51
|
Polak P, Oren A, Ben-Dror I, Steinberg D, Sapoznik S, Arditi-Duvdevany A, Vardimon L. The cytoskeletal network controls c-Jun translation in a UTR-dependent manner. Oncogene 2006; 25:665-76. [PMID: 16247475 DOI: 10.1038/sj.onc.1209114] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The cytoskeleton is a dynamic network that undergoes restructuring during various cellular events, influencing cell proliferation, differentiation, and apoptosis. Here, we report that accumulation of c-Jun, a member of the AP1 family of transcription factors that play a key role in normal and aberrant cell growth, dramatically increases upon depolymerization of the cytoskeleton, and that, unexpectedly, this increase is controlled translationally. Depolymerization of the actin or microtubule network induces an increase in c-Jun accumulation with no corresponding increase in c-Jun mRNA or in the half-life of the c-Jun protein, but rather in the translatability of its transcript. This increase is mediated by the untranslated regions (UTRs) of c-Jun mRNA, and is not dependent on activated mitogen-activated protein kinase pathways. This novel mechanism of c-Jun regulation might be relevant to physiological conditions in which c-Jun plays a pivotal role.
Collapse
Affiliation(s)
- P Polak
- Department of Biochemistry, George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | | | |
Collapse
|
52
|
le Maire A, Schiltz M, Braud S, Gondry M, Charbonnier JB, Zinn-Justin S, Stura E. Crystallization and halide phasing of the C-terminal domain of human KIN17. Acta Crystallogr Sect F Struct Biol Cryst Commun 2006; 62:245-8. [PMID: 16511313 PMCID: PMC2197188 DOI: 10.1107/s174430910600409x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2005] [Accepted: 02/02/2006] [Indexed: 11/10/2022]
Abstract
Here, the crystallization and initial phasing of the C-terminal domain of human KIN17, a 45 kDa protein mainly expressed in response to ionizing radiation and overexpressed in certain tumour cell lines, are reported. Crystals diffracting to 1.4 A resolution were obtained from 10% ethylene glycol, 27% PEG 6000, 500 mM LiCl and 100 mM sodium acetate pH 6.3 in space group P2(1)2(1)2(1), with unit-cell parameters a = 45.75, b = 46.31, c = 60.80 A and one molecule in the asymmetric unit. Since this domain has a basic pI, heavy-atom derivatives were obtained by soaking the crystals with negatively charged ions such as tungstate and iodine. The replacement of LiCl by KI in the cryosolution allowed the determination of phases from iodide ions to give an interpretable electron-density map.
Collapse
Affiliation(s)
- Albane le Maire
- Département d'Ingénierie et d'Etude des Protéines, Commissariat a l'Energie Atomique, 91191 Gif-sur-Yvette, France.
| | | | | | | | | | | | | |
Collapse
|
53
|
Lipniacki T, Paszek P, Brasier AR, Luxon BA, Kimmel M. Stochastic regulation in early immune response. Biophys J 2006; 90:725-42. [PMID: 16284261 PMCID: PMC1367099 DOI: 10.1529/biophysj.104.056754] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2004] [Accepted: 09/12/2005] [Indexed: 11/18/2022] Open
Abstract
Living cells may be considered noisy or stochastic biochemical reactors. In eukaryotic cells, in which the number of protein or mRNA molecules is relatively large, the stochastic effects originate primarily in regulation of gene activity. Transcriptional activity of a gene can be initiated by transactivator molecules binding to the specific regulatory site(s) in the target gene. The stochasticity of activator binding and dissociation is amplified by transcription and translation, since target gene activation results in a burst of mRNAs molecules, and each copy of mRNA then serves as a template for numerous protein molecules. In this article, we reformulate our model of the NF-kappaB regulatory module to analyze a single cell regulation. Ordinary differential equations, used for description of fast reaction channels of processes involving a large number of molecules, are combined with a stochastic switch to account for the activity of the genes involved. The stochasticity in gene transcription causes simulated cells to exhibit large variability. Moreover, none of them behaves like an average cell. Although the average mRNA and protein levels remain constant before tumor necrosis factor (TNF) stimulation, and stabilize after a prolonged TNF stimulation, in any single cell these levels oscillate stochastically in the absence of TNF and keep oscillating under the prolonged TNF stimulation. However, in a short period of approximately 90 min, most cells are synchronized by the TNF signal, and exhibit similar kinetics. We hypothesize that this synchronization is crucial for proper activation of early genes controlling inflammation. Our theoretical predictions of single cell kinetics are supported by recent experimental studies of oscillations in NF-kappaB signaling made on single cells.
Collapse
Affiliation(s)
- Tomasz Lipniacki
- Institute of Fundamental Technological Research, Warsaw, Poland.
| | | | | | | | | |
Collapse
|
54
|
Canguilhem B, Pradines A, Baudouin C, Boby C, Lajoie-Mazenc I, Charveron M, Favre G. RhoB protects human keratinocytes from UVB-induced apoptosis through epidermal growth factor receptor signaling. J Biol Chem 2005; 280:43257-63. [PMID: 16278215 DOI: 10.1074/jbc.m508650200] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Exposure of the skin to UVB light results in the formation of DNA photolesions that can give rise to cell death, mutations, and the onset of carcinogenic events. Specific proteins are activated by UVB and then trigger signal transduction pathways that lead to cellular responses. An alteration of these signaling molecules is thought to be a fundamental event in tumor promotion by UVB irradiation. RhoB, encoding a small GTPase has been identified as a DNA damage-inducible gene. RhoB is involved in epidermal growth factor (EGF) receptor trafficking, cytoskeletal organization, cell transformation, and survival. We have analyzed the regulation of RhoB and elucidated its role in the cellular response of HaCaT keratinocytes to relevant environmental UVB irradiation. We report here that the activated GTP-bound form of RhoB is increased rapidly within 5 min of exposure to UVB, and then RhoB protein levels increased concomitantly with EGF receptor (EGFR) activation. Inhibition of UVB-induced EGFR activation prevents RhoB protein expression and AKT phosphorylation but not the early activation of RhoB. Blocking UVB-induced RhoB expression with specific small interfering RNAs inhibits AKT and glycogen synthase kinase-3beta phosphorylation through inhibition of EGFR expression. Moreover, down-regulation of RhoB potentiates UVB-induced cell apoptosis. In contrast, RhoB overexpression protects keratinocytes against UVB-induced apoptosis. These results indicated that RhoB is regulated upon UVB exposure by a two-step process consisting of an early EGFR-independent RhoB activation followed by an EGFR-dependent induction of RhoB expression. Moreover, we have demonstrated that RhoB is essential in regulating keratinocyte cell survival after UVB exposure, suggesting its potential role in photocarcinogenesis.
Collapse
Affiliation(s)
- Bruno Canguilhem
- INSERM U563, Département Innovation Thérapeutique et Oncologie Moléculaire, Institut Claudius Regaud, Université Paul Sabatier, 20/24 rue du Pont Saint-Pierre, 31052 Toulouse Cedex France
| | | | | | | | | | | | | |
Collapse
|
55
|
Hamdi M, Kool J, Cornelissen-Steijger P, Carlotti F, Popeijus HE, van der Burgt C, Janssen JM, Yasui A, Hoeben RC, Terleth C, Mullenders LH, van Dam H. DNA damage in transcribed genes induces apoptosis via the JNK pathway and the JNK-phosphatase MKP-1. Oncogene 2005; 24:7135-44. [PMID: 16044158 DOI: 10.1038/sj.onc.1208875] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The nucleotide excision repair (NER) system consists of two sub-pathways, global genome repair (GGR) and transcription-coupled repair (TCR), which exhibit distinct functions in the cellular response to genotoxic stress. Defects in TCR result in prolonged UV light-induced stalling of RNA polymerase II and hypersensitivity to apoptosis induced by UV and certain chemotherapeutic drugs. Here, we show that low doses of UV trigger delayed activation of the stress-induced MAPkinase JNK and its proapoptotic targets c-Jun and ATF-3 in TCR-deficient primary human fibroblasts from Xeroderma Pigmentosum (XP) and Cockayne syndrome (CS) patients. This delayed activation of the JNK pathway is not observed in GGR-deficient TCR-proficient XP cells, is independent of functional p53, and is established through repression of the JNK-phosphatase MKP-1 rather than by activation of the JNK kinases MKK4 and 7. Enzymatic reversal of UV-induced cyclobutane pyrimidine dimers (CPDs) by CPD photolyase abrogated JNK activation, MKP-1 repression, and apoptosis in TCR-deficient XPA cells. Ectopic expression of MKP-1 inhibited DNA-damage-induced JNK activity and apoptosis. These results identify both MKP-1 and JNK as sensors and downstream effectors of persistent DNA damage in transcribed genes and suggest a link between the JNK pathway and UV-induced stalling of RNApol II.
