1: Cancer Cell. 2005 Aug;8(2):155-67. Comment in: Cancer Cell. 2005 Aug;8(2):91-3. Neuronal apoptosis linked to EglN3 prolyl hydroxylase and familial pheochromocytoma genes: developmental culling and cancer. Lee S, Nakamura E, Yang H, Wei W, Linggi MS, Sajan MP, Farese RV, Freeman RS, Carter BD, Kaelin WG Jr, Schlisio S. Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA. Germline NF1, c-RET, SDH, and VHL mutations cause familial pheochromocytoma. Pheochromocytomas derive from sympathetic neuronal precursor cells. Many of these cells undergo c-Jun-dependent apoptosis during normal development as NGF becomes limiting. NF1 encodes a GAP for the NGF receptor TrkA, and NF1 mutations promote survival after NGF withdrawal. We found that pheochromocytoma-associated c-RET and VHL mutations lead to increased JunB, which blunts neuronal apoptosis after NGF withdrawal. We also found that the prolyl hydroxylase EglN3 acts downstream of c-Jun and is specifically required among the three EglN family members for apoptosis in this setting. Moreover, EglN3 proapoptotic activity requires SDH activity because EglN3 is feedback inhibited by succinate. These studies suggest that failure of developmental apoptosis plays a role in pheochromocytoma pathogenesis. PMID: 16098468 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 2: Gene. 2004 Oct 27;341:209-18. E2F sites in the Op18 promoter are required for high level of expression in the human prostate carcinoma cell line PC-3-M. Polzin RG, Benlhabib H, Trepel J, Herrera JE. Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, 321 Church St. S.E., 6-155 Jackson Hall, Minneapolis, MN 55455, USA. Op18 (Oncoprotein 18, Stathmin) is an important mitotic regulator that is highly expressed in many cancers including the metastatic prostate carcinoma cell line PC-3-M. Recent studies indicate that antisense-mediated down-regulation of Op18 can inhibit cellular proliferation. However, the transcriptional mechanisms responsible for its normal regulation and for its high level of expression in proliferating cells remain poorly understood. In the study presented here, we have characterized transcription factor binding sites that together account for nearly 80% of the Op18 expression in PC-3-M cells. The 5' flanking region of the Op18 gene contains four putative E2F sites located at -700 (site 1), -28 (site 2), -19 (site 3), and +720 (site 4) relative to the transcriptional start site. E2F has been implicated in both the c-Jun-mediated up-regulation and the doxorubicin-induced repression of Op18 expression. We have used promoter-reporter assays and mobility shift assays to functionally examine each of these E2F sites. Mutagenesis studies indicate that all sites contribute to the basal expression of Op18. Mutagenesis of either site 1 or 4 reduced the reporter activity by 40%, mutagenesis of site 2 reduced reporter activity by 20%, and mutations in site 3 had no effect on reporter activity. Combinatorial mutagenesis indicates that site 1 and 4 function independently, whereas site 2 functions synergistically with either site 3 or 4. Mobility shift assays indicate that all elements bind factors in the nuclear extracts of PC-3-M cells. Characterization of the sites show that site 1, though a positive element, is not E2F specific; sites 2 and 3 may contain an overlapping binding site for E2F and NF1; and site 4, which resides in intron 1, is the only site shown to interact exclusively with E2F. These studies suggest that the overexpression of Op18 in PC-3-M cells is mediated predominantly through the E2F family of transcription factors. PMID: 15474303 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 3: J Virol. 2004 Jul;78(14):7590-601. Identification and characterization of cis-acting elements residing in the walleye dermal sarcoma virus promoter. Hronek BW, Meagher A, Rovnak J, Quackenbush SL. Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA. Walleye dermal sarcoma virus (WDSV) is a complex retrovirus found associated with tumors that appear and regress on a seasonal basis. There are quantitative and qualitative differences in the amount of virus expression between developing and regressing tumors. To understand the role of host cell factors in WDSV expression, DNase I footprint analysis, electrophoretic mobility shift assays (EMSA), and reporter gene assays were employed. DNase I footprint analysis of the U3 region of the WDSV long terminal repeat with nuclear extract prepared from a walleye cell line revealed protection of an Oct1, AP1, Whn, and two E4BP4 sites. Additionally, three regions that contained no putative transcription factor binding sites were protected. EMSA confirmed the specific binding of the protected sites and revealed three additional sites, NF1, AP3, and LVa, not protected in DNase I footprint analysis. Site-directed mutagenesis of the individual sites, in the context of a luciferase reporter plasmid, revealed that the NF1, Oct1, AP1, E4BP4#2, AP3, and LVa sites contributed to transcription activation driven by the WDSV U3 region. Mutation of Novel#2 resulted in an increase in luciferase activity, suggesting the Novel#2 site may function to bind a negative regulator of transcription. Anti-Jun and anti-Fos antiserum specifically