expression of the transcription factor CHOP (C/EBP homologous protein) and that CHOP in turn activates PUMA induction. Specifically, we demonstrate that CHOP binds to the PUMA promoter during ER stress and that CHOP knockdown attenuates PUMA induction and neuronal apoptosis. In summary, we have identified a key signaling pathway in ER-stress-induced neuronal death involving ATF4-CHOP-mediated transactivation of the proapoptotic Bcl-2 family member PUMA. We propose that this pathway may be an important therapeutic target relevant to a number of neurodegenerative conditions.
Collapse
|
124
|
Lee Y, Lee J, Kwon I, Nakajima Y, Ohmiya Y, Son GH, Lee KH, Kim K. Coactivation of the CLOCK-BMAL1 complex by CBP mediates resetting of the circadian clock. J Cell Sci 2011; 123:3547-57. [PMID: 20930143 DOI: 10.1242/jcs.070300] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The transcription factor CLOCK-BMAL1 is a core component of the molecular clock machinery that drives circadian gene expression and physiology in mammals. Recently, we reported that this heterodimeric transcription factor functions as a signaling molecule in response to the resetting stimuli via the Ca²+-dependent protein kinase C pathway. Here, we demonstrate that the CREB-binding protein (CBP) plays a key role in rapid activation of the CLOCK-BMAL1 heterodimer that leads to phase resetting of the circadian clock. Under physiological conditions, a bimolecular fluorescence complementation (BiFC) assay revealed that CLOCK and BMAL1 dimerize in the cytoplasm and subsequently translocate into the nucleus in response to serum stimuli (mean time duration was 29.2 minutes and mean velocity 0.7 μm/minute). Concomitantly, BMAL1 rapidly recruited CBP on Per1 promoter E-box, but not p300 (a functional analog of CBP), in the discrete nuclear foci. However, recruitment of CBP by cAMP/Ca²+ response element-binding (CREB) protein on CRE was not markedly increased upon delivery of the resetting stimuli. Furthermore, overexpression of CBP greatly potentiated the CLOCK-BMAL1-mediated Per1 transcription, and this effect was completely abolished by site-directed mutation of E-box elements, but not by the mutation of CRE in the Per1 promoter. Furthermore, molecular knockdown of CBP severely dampened circadian oscillation of clock gene expression triggered by the resetting stimuli. These findings suggest that CBP recruitment by BMAL1 mediates acute transactivation of CLOCK-BMAL1, thereby inducing immediate-early Per1 transcription and phase resetting of the circadian clock.
Collapse
Affiliation(s)
- Yool Lee
- Department of Biological Sciences and 21st Frontier Program in Neuroscience, Seoul National University, Seoul 151-742, Korea
| | | | | | | | | | | | | | | |
Collapse
|
125
|
Abstract
The non-canonical NF-κB pathway is an important arm of NF-κB signaling that predominantly targets activation of the p52/RelB NF-κB complex. This pathway depends on the inducible processing of p100, a molecule functioning as both the precursor of p52 and a RelB-specific inhibitor. A central signaling component of the non-canonical pathway is NF-κB-inducing kinase (NIK), which integrates signals from a subset of TNF receptor family members and activates a downstream kinase, IκB kinase-α (IKKα), for triggering p100 phosphorylation and processing. A unique mechanism of NIK regulation is through its fate control: the basal level of NIK is kept low by a TRAF-cIAP destruction complex and signal-induced non-canonical NF-κB signaling involves NIK stabilization. Tight control of the fate of NIK is important, since deregulated NIK accumulation is associated with lymphoid malignancies.
Collapse
Affiliation(s)
- Shao-Cong Sun
- Department of Immunology, The University of Texas MD Anderson Cancer Center, The University of Texas Graduate School of Biomedical Sciences at Houston, 7455 Fannin Street, Box 902, Houston, TX 77030, USA.
| |
Collapse
|
126
|
Lau AW, Pringle LM, Quick L, Riquelme DN, Ye Y, Oliveira AM, Chou MM. TRE17/ubiquitin-specific protease 6 (USP6) oncogene translocated in aneurysmal bone cyst blocks osteoblastic maturation via an autocrine mechanism involving bone morphogenetic protein dysregulation. J Biol Chem 2010; 285:37111-20. [PMID: 20864534 DOI: 10.1074/jbc.m110.175133] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Aneurysmal bone cyst (ABC) is a pediatric osseous tumor characterized by extensive destruction of the surrounding bone. The molecular mechanisms underlying its pathogenesis are completely unknown. Recent work showed that translocation of the TRE17/USP6 locus occurs in over 60% of ABC cases resulting in TRE17 overexpression. Immature osteoblasts are presumed to be the cell type harboring translocation of TRE17 in at least a subset of ABCs. However, the effects of TRE17 overexpression on transformation and osteoblast function are unknown. TRE17 encodes a ubiquitin-specific protease (USP) and a TBC (TRE2-Bub2-Cdc16) domain that promotes activation of the Arf6 GTPase. Here we report that TRE17 potently inhibits the maturation of MC3T3 pre-osteoblasts in a USP-dependent and Arf6-independent manner. Notably, we find that TRE17 function is mediated through an autocrine mechanism. Transcriptome analysis of TRE17-expressing cells reveals dysregulation of several pathways with established roles in osteoblast maturation. In particular, signaling through the bone morphogenetic protein (BMP) pathway, a key regulator of osteogenesis, is profoundly altered. TRE17 simultaneously inhibits the expression of BMP-4 while augmenting the BMP antagonist, Gremlin-1. Osteoblastic maturation is restored in TRE17-expressing cells by the addition of exogenous BMP-4, thus establishing a functional role for BMP-4 during TRE17-induced transformation. Because bone homeostasis involves a precise balance between the activities of osteoblasts and osteoclasts, our studies raise the possibility that attenuated osteoblast maturation caused by TRE17 overexpression may contribute to the bone loss/destruction observed in ABC.