Collapse
Affiliation(s)
- Mohamed Hamdi
- Department of Molecular Cell Biology, Leiden University Medical Center, Wassenaarseweg 72, 2333AL Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Miccoli L, Frouin I, Novac O, Di Paola D, Harper F, Zannis-Hadjopoulos M, Maga G, Biard DSF, Angulo JF. The human stress-activated protein kin17 belongs to the multiprotein DNA replication complex and associates in vivo with mammalian replication origins. Mol Cell Biol 2005; 25:3814-30. [PMID: 15831485 PMCID: PMC1084281 DOI: 10.1128/mcb.25.9.3814-3830.2005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The human stress-activated protein kin17 accumulates in the nuclei of proliferating cells with predominant colocalization with sites of active DNA replication. The distribution of kin17 protein is in equilibrium between chromatin-DNA and the nuclear matrix. An increased association with nonchromatin nuclear structure is observed in S-phase cells. We demonstrated here that kin17 protein strongly associates in vivo with DNA fragments containing replication origins in both human HeLa and monkey CV-1 cells. This association was 10-fold higher than that observed with nonorigin control DNA fragments in exponentially growing cells. In addition, the association of kin17 protein to DNA fragments containing replication origins was also analyzed as a function of the cell cycle. High binding of kin17 protein was found at the G(1)/S border and throughout the S phase and was negligible in both G(0) and M phases. Specific monoclonal antibodies against kin17 protein induced a threefold inhibition of in vitro DNA replication of a plasmid containing a minimal replication origin that could be partially restored by the addition of recombinant kin17 protein. Immunoelectron microscopy confirmed the colocalization of kin17 protein with replication proteins like RPA, PCNA, and DNA polymerase alpha. A two-step chromatographic fractionation of nuclear extracts from HeLa cells revealed that kin17 protein localized in vivo in distinct protein complexes of high molecular weight. We found that kin17 protein purified within an approximately 600-kDa protein complex able to support in vitro DNA replication by means of two different biochemical methods designed to isolate replication complexes. In addition, the reduced in vitro DNA replication activity of the multiprotein replication complex after immunodepletion for kin17 protein highlighted for a direct role in DNA replication at the origins.
Collapse
Affiliation(s)
- Laurent Miccoli
- Commissariat à l'Energie Atomique, Centre de Fontenay-aux-Roses, LGR/DRR/DSV, BP6, 92265 Fontenay-aux-Roses Cedex, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
57
|
Abstract
Every living organism on the surface of the earth is exposed to the ultraviolet (UV) fraction of the sunlight. This electromagnetic energy has both life-giving and life-endangering effects. UV radiation can damage DNA and thus mutagenize several genes involved in the development of the skin cancer. The presence of typical signature of UV-induced mutations on these genes indicates that the ultraviolet-B part of sunlight is responsible for the evolution of cutaneous carcinogenesis. During this process, variable alterations of the oncogenic, tumor-suppressive, and cell-cycle control signaling pathways occur. These pathways include (a) mutated PTCH (in the mitogenic Sonic Hedgehog pathway) and mutated p53 tumor-suppressor gene in basal cell carcinomas, (b) an activated mitogenic ras pathway and mutated p53 in squamous cell carcinomas, and (c) an activated ras pathway, inactive p16, and p53 tumor suppressors in melanomas. This review presents background information about the skin optics, UV radiation, and molecular events involved in photocarcinogenesis.
Collapse
Affiliation(s)
- Mahmoud R Hussein
- Pathology department, Assuit University Hospitals, Assuit University, Assuit, Egypt.
| |
Collapse
|
58
|
Pinon-Lataillade G, Masson C, Bernardino-Sgherri J, Henriot V, Mauffrey P, Frobert Y, Araneda S, Angulo JF. KIN17 encodes an RNA-binding protein and is expressed during mouse spermatogenesis. J Cell Sci 2005; 117:3691-702. [PMID: 15252136 DOI: 10.1242/jcs.01226] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Genotoxic agents deform DNA structure thus eliciting a complex genetic response allowing recovery and cell survival. The Kin17 gene is up-regulated during this response. This gene encodes a conserved nuclear protein that shares a DNA-binding domain with the bacterial RecA protein. The KIN17 protein binds DNA and displays enhanced expression levels in proliferating cultured cells, suggesting a role in nuclear metabolism. We investigated this by studying the expression profile of KIN17 protein during mouse spermatogenesis. As expected, the expression level of Kin17 is higher in proliferating than in differentiated cells. KIN17 is selectively extracted from this tissue by detergents and a fraction was tightly associated with the nuclear matrix. Germinal cells ubiquitously express Kin17 and the protein is located mainly in the nucleus except in elongated spermatids where cytoplasmic staining is also observed. Sertoli and germ cells that are no longer mitotically active express KIN17, suggesting a general role in all testicular cell types. In adult testis a significant proportion of KIN17 co-purifies with polyadenylated RNA. KIN17 directly binds RNA, preferentially poly(G) and poly(U) homopolymers. These results together with the identification of KIN17 as a component of the human spliceosome indicate that this protein may participate in RNA processing.
Collapse
Affiliation(s)
- Ghislaine Pinon-Lataillade
- Laboratoire de Génétique de la Radiosensibilité, CEA, Direction des Sciences du Vivant, Département de Radiobiologie et de Radiopathologie, B.P. 6, 92265 Fontenay aux Roses CEDEX, France.
| | | | | | | | | | | | | | | |
Collapse
|
59
|
Yao GQ, Itokawa T, Paliwal I, Insogna K. CSF-1 induces fos gene transcription and activates the transcription factor Elk-1 in mature osteoclasts. Calcif Tissue Int 2005; 76:371-8. [PMID: 15812575 DOI: 10.1007/s00223-004-0099-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2004] [Accepted: 10/22/2004] [Indexed: 01/20/2023]
Abstract
Mice with targeted deletion of the fos gene fail to develop mature osteoclasts, reflecting an absolute requirement for the c-Fos proto-oncogene in osteoclast precursors. C-Fos is also expressed in mature osteoclasts; however, the regulation of fos in these cells has not been studied. By using cultured murine osteoclast-like cells (OCLs) we found that treatment with colony-stimulation factor 1 (CSF-1) induced a 3.9-fold increase in c-Fos rnRNA at 30 minutes and a 2.6-fold increase at 60 minutes. With use of mature osteoclasts isolated from transgenic mice expressing the bacterial Lac-Z gene under the control of the murine fos promoter, we were able to directly demonstrate transcriptional activation of fos by CSF-1 in these cells. Transcriptional activation was 2.6-fold greater at 5 minutes and 2.8-fold greater at 15 minutes in CSF-1-treated cells than in vehicle-treated cells. CSF-1 induced nuclear protein binding to the fos serum response element that was significantly attenuated by antibodies to the transcription factor Elk-1 but not by Sap-1a. Treatment of mature osteoclasts with CSF-1 for 2 hours resulted in a significant increase in the levels of nuclear c-Fos protein. These data demonstrate that CSF-1 upregulates c-fos expression in mature osteoclasts at least in part via transcriptional activation of fos. CSF-1 induced binding of Elk-1 to the fos gene serum response element appears to be part of the molecular mechanism by which this occurs.
Collapse
Affiliation(s)
- G-Q Yao
- Section of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520-8020, USA.
| | | | | | | |
Collapse
|
60
|
Murata S, Yoshiara T, Lim CR, Sugino M, Kogure M, Ohnuki T, Komurasaki T, Matsubara K. Psychophysiological stress-regulated gene expression in mice. FEBS Lett 2005; 579:2137-42. [PMID: 15811331 DOI: 10.1016/j.febslet.2005.02.069] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2005] [Revised: 02/28/2005] [Accepted: 02/28/2005] [Indexed: 11/27/2022]
Abstract
Eight genes showed significant changes in expression in mice under psychophysiological stress provided by cage-restraint and water-immersion. The transcription level of most of these genes was affected in all the tissues analyzed, and some of them were responsive genes in several different stress systems. Peculiarly, the expression level of one gene, cdc2-like kinase 1 (CLK1), was reduced only in the brain, while the balance of partially- and alternatively-spliced CLK1 mRNA species changed in all the tissues including the brain. These results suggest that some stress-response mechanisms, including transcriptional and post-transcriptional events, are coordinated in the whole body in mice under psychophysiological stress.
Collapse
Affiliation(s)
- Shigenori Murata
- DNA Chip Research Inc., 1-1-43 Suehiro, Tsurumi, Yokohama 230-0045, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
61
|
Abstract
RhoB is a small GTP-binding protein that is involved in apoptotic signal transduction. We have cloned the mouse RhoB mRNA including a 1377 nucleotide 3'-untranslated region (UTR) that contains six AU-rich elements (AREs) as well as several uridine-rich stretches. There is 94% homology overall between the mouse and rat RhoB genes and 92% homology between the mouse and a putative human clone. Ultraviolet light (UVL) induces RhoB production through regulated changes in gene transcription and mRNA stabilization although the latter mechanism is unknown. We observed that UVL increased the half-life of RhoB mRNA from 63 min to 3.3 h in NIH/3T3 cells and from 87 min to 2.7 h in normal human keratinocyte cells. In vitro mobility shift assays demonstrated that HuR bound the 3'-UTR of RhoB at three distinct locations (nucleotides 1342-1696, 1765-1920 and 1897-1977) suggesting a regulatory role for this RNA-binding protein. HuR immunoprecipitations were positive for RhoB mRNA indicating an in vivo association, and Western blot analysis and immunofluorescence demonstrated that HuR rapidly partitions from the nucleus to the cytoplasm after UVL. Therefore, we propose a model in which UVL induces stress-activated signal transduction leading to nuclear/cytoplasmic shuttling of HuR and subsequent stabilization of RhoB mRNA.