inhibited protein-DNA complex formation, indicating the presence of c-Jun and c-Fos in the walleye cell nuclear extracts and their participation in binding to the AP1 site. Interestingly, degenerative 15-bp repeats found in the U3 region are differentially protected in DNase I footprint analysis by the walleye cell line nuclear extract and regressing-tumor nuclear extract. EMSA utilizing the 15-bp repeat probe revealed that there are similarities of binding with W12 cell and developing-tumor nuclear extracts and that the binding differs from that observed with regressing-tumor nuclear extract. PMID: 15220434 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 4: J Biol Chem. 2002 Sep 20;277(38):35232-9. Epub 2002 Jul 1. Role of AP-1 in the coordinate induction of rat glutamate-cysteine ligase and glutathione synthetase by tert-butylhydroquinone. Yang H, Zeng Y, Lee TD, Yang Y, Ou X, Chen L, Haque M, Rippe R, Lu SC. Division of Gastroenterology and Liver Diseases, University of Southern California Liver Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA. GSH synthesis occurs via two enzymatic steps catalyzed by glutamate-cysteine ligase (GCL, made up of two subunits) and GSH synthetase (GS). Recently, we described coordinate induction of GCL subunits and GS. To study GS transcriptional regulation, we have cloned and characterized a 2.2-kb 5'-flanking region of the rat GS (GenBank accession number AF333982). One transcriptional start site is located at 51 nucleotides upstream of the translational start site. The rat GS promoter drove efficiently luciferase expression in H4IIE cells. Sequential deletion analysis revealed DNA regions that are involved in positive and negative regulation. One repressor identified was NF1. tert-Butylhydroquinone (TBH) exerted a dose- and time-dependent increase in the mRNA level and promoter activity of both GCL subunits and GS. TBH increased protein binding to several regions of the GS promoter, c-jun expression, and activator protein 1 (AP-1) binding activity to several of the putative AP-1-binding sites of the GS promoter. Blocking AP-1 binding with dominant-negative c-jun led to decreased basal expression and significantly blocked the TBH-induced increase in promoter activity and mRNA level of all three genes. In conclusion, AP-1 is required for basal expression of GCL and GS; while NF1 serves as a repressor of GS, increased AP-1 transactivation is the predominant mechanism for coordinate induction of GCL and GS expression by TBH. PMID: 12093805 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 5: Eur J Biochem. 1998 Sep 15;256(3):541-9. Phorbol ester causes ligand-independent activation of the androgen receptor. Darne C, Veyssiere G, Jean C. Centre National de la Recherche Scientifique Unite Mixte de Recherche 6547, Universite Blaise Pascal-Clermont-Ferrand II, Aubiere, France. We have examined the human androgen receptor (hAR) for its ability to activate AR-dependent transcription of a transgene in a ligand-independent manner. The transcriptional activity was determined by analysis of chloramphenicol acetyltransferase (CAT) activity in T47D cells cotransfected with a plasmid expressing the hAR and a natural AR-regulated promoter (the MVDP androgen-dependent enhancer) ligated to the reporter CAT gene. In this study, the effects of the protein kinase C (PKC) activator 12-O-tetradecanoyphorbol-13 acetate (TPA) on AR activity were tested. We demonstrated that in the absence of androgen, TPA enhanced AR-mediated transactivation by 10-12-fold. This effect was specific of the PKC pathway since stimulation to the PKA pathway did not activate the unliganded AR. This ligand-independent pathway can function through another androgen-regulated promoter as shown by the use of the mouse mammary tumor virus MMTV-CAT reporter. The human glucocorticoid receptor (hGR) and the rabbit progesterone receptor (rPR) could not be activated by TPA, indicating that the effects are not universal for steroid receptors. A reporter plasmid containing the MVDP androgen response element (ARE) in front of the thymidine kinase promoter ligated to the CAT gene was activated by DHT but not by TPA, indicating that the context of the natural promoter is critical for ligand-independent activation of the AR. Exogenous c-jun enhanced transcriptional activation by the AR in a ligand-dependent manner, but had no effect in the absence of DHT. Base pair substitutions in both AR-binding (5'-TGTTCT-3' to 5'-TTTTTT-3') and NF1-binding (5'-GTGGCTG-3' to 5'-GTTTTTG-3') sites resulted in a loss of TPA responsiveness. Our results suggest that ligand-independent activation of the AR by TPA results from interaction of unliganded AR with other proteins in the transcription machinery. PMID: 9780230 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 6: Proc Natl Acad Sci U S A. 1998 Apr 28;95(9):5139-44. A role for Pak protein kinases in Schwann cell transformation. Tang Y, Marwaha S, Rutkowski JL, Tennekoon GI, Phillips PC, Field J. Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA. Neurofibromatosis type 1 (NF1), a common autosomal dominant disorder caused by loss of the NF1 gene, is characterized clinically by neurofibromas and more rarely by neurofibrosarcomas. Neurofibromin, the protein encoded by NF1, possesses an intrinsic GTPase accelerating activity for the Ras proto-oncogene. Through this activity, it is a negative regulator of Ras. The Pak protein kinase is a candidate for a downstream signaling protein that may mediate Ras signals because it is activated by Rac and Cdc42, two small G proteins required for Ras signaling. Here, we use Pak mutants to explore the role of Pak in Ras signaling in Schwann cells, the cells affected in NF1. Whereas an activated Pak mutant does not transform cells, dominant negative Pak mutants are potent inhibitors of Ras transformation of rat Schwann cells and of a neurofibrosarcoma cell line from an NF1 patient. Although activated Pak stimulated jun-N-terminal kinase, inhibition of Ras transformation by dominant negative Pak did not require inhibition of jun-N-terminal kinase. Instead, the Pak mutants appeared to inhibit transformation by preventing Ras activation of the ERK/mitogen-activated protein kinase cascade. These results have implications for our understanding of NF1 because a neurofibrosarcoma cell line derived from a patient with NF1 was reverted by stable expression of the Pak dominant negative mutants. PMID: 9560242 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 7: Oncogene. 1998 Mar 26;16(12):1525-31. The OMgp gene, a second growth suppressor within the NF1 gene. Habib AA, Gulcher JR, Hognason T, Zheng L, Stefansson K. Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02115, USA. The Oligodendrocyte-Myelin glycoprotein gene (OMgp) is placed within an intron of the NF1 gene. Neurofibromin, the product of NF1, acts as a RasGAP and suppresses growth; inactivating mutations in NF1 lead to neurofibromatosis type 1. We report that OMgp also has growth suppressive effects and downregulates mitogenic signaling pathways closely related to those influenced by neurofibromin. Overexpression of OMgp alters mitogenic signaling in NIH3T3 fibroblasts. Cells overexpressing OMgp grow more slowly in serum compared to controls and show a partial G1 block upon cell cycle analysis. PDGF is the primary mitogen for fibroblasts in serum. Overexpression of OMgp alters PDGF signaling in fibroblasts which results in a block of mitogenic signaling. PDGF induced activation of c-Src is blocked, as is the induction of c-Myc and c-Fos, while tyrosine phosphorylation of the PDGFbeta receptor, PLCgamma1 and induction of c-Jun are intact. Although a number of genes embedded within other genes have been described, the biological significance of this arrangement remains unknown. We demonstrate here that structurally unrelated products of two such genes may exercise closely related functions. Our data also raise the possibility of a role for OMgp in disorders of cell proliferation such as NF1. PMID: 9569019 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 8: Yakugaku Zasshi. 1996 Jul;116(7):505-18. [Regulation mechanism of specific expression of tumor marker gene during carcinogenesis] [Article in Japanese] Imagawa M. Faculty of Pharmaceutical Sciences, Osaka University, Japan. Rat glutathione transferase P (GST-P) is expressed at low levels in the normal liver but becomes highly expressed in hyperplastic nodules and in hepatocellular carcinomas during chemical hepatocarcinogenesis. To understand the regulation mechanisms of this gene, we have characterized the 5'-flanking region and have found that GST-P gene is regulated by at least two elements: one is a strong enhancer and the other is a silencer. GST-P enhancer I (GPEI), located at -2.5 Kb, consists of two TPA-responsive element (TRE)-like sequences that are palindromically oriented with 3 bp in between. It is well known that TRE is activated by two nuclear oncogenes, c-Jun and c-Fos. Although GPEI is trans-activated by these oncogenes, it is also active in F9 embryonal carcinoma cells that lack c-Jun protein, suggesting that it can function with some trans-activator other than AP-1 (c-Jun/c-Fos heterodimer). Indeed, another protein is identified from the F9 nuclear extract. We have also identified a silencer element at 300 bp upstream from the cap site. There are several cis-elements in this region and at least three trans-acting factors bind to these elements. We purified SF-A (silencer factor A) which binds to several regions in this silencer, and determined the partial amino acid sequence. Interestingly, SF-A seemed to be a related protein to NF1 (nuclear factor 1) which is an activator for the transcription and DNA replication. Another factor SF-B (silencer factor B) has been cloned and found to be the same as LIP (liver inhibitory protein) which is a competitor for LAP (liver activator protein), both are from the same gene designated as C/EBP beta. By transfection analysis using GAL4 DNA binding domain we found LIP is not only a competitor but a direct repressor. In the normal liver, another C/EBP family member, C/EBP alpha also acts as a negative regulator, and this expression decreases during hepatocarcinogenesis, resulting in the loss of silencer function. We carried out the carcinogenesis experiments using transgenic rats harboring a chloramphenicol acetyltransferase (CAT) reporter gene with -2900 to + 59 of the GST-P gene. Liver foci and nodules produced by chemical carcinogens were found to express high levels CAT activity by both CAT assay and immunohistochemical study, while normal liver cells did not express any CAT activity. These results demonstrate that the GST-P gene is trans-activated locus-independently during rat hepatocarcinogenesis. Moreover, the similar results were obtained using transgenic rats carrying GPEI-CAT, indicating that GPEI is an important cis-element for activation of GST-P gene during hepatocarcinogenesis. Publication Types: Review Review, Tutorial PMID: 8831256 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 9: Virus Genes. 