Collapse
Affiliation(s)
- Alan W Lau
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | |
Collapse
|
127
|
Frank CL, Ge X, Xie Z, Zhou Y, Tsai LH. Control of activating transcription factor 4 (ATF4) persistence by multisite phosphorylation impacts cell cycle progression and neurogenesis. J Biol Chem 2010; 285:33324-33337. [PMID: 20724472 PMCID: PMC2963346 DOI: 10.1074/jbc.m110.140699] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Organogenesis is a highly integrated process with a fundamental requirement for precise cell cycle control. Mechanistically, the cell cycle is composed of transitions and thresholds that are controlled by coordinated post-translational modifications. In this study, we describe a novel mechanism controlling the persistence of the transcription factor ATF4 by multisite phosphorylation. Proline-directed phosphorylation acted additively to regulate multiple aspects of ATF4 degradation. Stabilized ATF4 mutants exhibit decreased β-TrCP degron phosphorylation, β-TrCP interaction, and ubiquitination, as well as elicit early G1 arrest. Expression of stabilized ATF4 also had significant consequences in the developing neocortex. Mutant ATF4 expressing cells exhibited positioning and differentiation defects that were attributed to early G1 arrest, suggesting that neurogenesis is sensitive to ATF4 dosage. We propose that precise regulation of the ATF4 dosage impacts cell cycle control and impinges on neurogenesis.
Collapse
Affiliation(s)
- Christopher L Frank
- From the Massachusetts Institute of Technology, Picower Institute for Learning and Memory, the Howard Hughes Medical Institute, Cambridge, Massachusetts 02139, the Stanley Center for Psychiatric Research
| | - Xuecai Ge
- From the Massachusetts Institute of Technology, Picower Institute for Learning and Memory, the Howard Hughes Medical Institute, Cambridge, Massachusetts 02139, the Stanley Center for Psychiatric Research
| | - Zhigang Xie
- From the Massachusetts Institute of Technology, Picower Institute for Learning and Memory, the Howard Hughes Medical Institute, Cambridge, Massachusetts 02139, the Stanley Center for Psychiatric Research; Departments of Neurosurgery and Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Ying Zhou
- From the Massachusetts Institute of Technology, Picower Institute for Learning and Memory, the Howard Hughes Medical Institute, Cambridge, Massachusetts 02139, the Stanley Center for Psychiatric Research
| | - Li-Huei Tsai
- From the Massachusetts Institute of Technology, Picower Institute for Learning and Memory, the Howard Hughes Medical Institute, Cambridge, Massachusetts 02139, the Stanley Center for Psychiatric Research; Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02139.
| |
Collapse
|
128
|
Cao H, Yu S, Yao Z, Galson DL, Jiang Y, Zhang X, Fan J, Lu B, Guan Y, Luo M, Lai Y, Zhu Y, Kurihara N, Patrene K, Roodman GD, Xiao G. Activating transcription factor 4 regulates osteoclast differentiation in mice. J Clin Invest 2010; 120:2755-66. [PMID: 20628199 PMCID: PMC2912190 DOI: 10.1172/jci42106] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Accepted: 05/26/2010] [Indexed: 01/03/2023] Open
Abstract
Activating transcription factor 4 (ATF4) is a critical transcription factor for osteoblast (OBL) function and bone formation; however, a direct role in osteoclasts (OCLs) has not been established. Here, we targeted expression of ATF4 to the OCL lineage using the Trap promoter or through deletion of Atf4 in mice. OCL differentiation was drastically decreased in Atf4-/- bone marrow monocyte (BMM) cultures and bones. Coculture of Atf4-/- BMMs with WT OBLs or a high concentration of RANKL failed to restore the OCL differentiation defect. Conversely, Trap-Atf4-tg mice displayed severe osteopenia with dramatically increased osteoclastogenesis and bone resorption. We further showed that ATF4 was an upstream activator of the critical transcription factor Nfatc1 and was critical for RANKL activation of multiple MAPK pathways in OCL progenitors. Furthermore, ATF4 was crucial for M-CSF induction of RANK expression on BMMs, and lack of ATF4 caused a shift in OCL precursors to macrophages. Finally, ATF4 was largely modulated by M-CSF signaling and the PI3K/AKT pathways in BMMs. These results demonstrate that ATF4 plays a direct role in regulating OCL differentiation and suggest that it may be a therapeutic target for treating bone diseases associated with increased OCL activity.
Collapse
Affiliation(s)
- Huiling Cao
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Immunology, Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin, China.
Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China.
Department of Surgery and
Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
College of Life Sciences, Nankai University, Tianjin, China
| | - Shibing Yu
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Immunology, Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin, China.
Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China.
Department of Surgery and
Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
College of Life Sciences, Nankai University, Tianjin, China
| | - Zhi Yao
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Immunology, Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin, China.
Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China.
Department of Surgery and
Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
College of Life Sciences, Nankai University, Tianjin, China
| | - Deborah L. Galson
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Immunology, Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin, China.
Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China.
Department of Surgery and
Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
College of Life Sciences, Nankai University, Tianjin, China
| | - Yu Jiang
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Immunology, Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin, China.
Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China.
Department of Surgery and
Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaoyan Zhang
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Immunology, Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin, China.
Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China.
Department of Surgery and
Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
College of Life Sciences, Nankai University, Tianjin, China
| | - Jie Fan
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Immunology, Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin, China.
Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China.
Department of Surgery and
Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
College of Life Sciences, Nankai University, Tianjin, China
| | - Binfeng Lu
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Immunology, Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin, China.
Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China.
Department of Surgery and
Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
College of Life Sciences, Nankai University, Tianjin, China
| | - Youfei Guan
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Immunology, Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin, China.
Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China.
Department of Surgery and
Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
College of Life Sciences, Nankai University, Tianjin, China
| | - Min Luo
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Immunology, Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin, China.
Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China.
Department of Surgery and
Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
College of Life Sciences, Nankai University, Tianjin, China
| | - Yumei Lai
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Immunology, Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin, China.
Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China.
Department of Surgery and
Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
College of Life Sciences, Nankai University, Tianjin, China
| | - Yibei Zhu
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Immunology, Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin, China.
Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China.
Department of Surgery and
Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
College of Life Sciences, Nankai University, Tianjin, China
| | - Noriyoshi Kurihara
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Immunology, Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin, China.
Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China.
Department of Surgery and
Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
College of Life Sciences, Nankai University, Tianjin, China
| | - Kenneth Patrene
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Immunology, Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin, China.
Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China.
Department of Surgery and
Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
College of Life Sciences, Nankai University, Tianjin, China
| | - G. David Roodman
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Immunology, Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin, China.
Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China.