Collapse
Affiliation(s)
- Cara J Westmark
- Department of Pathology and Laboratory Medicine, Waisman Center for Developmental Disabilities, University of Wisconsin, Madison, WI 53705, USA
| | | | | |
Collapse
|
62
|
Paszek P, Lipniacki T, Brasier AR, Tian B, Nowak DE, Kimmel M. Stochastic effects of multiple regulators on expression profiles in eukaryotes. J Theor Biol 2004; 233:423-33. [PMID: 15652150 DOI: 10.1016/j.jtbi.2004.10.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2004] [Revised: 10/04/2004] [Accepted: 10/19/2004] [Indexed: 11/22/2022]
Abstract
The stochastic nature of gene regulation still remains not fully understood. In eukaryotes, the stochastic effects are primarily attributable to the binary nature of genes, which are considered either switched "on" or "off" due to the action of the transcription factors binding to the promoter. In the time period when the gene is activated, bursts of mRNA transcript are produced. In the present paper, we investigate regulation of gene expression at the single cell level. We propose a mechanism of gene regulation, which is able to explain the observed distinct transcription profiles assuming the number of co-regulatory activities, without attempting to identify the specific proteins involved. The model is motivated by our experiments on NF-kappaB-dependent genes in HeLa cells. Our experimental data shows that NF-kappaB-dependent genes can be stratified into three characteristic groups according to their expression profiles: early, intermediate and late having maximum of expression at about 1, 3 and 6 h, respectively, from the beginning of TNF stimulation. We provide a tractable analytical approach, not only in the terms of expected expression profiles and their moments, which corresponds to the measurements on the cell population, but also in the terms of single cell behavior. Comparison between these two modes of description reveals that single cells behave qualitatively different from the cell population. This analysis provides insights useful for understanding of microarray experiments.
Collapse
Affiliation(s)
- Pawel Paszek
- Department of Statistics, Rice University, 6100 Main Street, MS-138, Houston, TX 77005, USA.
| | | | | | | | | | | |
Collapse
|
63
|
Rieger KE, Chu G. Portrait of transcriptional responses to ultraviolet and ionizing radiation in human cells. Nucleic Acids Res 2004; 32:4786-803. [PMID: 15356296 PMCID: PMC519099 DOI: 10.1093/nar/gkh783] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2004] [Revised: 08/03/2004] [Accepted: 08/03/2004] [Indexed: 11/14/2022] Open
Abstract
To understand the human response to DNA damage, we used microarrays to measure transcriptional responses of 10 000 genes to ionizing radiation (IR) and ultraviolet radiation (UV). To identify bona fide responses, we used cell lines from 15 individuals and a rigorous statistical method, Significance Analysis of Microarrays (SAM). By exploring how sample number affects SAM, we rendered a portrait of the human damage response with a degree of accuracy unmatched by previous studies. By showing how SAM can be used to estimate the total number of responsive genes, we discovered that 24% of all genes respond to IR and 32% respond to UV, although most responses were less than 2-fold. Many genes were involved in known damage-response pathways for cell cycling and proliferation, apoptosis, DNA repair or the stress response. However, the majority of genes were involved in unexpected pathways, with functions in signal transduction, RNA binding and editing, protein synthesis and degradation, energy metabolism, metabolism of macromolecular precursors, cell structure and adhesion, vesicle transport, or lysosomal metabolism. Although these functions were not previously associated with the damage response in mammals, many were conserved in yeast. These insights reveal new directions for studying the human response to DNA damage.
Collapse
Affiliation(s)
- Kerri E Rieger
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | |
Collapse
|
64
|
Lipniacki T, Paszek P, Brasier ARAR, Luxon B, Kimmel M. Mathematical model of NF-kappaB regulatory module. J Theor Biol 2004; 228:195-215. [PMID: 15094015 DOI: 10.1016/j.jtbi.2004.01.001] [Citation(s) in RCA: 183] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2003] [Revised: 12/11/2003] [Accepted: 12/23/2003] [Indexed: 11/22/2022]
Abstract
The two-feedback-loop regulatory module of nuclear factor kappaB (NF-kappaB) signaling pathway is modeled by means of ordinary differential equations. The constructed model involves two-compartment kinetics of the activators IkappaB (IKK) and NF-kappaB, the inhibitors A20 and IkappaBalpha, and their complexes. In resting cells, the unphosphorylated IkappaBalpha binds to NF-kappaB and sequesters it in an inactive form in the cytoplasm. In response to extracellular signals such as tumor necrosis factor or interleukin-1, IKK is transformed from its neutral form (IKKn) into its active form (IKKa), a form capable of phosphorylating IkappaBalpha, leading to IkappaBalpha degradation. Degradation of IkappaBalpha releases the main activator NF-kappaB, which then enters the nucleus and triggers transcription of the inhibitors and numerous other genes. The newly synthesized IkappaBalpha leads NF-kappaB out of the nucleus and sequesters it in the cytoplasm, while A20 inhibits IKK converting IKKa into the inactive form (IKKi), a form different from IKKn, no longer capable of phosphorylating IkappaBalpha. After parameter fitting, the proposed model is able to properly reproduce time behavior of all variables for which the data are available: NF-kappaB, cytoplasmic IkappaBalpha, A20 and IkappaBalpha mRNA transcripts, IKK and IKK catalytic activity in both wild-type and A20-deficient cells. The model allows detailed analysis of kinetics of the involved proteins and their complexes and gives the predictions of the possible responses of whole kinetics to the change in the level of a given activator or inhibitor.
Collapse
Affiliation(s)
- Tomasz Lipniacki
- Institute of Fundamental Technological Research, Warsaw, Poland.
| | | | | | | | | |
Collapse
|
65
|
Pryor A, Tung L, Yang Z, Kapadia F, Chang TH, Johnson LF. Growth-regulated expression and G0-specific turnover of the mRNA that encodes URH49, a mammalian DExH/D box protein that is highly related to the mRNA export protein UAP56. Nucleic Acids Res 2004; 32:1857-65. [PMID: 15047853 PMCID: PMC390356 DOI: 10.1093/nar/gkh347] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
URH49 is a mammalian protein that is 90% identical to the DExH/D box protein UAP56, an RNA helicase that is important for splicing and nuclear export of mRNA. Although Saccharomyces cerevisiae and Drosophila express only a single protein corresponding to UAP56, mRNAs encoding URH49 and UAP56 are both expressed in human and mouse cells. Both proteins interact with the mRNA export factor Aly and both are able to rescue the loss of Sub2p (the yeast homolog of UAP56), indicating that both proteins have similar functions. UAP56 mRNA is more abundant than URH49 mRNA in many tissues, although in testes URH49 mRNA is much more abundant. UAP56 and URH49 mRNAs are present at similar levels in proliferating cultured cells. However, when the cells enter quiescence, the URH49 mRNA level decreases 3-6-fold while the UAP56 mRNA level remains relatively constant. The amount of URH49 mRNA increases to the level found in proliferating cells within 5 h when quiescent cells are growth-stimulated or when protein synthesis is inhibited. URH49 mRNA is relatively unstable (T(1/2) = 4 h) in quiescent cells, but is stabilized immediately following growth stimulation or inhibition of protein synthesis. In contrast, there is much less change in the content or stability of UAP56 mRNA following growth stimulation. Our observations suggest that in mammalian cells, two UAP56-like RNA helicases are involved in splicing and nuclear export of mRNA. Differential expression of these helicases may lead to quantitative or qualitative changes in mRNA expression.
Collapse
Affiliation(s)
- Anne Pryor
- The Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | |
Collapse
|
66
|
Malcolm T, Ettehadieh E, Sadowski I. Mitogen-responsive expression of RhoB is regulated by RNA stability. Oncogene 2003; 22:6142-50. [PMID: 13679852 DOI: 10.1038/sj.onc.1206638] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The small GTPase-encoding gene RhoB is strongly induced as part of the immediate early response of serum-stimulated fibroblasts. In this report, we have characterized the mechanism for growth factor responsiveness of RhoB in Rat-2 fibroblasts. By Northern blotting and ribonuclease protection, we observed low or barely detectable levels of RhoB mRNA in quiescent cells, but expression was transiently induced in response to serum stimulation, such that the mRNA peaked within 30 min and then declined over the next hour. Analysis of the rat promoter revealed cis-elements conserved with the mouse and human genes, including a pair of CEBP sites near the transcriptional start site. However, in contrast to the analysis of RNA, RhoB promoter fusions were constitutively expressed in quiescent cells in transient transfections, and were unaffected by serum. Similarly, stable RhoB promoter integrants were highly expressed in quiescent cells, and growth factor caused a slight decrease in activity. This indicates that growth factor-inducible RhoB expression cannot be mediated by transcriptional activation. We then examined decay of the RhoB mRNA and found that serum caused significant stabilization. Additionally, fusion of the 3' RhoB untranslated region (UTR) to a constitutively expressed reporter gene caused serum and growth factor as well as DNA damage-inducible expression. These observations are consistent with the view that RhoB mRNA is produced constitutively but its abundance is controlled in response to growth factors, and other signals including DNA damage, by stabilization through elements within the 3' UTR.