Department of Surgery and
Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
College of Life Sciences, Nankai University, Tianjin, China
| | - Guozhi Xiao
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Immunology, Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin, China.
Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China.
Department of Surgery and
Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
129
|
Inageda K. Insulin modulates induction of glucose-regulated protein 78 during endoplasmic reticulum stress via augmentation of ATF4 expression in human neuroblastoma cells. FEBS Lett 2010; 584:3649-54. [PMID: 20667453 DOI: 10.1016/j.febslet.2010.07.040] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2010] [Revised: 07/19/2010] [Accepted: 07/22/2010] [Indexed: 12/29/2022]
Abstract
The effect of insulin on endoplasmic reticulum (ER) stress was investigated. Insulin protected cell death induced by ER stress and increased glucose-regulated protein 78 (GRP78) mRNA and protein levels. Insulin also significantly increased activating transcription factor-4 (ATF4) protein in the nucleus, which was inhibited by LY294002, a phosphatidylinositol 3-kinase (PI-3 kinase) inhibitor. The increase of ATF4 protein by insulin was not due to transcriptional or translational up-regulation but to a post-translational mechanism. Knockdown of ATF4 by siRNA significantly inhibited GRP78 induction by insulin. These results indicate that insulin modulated ER stress-induced GRP78 expression occurs via ATF4 up-regulation.
Collapse
Affiliation(s)
- Kiyoshi Inageda
- Department of Hygiene and Public Health I, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan.
| |
Collapse
|
130
|
Suzuki T, Osumi N, Wakamatsu Y. Stabilization of ATF4 protein is required for the regulation of epithelial-mesenchymal transition of the avian neural crest. Dev Biol 2010; 344:658-68. [PMID: 20580702 DOI: 10.1016/j.ydbio.2010.05.492] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2010] [Revised: 05/13/2010] [Accepted: 05/16/2010] [Indexed: 12/20/2022]
Abstract
Epithelial-mesenchymal transition (EMT) permits neural crest cells to delaminate from the epithelial ectoderm and to migrate extensively in the embryonic environment. In this study, we have identified ATF4, a basic-leucine-zipper transcription factor, as one of the neural crest EMT regulators. Although ATF4 alone was not sufficient to drive the formation of migratory neural crest cells, ATF4 cooperated with Sox9 to induce neural crest EMT by controlling the expression of cell-cell and cell-extracellular matrix adhesion molecules. This was likely, at least in part, by inducing the expression of Foxd3, which encodes another neural crest transcription factor. We also found that the ATF4 protein level was strictly regulated by proteasomal degradation and p300-mediated stabilization, allowing ATF4 protein to accumulate in the nuclei of neural crest cells undergoing EMT. Thus, our results emphasize the importance of the regulation of protein stability in the neural crest EMT.
Collapse
Affiliation(s)
- Takashi Suzuki
- Center for Translational and Advanced Animal Research on Human Diseases, Division of Developmental Neuroscience, Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | | | | |
Collapse
|
131
|
Abstract
The basic domain-leucine zipper transcription factor activating transcription factor 4 (ATF4) regulates most functions of the osteoblast. It is therefore not surprising that its activity should be regulated through several mechanisms. Factor inhibiting ATF4-mediated transcription (FIAT) is a leucine zipper nuclear molecule lacking a basic domain for DNA binding that interacts with ATF4 to repress its transcriptional activity. FIAT expression was monitored in parallel with ATF4 during osteoblastogenesis. The mechanism of ATF4 repression by FIAT was investigated through structure-function analysis. The physiological significance of FIAT expression in osteoblasts was studied through silencing FIAT in osteoblasts by RNA interference, as well as through characterization of two genetic mouse models: FIAT transgenic mice which overexpress FIAT in osteoblasts, and FIAT knockout mice. Studies to date show that FIAT and ATF4 are co-expressed in osteoblasts, and that FIAT inhibition of matrix mineralization requires dimerization with ATF4 through the second leucine zipper. Furthermore, transgenic mice overexpressing FIAT exhibit osteopenia. The phenotype of FIAT knockout mice is still under evaluation but the salient aspects are discussed here. Taken together, the results accumulated to date support the hypothesis that FIAT is a transcriptional repressor that modulates osteoblast function.
Collapse
Affiliation(s)
- René St-Arnaud
- Genetics Unit, Shriners Hospital for Children, Montreal, Quebec, Canada H3G 1A6.
| | | |
Collapse
|
132
|
Drummond MJ, Glynn EL, Fry CS, Timmerman KL, Volpi E, Rasmussen BB. An increase in essential amino acid availability upregulates amino acid transporter expression in human skeletal muscle. Am J Physiol Endocrinol Metab 2010; 298:E1011-8. [PMID: 20304764 PMCID: PMC2867366 DOI: 10.1152/ajpendo.00690.2009] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Essential amino acids (EAA) stimulate skeletal muscle mammalian target of rapamycin complex 1 (mTORC1) signaling and protein synthesis. It has recently been reported that an increase in amino acid (AA) transporter expression during anabolic conditions is rapamycin-sensitive. The purpose of this study was to determine whether an increase in EAA availability increases AA transporter expression in human skeletal muscle. Muscle biopsies were obtained from the vastus lateralis of seven young adult subjects (3 male, 4 female) before and 1-3 h after EAA ingestion (10 g). Blood and muscle samples were analyzed for leucine kinetics using stable isotopic techniques. Quantitative RT-PCR, and immunoblotting were used to determine the mRNA and protein expression, respectively, of AA transporters and members of the general AA control pathway [general control nonrepressed (GCN2), activating transcription factor (ATF4), and eukaryotic initiation factor (eIF2) alpha-subunit (Ser(52))]. EAA ingestion increased blood leucine concentration, delivery of leucine to muscle, transport of leucine from blood into muscle, intracellular muscle leucine concentration, ribosomal protein S6 (Ser(240/244)) phosphorylation, and muscle protein synthesis. This was followed with increased L-type AA transporter (LAT1), CD98, sodium-coupled neutral AA transporter (SNAT2), and proton-coupled amino acid transporter (PAT1) mRNA expression at 1 h (P < 0.05) and modest increases in LAT1 protein expression (3 h post-EAA) and SNAT2 protein expression (2 and 3 h post-EAA, P < 0.05). Although there were no changes in GCN2 expression and eIF2 alpha phosphorylation, ATF4 protein expression reached significance by 2 h post-EAA (P < 0.05). We conclude that an increase in EAA availability upregulates human skeletal muscle AA transporter expression, perhaps in an mTORC1-dependent manner, which may be an adaptive response necessary for improved AA intracellular delivery.
Collapse
Affiliation(s)
- Micah J Drummond
- University of Texas Medical Branch, Department of Physical Therapy, Division of Rehabilitation Sciences, Sealy Center on Aging, 301 Univ. Blvd., Galveston, TX 77555-1144, USA
| | | | | | | | | | | |
Collapse
|
133
|
Wottawa M, Köditz J, Katschinski DM. Normoxic destabilization of ATF-4 depends on proteasomal degradation. Acta Physiol (Oxf) 2010; 198:457-63. [PMID: 19922526 DOI: 10.1111/j.1748-1716.2009.02060.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
AIM Hypoxia-inducible gene expression is an important physiological adaptive mechanism in response to a decreased oxygen supply. We have recently described an oxygen- and prolyl-4-hydroxylase (PHD)3-dependent stabilization of the activating transcription factor 4 (ATF-4). The aim of the present study was to examine if the normoxic destabilization of ATF-4 is regulated by oxygen-dependent proteasomal degradation. METHODS We determined poly-ubiquitination of ATF-4 in normoxia compared to hypoxia by immunoprecipitation and immunoblots. Furthermore, we analysed the expression of the ATF-4 target gene GADD153 as a function of oxygen concentration. RESULTS ATF-4 protein levels were not detectable in normoxia. Normoxic degradation correlated with an oxygen-dependent poly-ubiquitination of ATF-4, which was hindered by hypoxic incubation of the cells. As a result of hypoxia, GADD153 was expressed. The hypoxic GADD153 expression was attenuated or increased by transfecting the cells with ATF-4 siRNA or PHD3 siRNA respectively. CONCLUSION Our results demonstrate the involvement of oxygen-dependent proteasomal degradation of ATF-4 in the hypoxia-induced expression of GADD153. Taken together, hypoxia/PHD3-regulated stabilization of ATF-4 by hindering oxygen-dependent degradation may play a critical role in linking cell fate decisions to oxygen availability.
Collapse
Affiliation(s)
- M Wottawa
- Department of Cardiovascular Physiology, Georg-August University Göttingen, Germany
| | | | | |
Collapse
|
134
|
Abstract
Posttranslational modifications can cause profound changes in protein function. Typically, these modifications are reversible, and thus provide a biochemical on-off switch. In contrast, proline residues are the substrates for an irreversible reaction that is the most common posttranslational modification in humans. This reaction, which is catalyzed by prolyl 4-hydroxylase (P4H), yields (2S,4R)-4-hydroxyproline (Hyp). The protein substrates for P4Hs are diverse. Likewise, the biological consequences of prolyl hydroxylation vary widely, and include altering protein conformation and protein-protein interactions, and enabling further modification. The best known role for Hyp is in stabilizing the collagen triple helix. Hyp is also found in proteins with collagen-like domains, as well as elastin, conotoxins, and argonaute 2. A prolyl hydroxylase domain protein acts on the hypoxia inducible factor alpha, which plays a key role in sensing molecular oxygen, and could act on inhibitory kappaB kinase and RNA polymerase II. P4Hs are not unique to animals, being found in plants and microbes as well. Here, we review the enzymic catalysts of prolyl hydroxylation, along with the chemical and biochemical consequences of this subtle but abundant posttranslational modification.
Collapse
Affiliation(s)
- Kelly L. Gorres
- Department of Biochemistry, University of Wisconsin–Madison, Madison, WI 53706, U.S.A
| | - Ronald T. Raines
- Department of Biochemistry, University of Wisconsin–Madison, Madison, WI 53706, U.S.A
- Department of Chemistry, University of Wisconsin–Madison, Madison, WI 53706, U.S.A
| |
Collapse
|
135
|
Brown MK, Naidoo N. The UPR and the anti-oxidant response: relevance to sleep and sleep loss. Mol Neurobiol 2010; 42:103-13. [PMID: 20237865 DOI: 10.1007/s12035-010-8114-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Accepted: 03/01/2010] [Indexed: 12/21/2022]
Abstract
Oxidative stress has been linked to various physiological and pathological processes such as aging and neurological disorders. Recent evidence has now implicated a role for oxidative stress in sleep and sleep loss. Studies suggest that wakefulness results in an oxidative burden and sleep provides a protective mechanism against these harmful effects. Prolonged wakefulness/sleep deprivation activates an adaptive stress pathway termed the unfolded protein response (UPR), which temporarily guards against the deleterious consequences of reactive oxygen species. The UPR affects the function of the endoplasmic reticulum, which is the site for integral and secretory membrane processing and folding. Several downstream effectors of the UPR operate in an antioxidant capacity to reduce the load of these toxic species; a process that may be important in delaying the progression of neurodegenerative diseases. This review will highlight the molecular components of the UPR that ameliorate the accumulation of oxidative stress and may therefore provide potential therapeutic targets.
Collapse
Affiliation(s)
- Marishka K Brown
- Center for Sleep and Respiratory Neurobiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | |
Collapse
|
136
|
Jin HO, Seo SK, Woo SH, Choe TB, Hong SI, Kim JI, Park IC. Nuclear protein 1 induced by ATF4 in response to various stressors acts as a positive regulator on the transcriptional activation of ATF4. IUBMB Life 2010; 61:1153-8. [PMID: 19946894 DOI: 10.1002/iub.271] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Nuclear protein 1 (NUPR1) was originally identified as p8, a member of the family of HMG-I/Y transcription factors induced in response to various cellular stressors. However, the signaling pathway underlying NUPR1 induction by cellular stresses remains to be established. In this study, we found that the expression of NUPR1 by various stresses induced by activating transcription factor 4 (ATF4). Loss of ATF4 using siRNA significantly diminished NUPR1 expression. Overexpression of ATF4 caused NUPR1 levels to rise. NUPR1 expression was associated with enhanced transcriptional activation of genes of ATF4 downstream, suggesting that the protein promoted the transcription of stress-regulated genes via positive feedback on the ATF4 pathway.