Collapse
Affiliation(s)
- Tom Malcolm
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2146 Health Sciences Mall, Vancouver, B.C., Canada V6 T 1Z3
| | | | | |
Collapse
|
67
|
Choi M, Rolle S, Wellner M, Cardoso MC, Scheidereit C, Luft FC, Kettritz R. Inhibition of NF-kappaB by a TAT-NEMO-binding domain peptide accelerates constitutive apoptosis and abrogates LPS-delayed neutrophil apoptosis. Blood 2003; 102:2259-67. [PMID: 12763940 DOI: 10.1182/blood-2002-09-2960] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Delivery of biologically active peptides into human polymorphonuclear neutrophils (PMNs) has implications for studying cellular functions and may be therapeutically relevant. The transcription factor nuclear factor-kappaB (NF-kappaB) regulates the expression of multiple genes controlling inflammation, proliferation, and cell survival. PMNs play a crucial role in first-line defense. Targeting NF-kappaB in these cells may promote apoptosis and therefore facilitate resolution of inflammation. We used an 11-amino acid sequence NEMO-binding domain (NBD) that selectively inhibits the IKKgamma (NEMO)/IKKbeta interaction, preventing NF-kappaB activation. An HIV-TAT sequence served as a highly effective transducing shuttle. We show that lipopolysaccharide (LPS), granulocyte-macrophage colony-stimulating factor (GM-CSF), and dexamethasone (DEX) significantly reduced apoptosis after 20 hours. LPS, but not GM-CSF or DEX, activated NF-kappaB as shown by IkappaBalpha degradation, NF-kappaB DNA binding, and transcriptional activity. The TAT-NBD blocked LPS-induced NF-kappaB activation and NF-kappaB-dependent gene expression. TAT-NBD accelerated constitutive PMN apoptosis dose dependently and abrogated LPS-delayed apoptosis. These results provide a proof of principle for peptide delivery by TAT-derived protein transduction domains to specifically inhibit NF-kappaB activity in PMNs. This strategy may help in controlling various cellular functions even in short-lived, transfection-resistant primary human cells.
Collapse
Affiliation(s)
- Mira Choi
- Division of Nephrology, Franz Volhard Clinic, Medical Faculty of the Charité, Humboldt Univertsity of Berlin, Wiltbergstrasse 50, 13122 Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
68
|
Miccoli L, Biard DSF, Frouin I, Harper F, Maga G, Angulo JF. Selective interactions of human kin17 and RPA proteins with chromatin and the nuclear matrix in a DNA damage- and cell cycle-regulated manner. Nucleic Acids Res 2003; 31:4162-75. [PMID: 12853634 PMCID: PMC165974 DOI: 10.1093/nar/gkg459] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Several proteins involved in DNA synthesis are part of the so-called 'replication factories' that are anchored on non-chromatin nuclear structures. We report here that human kin17, a nuclear stress-activated protein, associates with both chromatin and non-chromatin nuclear structures in a cell cycle- and DNA damage-dependent manner. After L-mimosine block and withdrawal we observed that kin17 protein was recruited in the nucleus during re-entry and progression through S phase. These results are consistent with a role of kin17 protein in DNA replication. About 50% of the total amount of kin17 protein was detected on nuclear structures and could not be released by detergents. Furthermore, the amount of kin17 protein greatly increased in both G(1)/S and S phase-arrested cells in fractions containing proteins anchored to nuclear structures. The detection of kin17 protein showed for the first time its preferential assembly within non-chromatin nuclear structures in G(1)/S and S phase-arrested cells, while the association with these structures was found to be less stable in the G(2)/M phase, as judged by fractionation of human cells and immunostaining. In asynchronous growing cells, kin17 protein interacted with both chromatin DNA and non-chromatin nuclear structures, while in S phase-arrested cells it interacted mostly with non-chromatin nuclear structures, as judged by DNase I treatment and in vivo UV cross-linking. In the presence of DNA damage in S phase cells, the distribution of kin17 protein became mainly associated with chromosomal DNA, as judged by limited formaldehyde cross-linking of living cells. The physical interaction of kin17 protein with components of the nuclear matrix was confirmed and visualized by indirect immunofluorescence and immunoelectron microscopy. Our results indicate that, during S phase, a fraction of the human kin17 protein preferentially associates with the nuclear matrix, a fundamentally non-chromatin higher order nuclear structure, and to chromatin DNA in the presence of DNA damage.
Collapse
Affiliation(s)
- Laurent Miccoli
- Commissariat à l'Energie Atomique, Direction des Sciences du Vivant, Laboratoire de Génétique de la Radiosensibilité, Département de Radiobiologie et de Radiopathologie, F-92265 Fontenay-aux-Roses, France.
| | | | | | | | | | | |
Collapse
|
69
|
Figueroa A, Cuadrado A, Fan J, Atasoy U, Muscat GE, Muñoz-Canoves P, Gorospe M, Muñoz A. Role of HuR in skeletal myogenesis through coordinate regulation of muscle differentiation genes. Mol Cell Biol 2003; 23:4991-5004. [PMID: 12832484 PMCID: PMC162217 DOI: 10.1128/mcb.23.14.4991-5004.2003] [Citation(s) in RCA: 159] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In this report, we investigate the role of the RNA-binding protein HuR during skeletal myogenesis. At the onset of myogenesis in differentiating C2C12 myocytes and in vivo in regenerating mouse muscle, HuR cytoplasmic abundance increased dramatically, returning to a predominantly nuclear presence upon completion of myogenesis. mRNAs encoding key regulators of myogenesis-specific transcription (myogenin and MyoD) and cell cycle withdrawal (p21), bearing AU-rich regions, were found to be targets of HuR in a differentiation-dependent manner. Accordingly, mRNA half-lives were highest during differentiation, declining when differentiation was completed. Importantly, HuR-overexpressing C2C12 cells displayed increased target mRNA expression and half-life and underwent precocious differentiation. Our findings underscore a critical function for HuR during skeletal myogenesis linked to HuR's coordinate regulation of muscle differentiation genes.
Collapse
Affiliation(s)
- Angélica Figueroa
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
70
|
Despras E, Miccoli L, Créminon C, Rouillard D, Angulo JF, Biard DSF. Depletion of KIN17, a human DNA replication protein, increases the radiosensitivity of RKO cells. Radiat Res 2003; 159:748-58. [PMID: 12751957 DOI: 10.1667/0033-7587(2003)159[0748:dokahd]2.0.co;2] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The human KIN17 protein is a chromatin-associated protein involved in DNA replication. Certain tumor cell lines overproduce KIN17 protein. Among 16 cell lines, the highest KIN17 protein level was observed in H1299 non-small cell lung cancer cells, whereas the lowest was detected in MeWo melanoma cells. Cells displaying higher KIN17 protein levels exhibited elevated RPA70 protein contents. High KIN17 protein levels may be a consequence of the tumorigenic phenotype or a prerequisite for tumor progression. Twenty-four hours after exposure to ionizing radiation, after the completion of DNA repair, a co-induction of chromatin-bound KIN17 and RPA70 proteins was detected. Etoposide, an inhibitor of topoisomerase II generating double-strand breaks, triggered the concentration of KIN17 into punctuate intranuclear foci. KIN17 may be associated with unrepaired DNA sites. Flow cytometry analysis revealed that 48 h after transfection the uppermost KIN17-positive RKO cells shifted in the cell cycle toward higher DNA content, suggesting that KIN17 protein induced defects in chromatin conformation. Cells displaying reduced levels of KIN17 transcript exhibited a sixfold increased radiosensitivity at 2 Gy. The KIN17 protein may be a component of the DNA replication machinery that participates in the cellular response to unrepaired DSBs, and an impaired KIN17 pathway leads to an increased sensitivity to ionizing radiation.
Collapse
Affiliation(s)
- E Despras
- CEA, Commissariat à l'Energie Atomique, Laboratoire de Génétique de la Radiosensibilité, Département de Radiobiologie et de Radiopathologie, Direction des Sciences du Vivant, Fontenay-aux-Roses 92265, France
| | | | | | | | | | | |
Collapse
|
71
|
Masson C, Menaa F, Pinon-Lataillade G, Frobert Y, Chevillard S, Radicella JP, Sarasin A, Angulo JF. Global genome repair is required to activate KIN17, a UVC-responsive gene involved in DNA replication. Proc Natl Acad Sci U S A 2003; 100:616-21. [PMID: 12525703 PMCID: PMC141045 DOI: 10.1073/pnas.0236176100] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
UV light provokes DNA lesions that interfere with replication and transcription. These lesions may compromise cell viability and usually are removed by nucleotide excision repair (NER). In humans, inactivation of NER is associated with three rare autosomal recessive inherited disorders: xeroderma pigmentosum (XP), Cockayne syndrome, and trichothiodystrophy. The NER earliest step is lesion recognition by a complex formed by XPC and HHR23B proteins. In a subsequent step, XPA protein becomes associated to the repair complex. Here we investigate whether XPA and XPC proteins, involved in global genome repair, may contribute to a signal transduction pathway regulating the response to UVC-induced lesions. We monitored the expression of several UVC-induced genes in cells deficient in either a transduction pathway or mutated on an NER gene. Expression of the KIN17 gene is induced after UVC irradiation independently of p53 and of activating transcription factor 2. However, in human cells derived from XPA or XPC patients the UVC-induced accumulation of KIN17 RNA and protein is abolished. Our results indicate that the presence of functional XPA and XPC proteins is essential for the up-regulation of the KIN17 gene after UVC irradiation. They also show that the integrity of global genome repair is required to trigger KIN17 gene expression and probably other UVC-responsive genes.