Collapse
Affiliation(s)
- Hyeon-Ok Jin
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
137
|
Mangeat B, Gers-Huber G, Lehmann M, Zufferey M, Luban J, Piguet V. HIV-1 Vpu neutralizes the antiviral factor Tetherin/BST-2 by binding it and directing its beta-TrCP2-dependent degradation. PLoS Pathog 2009; 5:e1000574. [PMID: 19730691 PMCID: PMC2729927 DOI: 10.1371/journal.ppat.1000574] [Citation(s) in RCA: 240] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2009] [Accepted: 08/11/2009] [Indexed: 12/19/2022] Open
Abstract
Host cells impose a broad range of obstacles to the replication of retroviruses. Tetherin (also known as CD317, BST-2 or HM1.24) impedes viral release by retaining newly budded HIV-1 virions on the surface of cells. HIV-1 Vpu efficiently counteracts this restriction. Here, we show that HIV-1 Vpu induces the depletion of tetherin from cells. We demonstrate that this phenomenon correlates with the ability of Vpu to counteract the antiviral activity of both overexpressed and interferon-induced endogenous tetherin. In addition, we show that Vpu co-immunoprecipitates with tetherin and beta-TrCP in a tri-molecular complex. This interaction leads to Vpu-mediated proteasomal degradation of tetherin in a beta-TrCP2-dependent manner. Accordingly, in conditions where Vpu-beta-TrCP2-tetherin interplay was not operative, including cells stably knocked down for beta-TrCP2 expression or cells expressing a dominant negative form of beta-TrCP, the ability of Vpu to antagonize the antiviral activity of tetherin was severely impaired. Nevertheless, tetherin degradation did not account for the totality of Vpu-mediated counteraction against the antiviral factor, as binding of Vpu to tetherin was sufficient for a partial relief of the restriction. Finally, we show that the mechanism used by Vpu to induce tetherin depletion implicates the cellular ER-associated degradation (ERAD) pathway, which mediates the dislocation of ER membrane proteins into the cytosol for subsequent proteasomal degradation. In conclusion, we show that Vpu interacts with tetherin to direct its beta-TrCP2-dependent proteasomal degradation, thereby alleviating the blockade to the release of infectious virions. Identification of tetherin binding to Vpu provides a potential novel target for the development of drugs aimed at inhibiting HIV-1 replication.
Collapse
Affiliation(s)
- Bastien Mangeat
- Department of Dermatology and Venereology, University Hospitals and Medical School of Geneva, University of Geneva, Switzerland
- Department of Microbiology and Molecular Medicine, University Hospitals and Medical School of Geneva, University of Geneva, Switzerland
| | - Gustavo Gers-Huber
- Department of Dermatology and Venereology, University Hospitals and Medical School of Geneva, University of Geneva, Switzerland
- Department of Microbiology and Molecular Medicine, University Hospitals and Medical School of Geneva, University of Geneva, Switzerland
| | - Martin Lehmann
- Department of Dermatology and Venereology, University Hospitals and Medical School of Geneva, University of Geneva, Switzerland
- Department of Microbiology and Molecular Medicine, University Hospitals and Medical School of Geneva, University of Geneva, Switzerland
| | - Madeleine Zufferey
- Department of Microbiology and Molecular Medicine, University Hospitals and Medical School of Geneva, University of Geneva, Switzerland
| | - Jeremy Luban
- Department of Microbiology and Molecular Medicine, University Hospitals and Medical School of Geneva, University of Geneva, Switzerland
| | - Vincent Piguet
- Department of Dermatology and Venereology, University Hospitals and Medical School of Geneva, University of Geneva, Switzerland
- Department of Microbiology and Molecular Medicine, University Hospitals and Medical School of Geneva, University of Geneva, Switzerland
- * E-mail:
| |
Collapse
|
138
|
Kimbrel EA, Kung AL. The F-box protein beta-TrCp1/Fbw1a interacts with p300 to enhance beta-catenin transcriptional activity. J Biol Chem 2009; 284:13033-44. [PMID: 19297328 DOI: 10.1074/jbc.m901248200] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Hyperactivated beta-catenin is a commonly found molecular abnormality in colon cancer, and its nuclear accumulation is thought to promote the expression of genes associated with cellular proliferation and transformation. The p300 transcriptional co-activator binds to beta-catenin and facilitates transcription by recruiting chromatin remodeling complexes and general transcriptional apparatus. We have found that beta-TrCp1/Fbw1a, a member of the Skp1/Cullin/Rbx1/F-box E3 ubiquitin ligase complex, binds directly to p300 and co-localizes with it to beta-catenin target gene promoters. Our data show that Fbw1a, which normally targets beta-catenin for degradation, works together with p300 to enhance the transcriptional activity of beta-catenin, whereas other F-box/WD40 proteins do not. Fbw1a also cooperates with p300 to co-activate transcription by SMAD3, another Fbw1a ubiquitylation target, but not p53 or HIF-1alpha, which are substrates for other ubiquitin ligase complexes. These results suggest that, although Fbw1a is part of a negative feedback loop for controlling beta-catenin levels in normal cells, its overexpression and binding to p300 may contribute to hyperactivated beta-catenin transcriptional activity in colon cancer cells.
Collapse
Affiliation(s)
- Erin A Kimbrel
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Children's Hospital Boston and Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
139
|
Cadmium interferes with the degradation of ATF5 via a post-ubiquitination step of the proteasome degradation pathway. Biochem Biophys Res Commun 2009; 380:673-8. [PMID: 19285020 DOI: 10.1016/j.bbrc.2009.01.158] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2009] [Accepted: 01/26/2009] [Indexed: 11/21/2022]
Abstract
ATF5 is a member of the CREB/ATF family of transcription factors. In the current study, using a transient transfection system to express FLAG epitope fusion proteins of ATF5, we have shown that CdCl(2) or NaAsO(3) increases the protein levels of ATF5 in cells, and that cadmium stabilizes the ATF5 protein. Proteasome inhibitors had a similar effect to cadmium on the cellular accumulation of ATF5. Proteasome inhibition led to an increase in ubiquitinated ATF5, while cadmium did not appear to reduce the extent of ATF5 ubiquitination. ATF5 contains a putative nuclear export signal within its N-terminus. We demonstrated that whereas deletion of N-terminal region resulted in a increase of ATF5 levels, this region does not appear to be involved in the ubiquitination of ATF5. These results indicate that ATF5 is targeted for degradation by the ubiquitin-proteasome pathway, and that cadmium slows the rate of ATF5 degradation via a post-ubiquitination mechanism.
Collapse
|
140
|
Seo E, Kim H, Kim R, Yun S, Kim M, Han JK, Costantini F, Jho EH. Multiple isoforms of beta-TrCP display differential activities in the regulation of Wnt signaling. Cell Signal 2009; 21:43-51. [PMID: 18929646 PMCID: PMC2645026 DOI: 10.1016/j.cellsig.2008.09.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2008] [Accepted: 09/09/2008] [Indexed: 10/21/2022]
Abstract
The F-box proteins beta-TrCP1 and 2 (beta-transducin repeat containing protein) have 2 and 3 isoforms, respectively, due to alternative splicing of exons encoding the N-terminal region. We identified an extra exon in between the previously known exons 1 and 2 of beta-TrCP1 and beta-TrCP2. Interestingly, sequence analysis suggested that many more isoforms are produced than previously identified, via the alternative splicing of all possible combination of exons II to V of beta-TrCP1 and exons II to IV of beta-TrCP2. Different mouse tissues show specific expression patterns of the isoforms, and the level of expression of the isoform that has been used in most published papers was very low. Yeast two-hybrid assays show that beta-TrCP1 isoforms containing exon III, which are the most highly expressed isoforms in most tissues, do not interact with Skp1. Indirect immunofluorescence analysis of transiently expressed beta-TrCP1 isoforms suggests that the presence of exon III causes beta-TrCP1 to localize in nuclei. Consistent with the above findings, isoforms including exon III showed a reduced ability to block ectopic embryonic axes induced via injection of Wnt8 or beta-catenin in Xenopus embryos. Overall, our data suggest that isoforms of beta-TrCPs generated by alternative splicing may have different biological roles.
Collapse
Affiliation(s)
- Eunjeong Seo
- Department of Life Science, The University of Seoul, Seoul, Korea, 130-743
| | - Hyunjoon Kim
- Division of Molecular and Life Sciences, Pohang University of Science and Technology, San 31, Hyoja-dong, Pohang, Kyungbuk, Korea, 790-784
| | - Rokki Kim
- Department of Life Science, The University of Seoul, Seoul, Korea, 130-743
| | - Sangmoon Yun
- Department of Life Science, The University of Seoul, Seoul, Korea, 130-743
| | - Minseong Kim
- Department of Life Science, The University of Seoul, Seoul, Korea, 130-743
| | - Jin-Kwan Han
- Division of Molecular and Life Sciences, Pohang University of Science and Technology, San 31, Hyoja-dong, Pohang, Kyungbuk, Korea, 790-784
| | - Frank Costantini
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Eek-hoon Jho
- Department of Life Science, The University of Seoul, Seoul, Korea, 130-743
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
141
|
Qi M, Lei T, Zhou L, Chen XD, Long H, Long QQ, Zhang RR, Yang ZQ, Gan L. Cloning, characterization, chromosomal mapping and tissue transcription analysis of porcine CREB2 and CREB3 genes. Folia Biol (Praha) 2009; 55:137-144. [PMID: 19691921 DOI: 10.14712/fb2009055040137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
CREB2 and CREB3 are two important members of the ATF/CREB family, which negatively and positively regulates CRE-dependent transcription in vitro. Here we report the cloning, chromosome mapping and tissue transcription analysis of CREB2 and CREB3 in pigs. The full-length coding sequence of CREB2 and CREB3 is 1047 bp and 1098 bp, encoding 348 and 365 amino acids, respectively. Porcine CREB3 comprises nine exons and eight introns, whereas CREB2 consists of three exons and two introns. CREB2 and CREB3 were cytogenetically assigned to porcine chromosome 5p and 1q28, respectively. Tissue transcription analysis revealed that both porcine CREB2 and CREB3 mRNA were ubiquitously detected in all examined tissues. Additionally, we cloned the 5' flank genomic sequence of porcine CREB3 and characterized several putative transcription factor recognition sites including SP1, NF-kappaB, AP-1 and AP-2 in its promoter region. Our studies provide basic molecular information helpful for further investigation of the function of the two genes in pig models.
Collapse
Affiliation(s)
- M Qi
- College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Abstract
The maintenance and preservation of distinct phases during the cell cycle is a highly complex and coordinated process. It is regulated by phosphorylation--through the activity of cyclin-dependent kinases (CDKs)--and protein degradation, which occurs through ubiquitin ligases such as SCF (SKP1-CUL1-F-box protein) complexes and APC/C (anaphase-promoting complex/cyclosome). Here, we explore the functionality and biology of the F-box proteins, SKP2 (S-phase kinase-associated protein 2) and beta-TrCP (beta-transducin repeat-containing protein), which are emerging as important players in cancer biogenesis owing to the deregulated proteolysis of their substrates.
Collapse
Affiliation(s)
- David Frescas
- Department of Pathology, NYU Cancer Institute, New York University School of Medicine, 550 First Avenue, New York, New York 10016, USA
| | | |
Collapse
|
143
|
Malmberg SE, Adams CM. Insulin signaling and the general amino acid control response. Two distinct pathways to amino acid synthesis and uptake. J Biol Chem 2008; 283:19229-34. [PMID: 18480057 DOI: 10.1074/jbc.m801331200] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
ATF4 is a transcription factor that induces a genetic program for amino acid synthesis and amino acid uptake. Previous work demonstrated that ATF4 expression is increased either by insulin or by the general amino acid control (GAAC) response, an evolutionarily ancient pathway that is activated when eukaryotic cells are deprived of amino acids. It is not known whether insulin and the GAAC pathway increase ATF4 expression by the same or different mechanisms. In these studies, we demonstrate that insulin-mediated ATF4 expression occurs as part of a coordinated anabolic program that does not require an essential component of the GAAC pathway, the protein kinase GCN2. Moreover, insulin and the GAAC pathway have an additive effect on expression of ATF4 and downstream mRNAs for amino acid synthesis and uptake. These data suggest that the GAAC pathway may facilitate insulin-mediated anabolism when exogenous amino acids are limiting. We conclude that insulin signaling and the GAAC response comprise two distinct yet complimentary pathways to ATF4 expression, allowing anabolism to be finely tuned to amino acid availability.