Collapse
Affiliation(s)
- Christel Masson
- Laboratoire de Génétique de la Radiosensibilité, Département de Radiobiologie et de Radiopathologie (DRR), Direction des Sciences du Vivant (DSV), Commissariat à l'Energie Atomique (CEA), B.P. 6, 92265 Fontenay aux Roses, France
| | | | | | | | | | | | | | | |
Collapse
|
72
|
Bollig F, Winzen R, Kracht M, Ghebremedhin B, Ritter B, Wilhelm A, Resch K, Holtmann H. Evidence for general stabilization of mRNAs in response to UV light. EUROPEAN JOURNAL OF BIOCHEMISTRY 2002; 269:5830-9. [PMID: 12444971 DOI: 10.1046/j.1432-1033.2002.03300.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
mRNA stabilization plays an important role in the changes in protein expression initiated by inducers of inflammation or direct cell stress such as UV light. This study provides evidence that stabilization in response to UV light differs from that induced by proinflammatory stimuli such as bacterial lipopolysaccharide or interleukin (IL)-1. Firstly, UV-induced stabilization is independent of the p38 MAP kinase pathway, which has previously been shown to mediate stabilization induced by IL-1 or lipopolysaccharide. UV-induced mRNA stabilization was insensitive to the dominant negative forms of p38 MAP kinase and its substrate MAP kinase-activated protein kinase 2 (MK2), or to the p38 MAP kinase inhibitor SB 203580, demonstrating that it occurs through a different signaling mechanism. Secondly, UV-induced stabilization exhibits a different transcript selectivity. Activation of the p38 MAP kinase pathway, by expressing active MAP kinase kinase 6, induced stabilization only of transcripts containing AU-rich elements. UV light also induced stabilization of transcripts lacking AU-rich elements. This effect could not be mimicked by expressing MEKK1, an upstream activator of the p38, JNK, ERK and NF-kappaB pathways. UV light also stabilized endogenous histone mRNA, which lacks AU-rich elements and a poly(A) tail. This effect was not mimicked by active MAP kinase kinase 6 and not sensitive to a p38 MAP kinase inhibitor. This suggests that UV light induces stabilization through a mechanism that is independent of p38 MAP kinase and affects a broad spectrum of mRNAs.
Collapse
Affiliation(s)
- Frank Bollig
- Institute of Pharmacology, Medical School Hannover, Germany
| | | | | | | | | | | | | | | |
Collapse
|
73
|
Kracht M, Saklatvala J. Transcriptional and post-transcriptional control of gene expression in inflammation. Cytokine 2002; 20:91-106. [PMID: 12453467 DOI: 10.1006/cyto.2002.0895] [Citation(s) in RCA: 181] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Michael Kracht
- Institute of Phamacology, Medical School Hannover, Carl-Neuberg Strasse 1, D-30625, Hannover, Germany.
| | | |
Collapse
|
74
|
Dodson RE, Shapiro DJ. Regulation of pathways of mRNA destabilization and stabilization. PROGRESS IN NUCLEIC ACID RESEARCH AND MOLECULAR BIOLOGY 2002; 72:129-64. [PMID: 12206451 DOI: 10.1016/s0079-6603(02)72069-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The level of an mRNA in the cytoplasm represents a balance between the rate at which the mRNA precursor is synthesized in the nucleus and the rates of nuclear RNA processing and export and cytoplasmic mRNA degradation. Although most studies of gene expression have focused on gene transcription and in the area of eukaryotic mRNA degradation, but to provide a short general discussion of the importance of mRNA degradation and its regulation and a brief overview of recent findings and present knowledge. The overview is followed by a more in-depth discussion of one of the several pathways for mRNA degradation. We concentrate on the pathway for regulated mRNA degradation mediated by mRNA-binding proteins and endonucleases that cleave within the body of mRNAs. As a potential example of this type of control, we focus on the regulated degradation of the egg yolk precursor protein vitellogenin on the mRNA-binding protein vigilin and the mRNA endonuclease polysomal ribonuclease 1 (PMR-1).
Collapse
Affiliation(s)
- Robin E Dodson
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana 61801, USA
| | | |
Collapse
|
75
|
Mathas S, Hinz M, Anagnostopoulos I, Krappmann D, Lietz A, Jundt F, Bommert K, Mechta-Grigoriou F, Stein H, Dörken B, Scheidereit C. Aberrantly expressed c-Jun and JunB are a hallmark of Hodgkin lymphoma cells, stimulate proliferation and synergize with NF-kappa B. EMBO J 2002; 21:4104-13. [PMID: 12145210 PMCID: PMC126136 DOI: 10.1093/emboj/cdf389] [Citation(s) in RCA: 278] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AP-1 family transcription factors have been implicated in the control of proliferation, apoptosis and malignant transformation. However, their role in oncogenesis is unclear and no recurrent alterations of AP-1 activities have been described in human cancers. Here, we show that constitutively activated AP-1 with robust c-Jun and JunB overexpression is found in all tumor cells of patients with classical Hodgkin's disease. A similar AP-1 activation is present in anaplastic large cell lymphoma (ALCL), but is absent in other lymphoma types. Whereas c-Jun is up-regulated by an autoregulatory process, JunB is under control of NF-kappa B. Activated AP-1 supports proliferation of Hodgkin cells, while it suppresses apoptosis of ALCL cells. Furthermore, AP-1 cooperates with NF-kappa B and stimulates expression of the cell-cycle regulator cyclin D2, proto-oncogene c-met and the lymphocyte homing receptor CCR7, which are all strongly expressed in primary HRS cells. Together, these data suggest an important role of AP-1 in lymphoma pathogenesis.
Collapse
MESH Headings
- Cell Division
- Cell Transformation, Neoplastic/genetics
- Cyclin D2
- Cyclins/biosynthesis
- Cyclins/genetics
- DNA, Neoplasm/metabolism
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/physiology
- Gene Expression Regulation, Neoplastic/radiation effects
- Genes, jun
- Hodgkin Disease/genetics
- Hodgkin Disease/pathology
- Humans
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Non-Hodgkin/genetics
- Lymphoma, Non-Hodgkin/pathology
- MAP Kinase Signaling System/drug effects
- MAP Kinase Signaling System/radiation effects
- Mitogens/pharmacology
- NF-kappa B/physiology
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neoplasm Proteins/physiology
- Proto-Oncogene Mas
- Proto-Oncogene Proteins c-jun/biosynthesis
- Proto-Oncogene Proteins c-jun/genetics
- Proto-Oncogene Proteins c-jun/physiology
- Proto-Oncogene Proteins c-met/biosynthesis
- Proto-Oncogene Proteins c-met/genetics
- Receptors, CCR7
- Receptors, Chemokine/biosynthesis
- Receptors, Chemokine/genetics
- Recombinant Fusion Proteins/physiology
- Reed-Sternberg Cells/drug effects
- Reed-Sternberg Cells/metabolism
- Reed-Sternberg Cells/radiation effects
- Tetradecanoylphorbol Acetate/pharmacology
- Transcription Factor AP-1/genetics
- Transcription Factor AP-1/physiology
- Transcription, Genetic
- Transfection
- Tumor Cells, Cultured/drug effects
- Tumor Cells, Cultured/metabolism
- Tumor Cells, Cultured/radiation effects
- Ultraviolet Rays
Collapse
Affiliation(s)
- Stephan Mathas
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, D-13125 Berlin, Universitätsklinikum Charité, Robert-Rössle-Klinik, Humboldt University, Lindenberger Weg 80, D-13125 Berlin, Institute for Pathology, Universitätsklinikum Benjamin Franklin, Free University, Hindenburgdamm 30, 12200 Berlin, Germany and Unité des Virus Oncogènes, URA CNRS 1644, Institut Pasteur, 28 Rue du Dr Roux, 75724 Paris cedex 15, France Corresponding author e-mail:
| | - Michael Hinz
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, D-13125 Berlin, Universitätsklinikum Charité, Robert-Rössle-Klinik, Humboldt University, Lindenberger Weg 80, D-13125 Berlin, Institute for Pathology, Universitätsklinikum Benjamin Franklin, Free University, Hindenburgdamm 30, 12200 Berlin, Germany and Unité des Virus Oncogènes, URA CNRS 1644, Institut Pasteur, 28 Rue du Dr Roux, 75724 Paris cedex 15, France Corresponding author e-mail:
| | - Ioannis Anagnostopoulos
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, D-13125 Berlin, Universitätsklinikum Charité, Robert-Rössle-Klinik, Humboldt University, Lindenberger Weg 80, D-13125 Berlin, Institute for Pathology, Universitätsklinikum Benjamin Franklin, Free University, Hindenburgdamm 30, 12200 Berlin, Germany and Unité des Virus Oncogènes, URA CNRS 1644, Institut Pasteur, 28 Rue du Dr Roux, 75724 Paris cedex 15, France Corresponding author e-mail:
| | - Daniel Krappmann
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, D-13125 Berlin, Universitätsklinikum Charité, Robert-Rössle-Klinik, Humboldt University, Lindenberger Weg 80, D-13125 Berlin, Institute for Pathology, Universitätsklinikum Benjamin Franklin, Free University, Hindenburgdamm 30, 12200 Berlin, Germany and Unité des Virus Oncogènes, URA CNRS 1644, Institut Pasteur, 28 Rue du Dr Roux, 75724 Paris cedex 15, France Corresponding author e-mail:
| | - Andreas Lietz