Collapse
Affiliation(s)
- Sharon E Malmberg
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA
| | | |
Collapse
|
144
|
Lange PS, Chavez JC, Pinto JT, Coppola G, Sun CW, Townes TM, Geschwind DH, Ratan RR. ATF4 is an oxidative stress-inducible, prodeath transcription factor in neurons in vitro and in vivo. ACTA ACUST UNITED AC 2008; 205:1227-42. [PMID: 18458112 PMCID: PMC2373852 DOI: 10.1084/jem.20071460] [Citation(s) in RCA: 217] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Oxidative stress is pathogenic in neurological diseases, including stroke. The identity of oxidative stress–inducible transcription factors and their role in propagating the death cascade are not well known. In an in vitro model of oxidative stress, the expression of the bZip transcription factor activating transcription factor 4 (ATF4) was induced by glutathione depletion and localized to the promoter of a putative death gene in neurons. Germline deletion of ATF4 resulted in a profound reduction in oxidative stress–induced gene expression and resistance to oxidative death. In neurons, ATF4 modulates an early, upstream event in the death pathway, as resistance to oxidative death by ATF4 deletion was associated with decreased consumption of the antioxidant glutathione. Forced expression of ATF4 was sufficient to promote cell death and loss of glutathione. In ATF4−/− neurons, restoration of ATF4 protein expression reinstated sensitivity to oxidative death. In addition, ATF4−/− mice experienced significantly smaller infarcts and improved behavioral recovery as compared with wild-type mice subjected to the same reductions in blood flow in a rodent model of ischemic stroke. Collectively, these findings establish ATF4 as a redox-regulated, prodeath transcriptional activator in the nervous system that propagates death responses to oxidative stress in vitro and to stroke in vivo.
Collapse
Affiliation(s)
- Philipp S Lange
- Burke Medical Research Institute, White Plains, NY 10605, USA.
| | | | | | | | | | | | | | | |
Collapse
|
145
|
Dong C, Upadhya SC, Ding L, Smith TK, Hegde AN. Proteasome inhibition enhances the induction and impairs the maintenance of late-phase long-term potentiation. Learn Mem 2008; 15:335-47. [PMID: 18441292 PMCID: PMC2364605 DOI: 10.1101/lm.984508] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2008] [Accepted: 03/20/2008] [Indexed: 01/21/2023]
Abstract
Protein degradation by the ubiquitin-proteasome pathway plays important roles in synaptic plasticity, but the molecular mechanisms by which proteolysis regulates synaptic strength are not well understood. We investigated the role of the proteasome in hippocampal late-phase long-term potentiation (L-LTP), a model for enduring synaptic plasticity. We show here that inhibition of the proteasome enhances the induction of L-LTP, but inhibits its maintenance. Proteasome inhibitor-mediated enhancement of the early part of L-LTP requires activation of NMDA receptors and the cAMP-dependent protein kinase. Augmentation of L-LTP induction by proteasome inhibition is blocked by a protein synthesis inhibitor anisomycin and is sensitive to the drug rapamycin. Our findings indicate that proteasome inhibition increases the induction of L-LTP by stabilizing locally translated proteins in dendrites. In addition, our data show that inhibition of the proteasome blocks transcription of brain-derived neurotrophic factor (BDNF), which is a cAMP-responsive element-binding protein (CREB)-inducible gene. Furthermore, our results demonstrate that the proteasome inhibitors block degradation of ATF4, a CREB repressor. Thus, proteasome inhibition appears to hinder CREB-mediated transcription. Our results indicate that blockade of proteasome activity obstructs the maintenance of L-LTP by interfering with transcription as well as translation required to sustain L-LTP. Thus, proteasome-mediated proteolysis has different roles during the induction and the maintenance of L-LTP.
Collapse
Affiliation(s)
- Chenghai Dong
- Department of Neurobiology and Anatomy, Wake Forest University Health Sciences, Winston-Salem, North Carolina 27157, USA
| | - Sudarshan C. Upadhya
- Department of Neurobiology and Anatomy, Wake Forest University Health Sciences, Winston-Salem, North Carolina 27157, USA
| | - Lan Ding
- Department of Neurobiology and Anatomy, Wake Forest University Health Sciences, Winston-Salem, North Carolina 27157, USA
| | - Thuy K. Smith
- Department of Neurobiology and Anatomy, Wake Forest University Health Sciences, Winston-Salem, North Carolina 27157, USA
| | - Ashok N. Hegde
- Department of Neurobiology and Anatomy, Wake Forest University Health Sciences, Winston-Salem, North Carolina 27157, USA
| |
Collapse
|
146
|
Yu S, Jiang Y, Galson DL, Luo M, Lai Y, Lu Y, Ouyang HJ, Zhang J, Xiao G. General transcription factor IIA-gamma increases osteoblast-specific osteocalcin gene expression via activating transcription factor 4 and runt-related transcription factor 2. J Biol Chem 2008; 283:5542-53. [PMID: 18171674 PMCID: PMC2736298 DOI: 10.1074/jbc.m705653200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
ATF4 (activating transcription factor 4) is an osteoblast-enriched transcription factor that regulates terminal osteoblast differentiation and bone formation. ATF4 knock-out mice have reduced bone mass (severe osteoporosis) throughout life. Runx2 (runt-related transcription factor 2) is a runt domain-containing transcription factor that is essential for bone formation during embryogenesis and postnatal life. In this study, we identified general transcription factor IIA gamma (TFIIA gamma) as a Runx2-interacting factor in a yeast two-hybrid screen. Immunoprecipitation assays confirmed that TFIIA gamma interacts with Runx2 in osteoblasts and when coexpressed in COS-7 cells or using purified glutathione S-transferase fusion proteins. Chromatin immunoprecipitation assay of MC3T3-E1 (clone MC-4) preosteoblast cells showed that in intact cells TFIIA gamma is recruited to the region of the osteocalcin promoter previously shown to bind Runx2 and ATF4. A small region of Runx2 (amino acids 258-286) was found to be required for TFIIA gamma binding. Although TFIIA gamma interacts with Runx2, it does not activate Runx2. Instead, TFIIA gamma binds to and activates ATF4. Furthermore, TFIIA gamma together with ATF4 and Runx2 stimulates osteocalcin promoter activity and endogenous mRNA expression. Small interfering RNA silencing of TFIIA gamma markedly reduces levels of endogenous ATF4 protein and Ocn mRNA in osteoblastic cells. Overexpression of TFIIA gamma increases levels of ATF4 protein. Finally, TFIIA gamma significantly prevents ATF4 degradation. This study shows that a general transcription factor, TFIIA gamma, facilitates osteoblast-specific gene expression through interactions with two important bone transcription factors ATF4 and Runx2.