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, D-13125 Berlin, Universitätsklinikum Charité, Robert-Rössle-Klinik, Humboldt University, Lindenberger Weg 80, D-13125 Berlin, Institute for Pathology, Universitätsklinikum Benjamin Franklin, Free University, Hindenburgdamm 30, 12200 Berlin, Germany and Unité des Virus Oncogènes, URA CNRS 1644, Institut Pasteur, 28 Rue du Dr Roux, 75724 Paris cedex 15, France Corresponding author e-mail:
| | - Franziska Jundt
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, D-13125 Berlin, Universitätsklinikum Charité, Robert-Rössle-Klinik, Humboldt University, Lindenberger Weg 80, D-13125 Berlin, Institute for Pathology, Universitätsklinikum Benjamin Franklin, Free University, Hindenburgdamm 30, 12200 Berlin, Germany and Unité des Virus Oncogènes, URA CNRS 1644, Institut Pasteur, 28 Rue du Dr Roux, 75724 Paris cedex 15, France Corresponding author e-mail:
| | - Kurt Bommert
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, D-13125 Berlin, Universitätsklinikum Charité, Robert-Rössle-Klinik, Humboldt University, Lindenberger Weg 80, D-13125 Berlin, Institute for Pathology, Universitätsklinikum Benjamin Franklin, Free University, Hindenburgdamm 30, 12200 Berlin, Germany and Unité des Virus Oncogènes, URA CNRS 1644, Institut Pasteur, 28 Rue du Dr Roux, 75724 Paris cedex 15, France Corresponding author e-mail:
| | - Fatima Mechta-Grigoriou
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, D-13125 Berlin, Universitätsklinikum Charité, Robert-Rössle-Klinik, Humboldt University, Lindenberger Weg 80, D-13125 Berlin, Institute for Pathology, Universitätsklinikum Benjamin Franklin, Free University, Hindenburgdamm 30, 12200 Berlin, Germany and Unité des Virus Oncogènes, URA CNRS 1644, Institut Pasteur, 28 Rue du Dr Roux, 75724 Paris cedex 15, France Corresponding author e-mail:
| | - Harald Stein
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, D-13125 Berlin, Universitätsklinikum Charité, Robert-Rössle-Klinik, Humboldt University, Lindenberger Weg 80, D-13125 Berlin, Institute for Pathology, Universitätsklinikum Benjamin Franklin, Free University, Hindenburgdamm 30, 12200 Berlin, Germany and Unité des Virus Oncogènes, URA CNRS 1644, Institut Pasteur, 28 Rue du Dr Roux, 75724 Paris cedex 15, France Corresponding author e-mail:
| | - Bernd Dörken
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, D-13125 Berlin, Universitätsklinikum Charité, Robert-Rössle-Klinik, Humboldt University, Lindenberger Weg 80, D-13125 Berlin, Institute for Pathology, Universitätsklinikum Benjamin Franklin, Free University, Hindenburgdamm 30, 12200 Berlin, Germany and Unité des Virus Oncogènes, URA CNRS 1644, Institut Pasteur, 28 Rue du Dr Roux, 75724 Paris cedex 15, France Corresponding author e-mail:
| | - Claus Scheidereit
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, D-13125 Berlin, Universitätsklinikum Charité, Robert-Rössle-Klinik, Humboldt University, Lindenberger Weg 80, D-13125 Berlin, Institute for Pathology, Universitätsklinikum Benjamin Franklin, Free University, Hindenburgdamm 30, 12200 Berlin, Germany and Unité des Virus Oncogènes, URA CNRS 1644, Institut Pasteur, 28 Rue du Dr Roux, 75724 Paris cedex 15, France Corresponding author e-mail:
| |
Collapse
|
76
|
Alarcon-Vargas D, Tansey WP, Ronai Z. Regulation of c-myc stability by selective stress conditions and by MEKK1 requires aa 127-189 of c-myc. Oncogene 2002; 21:4384-91. [PMID: 12080469 DOI: 10.1038/sj.onc.1205543] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2001] [Revised: 03/27/2002] [Accepted: 04/02/2002] [Indexed: 11/09/2022]
Abstract
C-myc availability is central for its ability to serve as a regulator of cell growth and death. Here we study the regulation of c-myc protein stability and identify domains of c-myc that are important for its stabilization in response to stress kinases activated following selective stress conditions. UV-irradiation elicited an increase in c-myc protein levels, which could be attenuated by inhibitors of stress kinases but also by actinomycin D-inhibitor of transcription. Inhibition of protein synthesis results in a noticeable decrease in c-myc levels, further pointing to the short half-life of the protein. However, in combination with tumor necrosis factor-alpha (TNF-alpha), cycloheximide efficiently increases steady-state levels of c-myc, suggesting that selective stress conditions are required to increase c-myc protein stability. Expression of MEKK1, an upstream regulator of protein kinases that has been implicated in mediating the response to diverse stress conditions, also results in an efficient increase in the half-life of c-myc protein. To map c-myc domains that are responsive to stress kinases, we monitored changes in the level of c-myc deletion mutants following MEKK1 expression. Of the seven c-myc deletion mutants analysed, the domain spanning amino acids 127-189 was found to be required for MEKK1-dependent increase in c-myc stability. In all, the present study identifies a novel domain that is important for the regulation of c-myc stability by stress kinases in response to selective stress conditions.
Collapse
Affiliation(s)
- Dania Alarcon-Vargas
- Ruttenberg Cancer Center, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | |
Collapse
|
77
|
Gonzales M, Bowden GT. Nordihydroguaiaretic acid-mediated inhibition of ultraviolet B-induced activator protein-1 activation in human keratinocytes. Mol Carcinog 2002; 34:102-11. [PMID: 12112316 DOI: 10.1002/mc.10052] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Nordihydroguaiaretic acid (NDGA) is a polyphenolic compound from the Larrea tridentata bush that has been identified as a chemopreventive drug in animal studies. Topically applied NDGA has been shown to prevent phorbol ester promotion of tumors in mouse skin, suggesting that NDGA may be a candidate drug for the chemoprevention of skin cancer. Ultraviolet (UV) B irradiation from sunlight exposure is the major cause of human skin cancer. UVB irradiation causes epigenetic alterations in target keratinocytes, such as the upregulation of signal transduction pathways that induce the expression of transcription factors. Specifically, UVB induces activator protein-1 (AP-1), a transcription factor complex that alters normal cellular gene expression. A component of the UVB-induced AP-1 complex, c-fos, also was identified as a mediator of the signaling pathway that leads to AP-1 activation. Thus, NDGA was investigated as a potential inhibitory agent for UVB-induced signaling pathways in the human keratinocyte cell line HaCaT. NDGA significantly inhibited UVB-induced c-fos and AP-1 transactivation. In addition, NDGA was found to inhibit activity of phosphatidylinositol 3-kinase (PI 3-kinase), a UVB-inducible enzyme that contributes to the induced expression of c-fos and AP-1. Therefore, NDGA prevents UVB-induced c-fos expression and AP-1 transactivation by inhibiting the PI 3-kinase signaling pathway. Effective skin chemoprevention strategies may incorporate NDGA to inhibit components of the UVB-induced cell signaling pathways that increase AP-1 activity.
Collapse
Affiliation(s)
- Melissa Gonzales
- Department of Molecular and Cellular Biology, Arizona Cancer Center, College of Medicine, University of Arizona, Tucson, USA
| | | |
Collapse
|
78
|
Tapinos NI, Polihronis M, Thyphronitis G, Moutsopoulos HM. Characterization of the cysteine-rich secretory protein 3 gene as an early-transcribed gene with a putative role in the pathophysiology of Sjögren's syndrome. ARTHRITIS AND RHEUMATISM 2002; 46:215-22. [PMID: 11817594 DOI: 10.1002/1529-0131(200201)46:1<215::aid-art10024>3.0.co;2-m] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To identify genes that may participate in the pathophysiology of Sjögren's syndrome (SS), the technique of differential display was applied to labial minor salivary gland (MSG) biopsy samples. METHODS Total RNA was isolated from MSG biopsy samples from a woman with primary SS and a control subject, and the differential display protocol with 8 different random oligonucleotide primers was performed. One particular differentially expressed fragment showed 98% homology with the cysteine-rich secretory protein 3 (CRISP-3) gene. The result was verified by reverse transcription-polymerase chain reaction (RT-PCR) with messenger RNA (mRNA) samples from MSG biopsy tissues obtained from 4 women with primary SS. A CRISP-3 RNA probe was synthesized for in situ hybridization of 7 MSG biopsy samples from patients with primary SS. In an attempt to interpret the expression of CRISP-3, normal peripheral blood lymphocytes (PBLs) were activated in vitro at different time points and assayed for CRISP-3 expression. Finally, B cells were transfected with the coding region of CRISP-3 and monitored for the up-regulation of different B cell activation markers. RESULTS The CRISP-3 gene was detected by RT-PCR in all SS patients tested. Mainly the mononuclear cells infiltrating the MSGs of patients expressed CRISP-3 mRNA. In addition, CRISP-3 was detected by RT-PCR between 30 minutes and 6 hours in phorbol myristate acetate-activated normal PBLs, while staurosporine inhibited this expression. CRISP-3-transfected B cells exhibited an up-regulation in CD25 surface expression. CONCLUSION The CRISP-3 gene is identified as a novel early response gene that may participate in the pathophysiology of the autoimmune lesions of SS.