Collapse
Affiliation(s)
- Shibing Yu
- Department of Medicine University of Pittsburgh, Pittsburgh, Pennsylvania 15240
| | - Yu Jiang
- Department of Pharmacology, University of Pittsburgh, Pittsburgh, Pennsylvania 15240
| | - Deborah L. Galson
- Department of Medicine University of Pittsburgh, Pittsburgh, Pennsylvania 15240
| | - Min Luo
- Department of Medicine University of Pittsburgh, Pittsburgh, Pennsylvania 15240
| | - Yumei Lai
- Department of Medicine University of Pittsburgh, Pittsburgh, Pennsylvania 15240
| | - Yi Lu
- Department of Medicine University of Pittsburgh, Pittsburgh, Pennsylvania 15240
| | - Hong-Jiao Ouyang
- Department of Medicine University of Pittsburgh, Pittsburgh, Pennsylvania 15240
| | - Jian Zhang
- Department of Medicine University of Pittsburgh, Pittsburgh, Pennsylvania 15240
| | - Guozhi Xiao
- Department of Medicine University of Pittsburgh, Pittsburgh, Pennsylvania 15240
| |
Collapse
|
147
|
Abstract
Cellular stresses that disrupt the processing of proteins slated for the secretory pathway induce the unfolded protein response (UPR), a regulatory network involving both translational and transcriptional control mechanisms that is designed to expand the secretory pathway and alleviate cellular injury. PERK (PEK/EIF2AK3) mediates the translational control arm of the UPR by enhancing phosphorylation of eIF2. Phosphorylation of eIF2 reduces global protein synthesis, preventing further overload of the secretory pathway and allowing the cell to direct a new pattern of mRNA synthesis that enhances the processing capacity of the endoplasmic reticulum (ER). PERK also directs preferential translation of stress-related transcripts, including that encoding ATF4, a transcriptional activator that contributes to the UPR. Reduced global translation also leads to reduced levels of key regulatory proteins that are subject to rapid turnover, facilitating activation of transcription factors such as NF-B during cellular stress. This review highlights the mechanisms by which PERK monitors and is activated by accumulated misfolded protein in the ER, the processes by which PERK regulates both general and gene-specific translation that is central for the UPR, and the role of PERK in the process of cellular adaptation to ER stress and its impact in disease.
Collapse
Affiliation(s)
- Ronald C Wek
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.
| | | |
Collapse
|
148
|
Pons J, Evrard-Todeschi N, Bertho G, Gharbi-Benarous J, Benarous R, Girault JP. Phosphorylation-dependent structure of ATF4 peptides derived from a human ATF4 protein, a member of the family of transcription factors. Peptides 2007; 28:2253-67. [PMID: 17996332 DOI: 10.1016/j.peptides.2007.09.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Revised: 09/14/2007] [Accepted: 09/17/2007] [Indexed: 11/21/2022]
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.
Collapse
Affiliation(s)
- Julien Pons
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, Université Paris Descartes, UMR 8601 CNRS, 45 rue des Saint-Pères, 75270 Paris Cedex 06, France
| | | | | | | | | | | |
Collapse
|
149
|
Köditz J, Nesper J, Wottawa M, Stiehl DP, Camenisch G, Franke C, Myllyharju J, Wenger RH, Katschinski DM. Oxygen-dependent ATF-4 stability is mediated by the PHD3 oxygen sensor. Blood 2007; 110:3610-7. [PMID: 17684156 DOI: 10.1182/blood-2007-06-094441] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
AbstractThe 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.
Collapse
Affiliation(s)
- Jens Köditz
- Department of Heart and Circulatory Physiology, Center of Physiology and Pathophysiology, Georg-August University Göttingen, Göttingen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Ord D, Meerits K, Ord T. TRB3 protects cells against the growth inhibitory and cytotoxic effect of ATF4. Exp Cell Res 2007; 313:3556-67. [PMID: 17707795 DOI: 10.1016/j.yexcr.2007.07.017] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Revised: 07/11/2007] [Accepted: 07/19/2007] [Indexed: 01/03/2023]
Abstract
Tribbles homolog 3 (TRB3) is a pseudokinase the level of which is increased in response to various stresses. We and other researchers have previously shown that TRB3 interacts with activating transcription factor 4 (ATF4) and may function as a negative feedback regulator of ATF4. In the present study, we investigate the effect of ATF4 and TRB3 on cell growth and viability, using both the enforced expression and silencing of the genes. HEK293 cells overexpressing ATF4 show retarded growth in the complete medium and decreased viability in the glucose-free medium. The enforced expression of ATF4 increases the level of reactive oxygen species (ROS) and the supplementation of the medium with ROS scavenging and reducing compounds supports the growth and survival of cells overexpressing ATF4. The deleterious effects of elevated ATF4 are suppressed by the coexpression of TRB3, which downregulates ATF4 transcriptional activity and results in the decrease of intracellular ROS. Also, the coexpression of TRB3 rescues postmitotic neuronally differentiated PC12 cells from the apoptosis evoked by ATF4 overexpression. The silencing of ATF4 and TRB3 genes by RNA interference reveals that endogenous ATF4 promotes and TRB3 suppresses the death of glucose-deprived SaOS2 cells. Together, the results indicate that TRB3 protects cells against the growth inhibitory and cytotoxic effect of ATF4.
Collapse
Affiliation(s)
- Daima Ord
- Institute of Molecular and Cell Biology, Tartu University, Tartu, Estonia; Estonian Biocentre, 23 Riia St, Tartu, Estonia
| | | | | |
Collapse
|