Collapse
|
79
|
Jean S, Bideau C, Bellon L, Halimi G, De Méo M, Orsière T, Dumenil G, Bergé-Lefranc JL, Botta A. The expression of genes induced in melanocytes by exposure to 365-nm UVA: study by cDNA arrays and real-time quantitative RT-PCR. BIOCHIMICA ET BIOPHYSICA ACTA 2001; 1522:89-96. [PMID: 11750059 DOI: 10.1016/s0167-4781(01)00326-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ultraviolet A radiation (UVA; 320-400 nm) constitutes more than 90% of the terrestrial UV solar energy. This type of radiation generates reactive oxygen species and consequently induces DNA damage. UVA irradiation is now considered to be an important carcinogen agent especially in the development of melanoma. UVA radiation is known to activate several pathways in mammalian cells. We have used cDNA arrays to analyze differential gene expression in primary cultures of human melanocytes in response to 365-nm UVA. Among 588 genes studied, 11 were overexpressed. These genes included genes involved in cell cycle regulation (GADD45, CIP1/WAF1), in stress response (HSP70, HSP40, HSP86), in apoptosis (GADD153, tristetraproline) and genes encoding transcription factors (EGR-1, ETR-101, c-JUN, ATF4). This coordinate gene regulation was confirmed by real-time quantitative RT-PCR.
Collapse
Affiliation(s)
- S Jean
- Laboratoire de Biogénotoxicologie et Mutagenèse Environnementale, Facultés de Médecine et de Pharmacie, Université de la Méditerranée, Marseille, France
| | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Thakur M, Wernick M, Collins C, Limoli CL, Crowley E, Cleaver JE. DNA polymerase eta undergoes alternative splicing, protects against UV sensitivity and apoptosis, and suppresses Mre11-dependent recombination. Genes Chromosomes Cancer 2001; 32:222-35. [PMID: 11579462 DOI: 10.1002/gcc.1186] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Polymerase eta (pol eta) is a low-fidelity DNA polymerase that is the product of the gene, POLH, associated with the human XP variant disorder in which there is an extremely high level of solar-induced skin carcinogenesis. The complete human genomic sequence spans about 40 kb containing 10 coding exons and a cDNA of 2.14 kb; exon I is untranslated and is 6 kb upstream from the first coding exon. Using bacterial artificial chromosomes (BACs), the gene was mapped to human chromosome band 6p21 and mouse band 17D. The gene is expressed in most tissues, except for very low or undetectable levels in peripheral lymphocytes, fetal spleen, and adult muscle; exon II, however, is frequently spliced out in normal cells and in almost half the transcripts in the testis and fetal liver. Expression of POLH in a multicopy episomal vector proved nonviable, suggesting that overexpression is toxic. Expression from chromosomally integrated linear copies using either an EF1-alpha or CMV promoter was functional, resulting in cell lines with low or high levels of pol eta protein, respectively. Point mutations in the center of the gene and in a C-terminal cysteine and deletion of exon II resulted in inactivation, but addition of a terminal 3 amino acid C-terminal tag, or an N- or C-terminal green fluorescent protein, had no effect on function. A low level of expression of pol eta eliminated hMre11 recombination and partially restored UV survival, but did not prevent UV-induced apoptosis, which required higher levels of expression. Polymerase eta is therefore involved in S-phase checkpoint and signal transduction pathways that lead to arrest in S, apoptosis, and recombination. In normal cells, the predominant mechanism of replication of UV damage involves pol eta-dependent bypass, and Mre11-dependent recombination that acts is a secondary, backup mechanism when cells are severely depleted of pol eta.
Collapse
Affiliation(s)
- M Thakur
- UCSF Comprehensive Cancer Center, University of California, San Francisco, California 94115, USA
| | | | | | | | | | | |
Collapse
|
81
|
Masson C, Menaa F, Pinon-Lataillade G, Frobert Y, Radicella JP, Angulo JF. Identification of KIN (KIN17), a human gene encoding a nuclear DNA-binding protein, as a novel component of the TP53-independent response to ionizing radiation. Radiat Res 2001; 156:535-44. [PMID: 11604067 DOI: 10.1667/0033-7587(2001)156[0535:iokkah]2.0.co;2] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Ionizing radiation elicits a genetic response in human cells that allows cell survival. The human KIN (also known as KIN17) gene encodes a 45-kDa nuclear DNA-binding protein that participates in the response to UVC radiation and is immunologically related to the bacterial RecA protein. We report for the first time that ionizing radiation and bleomycin, a radiomimetic drug, which produce single- and double-strand breaks, increased expression of KIN in human cells established from tumors, including MeWo melanoma, MCF7 breast adenocarcinoma, and ATM+ GM3657 lymphoblast cells. KIN expression increased rapidly in a dose-dependent manner after irradiation. Under the same conditions, several genes controlled by TP53 were induced with kinetics similar to that of KIN. Using the CDKN1A gene as a marker of TP53 responsiveness, we analyzed the up-regulation of KIN and showed that is independent of the status of TP53 and ATM. In contrast, the presence of a dominant mutant for activating transcription factor 2 (ATF2) completely abolished the up-regulation of KIN. Our results suggest a role for ATF2 in the TP53-independent increase in KIN expression after gamma irradiation.
Collapse
Affiliation(s)
- C Masson
- Laboratoire de Génétique de la Radiosensibilité, Direction des Sciences du Vivant, Centre d'Etudes de Fontenay-aux-Roses, CEA, 60-68, Avenue du Général-Leclerc, B.P. no. 6, 92265 Fontenay-aux-Roses Cedex, France
| | | | | | | | | | | |
Collapse
|
82
|
Evdokimova V, Ruzanov P, Imataka H, Raught B, Svitkin Y, Ovchinnikov LP, Sonenberg N. The major mRNA-associated protein YB-1 is a potent 5' cap-dependent mRNA stabilizer. EMBO J 2001; 20:5491-502. [PMID: 11574481 PMCID: PMC125650 DOI: 10.1093/emboj/20.19.5491] [Citation(s) in RCA: 224] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
mRNA silencing and storage play an important role in gene expression under diverse circumstances, such as throughout early metazoan development and in response to many types of environmental stress. Here we demonstrate that the major mRNA-associated protein YB-1, also termed p50, is a potent cap-dependent mRNA stabilizer. YB-1 addition or overexpression dramatically increases mRNA stability in vitro and in vivo, whereas YB-1 depletion results in accelerated mRNA decay. The cold shock domain of YB-1 is responsible for the mRNA stabilizing activity, and a blocked mRNA 5' end is required for YB-1-mediated stabilization. Significantly, exogenously added YB-1 destabilizes the interaction of the cap binding protein, eIF4E, with the mRNA cap structure. Conversely, sequestration of eIF4E from the cap increases the association of endogenous YB-1 with mRNA at or near the cap, and significantly enhances mRNA stability. These data support a model whereby down-regulation of eIF4E activity or increasing the YB-1 mRNA binding activity or concentration in cells activates a general default pathway for mRNA stabilization.
Collapse
Affiliation(s)
- Valentina Evdokimova
- Department of Biochemistry and McGill Cancer Centre, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada, H3G 1Y6 and Institute of Protein Research, Russian Academy of Sciences, Puschino, Moscow Region, Russian Federation, 142 292 Corresponding author e-mail:
| | - Peter Ruzanov
- Department of Biochemistry and McGill Cancer Centre, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada, H3G 1Y6 and Institute of Protein Research, Russian Academy of Sciences, Puschino, Moscow Region, Russian Federation, 142 292 Corresponding author e-mail:
| | - Hiroaki Imataka
- Department of Biochemistry and McGill Cancer Centre, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada, H3G 1Y6 and Institute of Protein Research, Russian Academy of Sciences, Puschino, Moscow Region, Russian Federation, 142 292 Corresponding author e-mail:
| | - Brian Raught
- Department of Biochemistry and McGill Cancer Centre, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada, H3G 1Y6 and Institute of Protein Research, Russian Academy of Sciences, Puschino, Moscow Region, Russian Federation, 142 292 Corresponding author e-mail:
| | - Yuri Svitkin
- Department of Biochemistry and McGill Cancer Centre, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada, H3G 1Y6 and Institute of Protein Research, Russian Academy of Sciences, Puschino, Moscow Region, Russian Federation, 142 292 Corresponding author e-mail:
| | - Lev P. Ovchinnikov
- Department of Biochemistry and McGill Cancer Centre, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada, H3G 1Y6 and Institute of Protein Research, Russian Academy of Sciences, Puschino, Moscow Region, Russian Federation, 142 292 Corresponding author e-mail:
| | - Nahum Sonenberg
- Department of Biochemistry and McGill Cancer Centre, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada, H3G 1Y6 and Institute of Protein Research, Russian Academy of Sciences, Puschino, Moscow Region, Russian Federation, 142 292 Corresponding author e-mail:
| |
Collapse
|
83
|
Araneda S, Mermet N, Verjat T, Angulo JF, Radicella JP. Expression of Kin17 and 8-OxoG DNA glycosylase in cells of rodent and quail central nervous system. Brain Res Bull 2001; 56:139-46. [PMID: 11704351 DOI: 10.1016/s0361-9230(01)00620-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Kin17 and 8-Oxoguanine DNA glycosylase (Ogg1) are proteins, respectively, involved in illegitimate recombination and DNA repair in eukaryotic cells. To characterize the expression of these proteins in cell types of rodent and avian brains, we combined immunocytochemistry for either Kin17 or Ogg1 proteins with glial fibrillary acidic protein (GFAP, an astrocyte marker) immunodetection on the same tissue section. Both Kin17 and Ogg1 proteins were localized in cell nuclei and were extensively distributed in neuronal populations of quail and rodent brains. However, GFAP-immunoreactive cells were never labeled by Kin17 protein. This was observed in nerve fiber tracts, in the cerebral cortex, the hippocampal formation, the hypothalamic region, and the periventricular regions of the brain of both species studied. These results were confirmed by combining in situ hybridization of kin17 mRNA and GFAP immunodetection. On the contrary, GFAP-immunoreactive cells were often labeled by the Ogg1 protein in brain structures such as fiber tracts, the cortical surface, the cerebellum, and the ependymal surface of both quail and mouse brains. Our results suggest that the expression of the Kin17 protein (observed in neurons) and that of the Ogg1 protein (observed in neurons and glial cells) is conserved in brain phylogeny.
Collapse
Affiliation(s)
- S Araneda
- Neurobiologie des états de sommeils et d'éveil, INSERM U480, Université Claude Bernard, Lyon Cedex, France.
| | | | | | | | | |
Collapse
|
84
|
Piu F, Aronheim A, Katz S, Karin M. AP-1 repressor protein JDP-2: inhibition of UV-mediated apoptosis through p53 down-regulation. Mol Cell Biol 2001; 21:3012-24. [PMID: 11287607 PMCID: PMC86930 DOI: 10.1128/mcb.21.9.3012-3024.2001] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2000] [Accepted: 02/08/2001] [Indexed: 11/20/2022] Open
Abstract
Members of the AP-1 transcription factor family, especially c-Jun and c-Fos, have long been known to mediate critical steps in the cellular response to ultraviolet (UV) irradiation. We sought to examine whether two newly discovered members of the AP-1 family, JDP-1 and JDP-2, also participate in the mammalian UV response. Here we report that JDP-2, but not JDP-1, is transiently induced upon UV challenge and that elevated levels of JDP-2 increase cell survival following UV exposure. This protective function of JDP-2 appears to be mediated through repression of p53 expression at the transcriptional level, via a conserved atypical AP-1 site in the p53 promoter.
Collapse
Affiliation(s)
- F Piu
- Department of Pharmacology, Laboratory of Gene Regulation and Signal Transduction, , University of California at San Diego, La Jolla, California 92093-0636, USA.
| | | | | | | |
Collapse
|
85
|
Marbach I, Licht R, Frohnmeyer H, Engelberg D. Gcn2 mediates Gcn4 activation in response to glucose stimulation or UV radiation not via GCN4 translation. J Biol Chem 2001; 276:16944-51. [PMID: 11350978 DOI: 10.1074/jbc.m100383200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In mammalian cells transcription factors of the AP-1 family are activated by either stress signals such as UV radiation, or mitogenic signals such as growth factors. Here we show that a similar situation exists in the yeast Saccharomyces cerevisiae. The AP-1 transcriptional activator Gcn4, known to be activated by stress signals such as UV radiation and amino acids starvation, is also induced by growth stimulation such as glucose. We show that glucose-dependent Gcn4 activation is mediated through the Ras/cAMP pathway. This pathway is also responsible for UV-dependent Gcn4 activation but is not involved in Gcn4 activation by amino acid starvation. Thus, the unusual phenomenon of activation of mitogenic pathways and AP-1 factors by contradictory stimuli through Ras is conserved from yeast to mammals. We also show that activation of Gcn4 by glucose and UV requires Gcn2 activity. However, in contrast to its role in amino acid starvation, Gcn2 does not increase eIF2alpha phosphorylation or translation of GCN4 mRNA in response to glucose or UV. These findings suggest a novel mechanism of action for Gcn2. The finding that Gcn4 is activated in response to glucose via the Ras/cAMP pathway suggests that this cascade coordinates glucose metabolism with amino acids and purine biosynthesis and thereby ensures availability of both energy and essential building blocks for continuation of the cell cycle.
Collapse
Affiliation(s)
- I Marbach
- Department of Biological Chemistry, The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | | | | | | |
Collapse
|
86
|
Schmitt-Ney M, Habener JF. CHOP/GADD153 gene expression response to cellular stresses inhibited by prior exposure to ultraviolet light wavelength band C (UVC). Inhibitory sequence mediating the UVC response localized to exon 1. J Biol Chem 2000; 275:40839-45. [PMID: 11010973 DOI: 10.1074/jbc.m007440200] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
CHOP/GADD153 is both an activating and repressing transcription factor that is markedly induced in response to a variety of cellular stresses. The CHOP/GADD153 gene was originally cloned because of its inducibility by ultraviolet light wavelength band C (UVC) and has since been found to be activated in response to many different cellular stresses. Some of the recent studies have questioned the UVC responsiveness of the CHOP gene. Contradiction in our own data led us to reexamine the UVC effects on CHOP expression. UVC is capable of strongly activating the mouse CHOP promoter in stably transfected NIH 3T3 cells but has only a modest and transient effect on the level of the CHOP messenger RNA. In addition to its positive effect on CHOP promoter activity, we show that UVC negatively affects CHOP mRNA and protein expression. Pretreatment of NIH 3T3 cells with UVC markedly attenuates the subsequent induction of CHOP mRNA by the cellular stress activators methylmethane sulfate, tunicamycin, glucose deprivation, and methionine deprivation for as long as at least 16 h. This inhibitory effect of UVC on CHOP expression in response to stress is independent of the presence or absence of p53 and does not involve mRNA degradation as opposed to the UVC effect that inhibits p21 expression seen only in the absence of p53. The target of the inhibitory effect of UVC on CHOP expression is located in the first exon of the gene, a 5'-untranslated region that is unusually conserved between different species. These findings suggest that an unknown function encoded by the 5'-untranslated region somehow modifies the response of CHOP gene transcription to UVC.
Collapse
Affiliation(s)
- M Schmitt-Ney
- Laboratory of Molecular Endocrinology, Massachusetts General Hospital, Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | |
Collapse
|
87
|
Kannouche P, Mauffrey P, Pinon-Lataillade G, Mattei MG, Sarasin A, Daya-Grosjean L, Angulo JF. Molecular cloning and characterization of the human KIN17 cDNA encoding a component of the UVC response that is conserved among metazoans. Carcinogenesis 2000; 21:1701-10. [PMID: 10964102 DOI: 10.1093/carcin/21.9.1701] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We describe the cloning and characterization of the human KIN17 cDNA encoding a 45 kDa zinc finger nuclear protein. Previous reports indicated that mouse kin17 protein may play a role in illegitimate recombination and in gene regulation. Furthermore, overproduction of mouse kin17 protein inhibits the growth of mammalian cells, particularly the proliferation of human tumour-derived cells. We show here that the KIN17 gene is remarkably conserved during evolution. Indeed, the human and mouse kin17 proteins are 92.4% identical. Furthermore, DNA sequences from fruit fly and filaria code for proteins that are 60% identical to the mammalian kin17 proteins, indicating conservation of the KIN17 gene among metazoans. The human KIN17 gene, named (HSA)KIN17, is located on human chromosome 10 at p15-p14. The (HSA)KIN17 RNA is ubiquitously expressed in all the tissues and organs examined, although muscle, heart and testis display the highest levels. UVC irradiation of quiescent human primary fibroblasts increases (HSA)KIN17 RNA with kinetics similar to those observed in mouse cells, suggesting that up-regulation of the (HSA)KIN17 gene after UVC irradiation is a conserved response in mammalian cells. (HSA)kin17 protein is concentrated in intranuclear focal structures in proliferating cells as judged by indirect immunofluorescence. UVC irradiation disassembles (HSA)kin17 foci in cycling cells, indicating a link between the intranuclear distribution of (HSA)kin17 protein and the DNA damage response.
Collapse
Affiliation(s)
- P Kannouche
- Laboratoire de Génétique de la Radiosensibilité, Département de Radiobiologie et de Radiopathologie, Direction des Sciences du Vivant, Centre d'Etudes de Fontenay-aux-Roses, CEA, 92265 Fontenay-aux-Roses, France
| | | | | | | | | | | | | |
Collapse
|