1996;13(1):45-52. Induction of the HPV16 enhancer activity by Jun-B and c-Fos through cooperation of the promoter-proximal AP-1 site and the epithelial cell type--specific regulatory element in fibroblasts. Kikuchi K, Taniguchi A, Yasumoto S. Laboratory of Molecular and Cellular Biology, Kanagawa Cancer Center Research Institute, Yokohama, Japan. The epithelial cell type-specific enhancer of the human papillomavirus (HPV) type 16 termed the long control region (LCR) carries three AP-1 binding sites. We investigated the roles of the AP-1 sites for transactivation of the LCR by Jun-B that may be a cell type specific-transactivator for the HPVs in human fibroblasts in which expression of the endogenous Jun-B gene is low. Transient expression of Jun-B alone poorly activated transcriptional activity of the LCR. However, when combined with c-Fos, Jun-B activated the LCR. The promoter-proximal AP-1 site was required for transactivation of the LCR by Jun-B:c-Fos, but the site itself was not sufficient for the maximal response. The proposed cell type specific--regulatory element that harbors binding sites for NF1 as well as TEF-1 and PEA3 motifs, but neither other AP-1 sites nor the proximal enhancer region, could augment the transcriptional response of the promoter-proximal AP-1 site to Jun-B:c-Fos. PMID: 8938978 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 10: Virology. 1995 Aug 1;211(1):184-97. Transcriptional activity of human papillomavirus type 31b enhancer is regulated through synergistic interaction of AP1 with two novel cellular factors. Kyo S, Tam A, Laimins LA. Department of Microbiology-Immunology, Northwestern University, Chicago, Illinois 60611, USA. Transcription of human papillomaviruses (HPV) is regulated by enhancer sequences located in the upstream regulatory region. The factors regulating expression of one of the high risk genital HPV types, HPV 31b, were investigated using transient expression and protein binding assays. A region of 262 base pairs in length was identified as the minimal functional enhancer and a series of five protected binding sites were observed by footprint analyses. Electrophoretic mobility shift assays demonstrated that AP1, Oct-1, as well as three novel factors bound these sequences. Mutational analyses indicated that AP1 synergistically activated the HPV31b enhancer together with either of two novel factors. One of these novel factors bound a sequence similar to an NF1 site but was distinct from NF-1. The second factor bound sequences bearing similarity to KRF-1 binding sites which have previously been characterized in HPV 18. Competition binding assays demonstrated that this factor was not identical to KRF-1. Additional studies implicated Oct-1 as a negative regulator of HPV 31b expression as mutation of Oct-1 binding sequences resulted in an increase in viral expression. None of the factors observed to be important for HPV 31b enhancer activity was found exclusively in epithelial cells and instead were detected in a variety of cell types. Of these factors, AP1 binding correlated most strongly with enhancer function in a variety of cell types, implicating it as a principal regulator of HPV expression. Variations in the constituents of the AP1 complex that bind the HPV 31b enhancer were also observed in different cell types, suggesting that changes in the distribution of jun proteins may play a significant role in determining the tropism of HPV. These results indicate that AP1 may be a common regulator for various HPV types and that it contributes to enhancer specificity. In addition, a set of novel factors, which may be specific for each HPV type, act synergistically with AP1 for full activation of the enhancer. PMID: 7645210 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 11: Proc Natl Acad Sci U S A. 1995 May 9;92(10):4497-501. Inhibition of AP-1 binding and transcription by gold and selenium involving conserved cysteine residues in Jun and Fos. Handel ML, Watts CK, deFazio A, Day RO, Sutherland RL. Cancer Biology Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia. Gold(I) salts and selenite, which have diverse therapeutic and biological effects, are noted for their reactivity with thiols. Since the binding of Jun-Jun and Jun-Fos dimers to the AP-1 DNA binding site is regulated in vitro by a redox process involving conserved cysteine residues, we hypothesized that some of the biological actions of gold and selenium are mediated via these residues. In electrophoretic mobility-shift analyses, AP-1 DNA binding was inhibited by gold(I) thiolates and selenite, with 50% inhibition occurring at approximately 5 microM and 1 microM, respectively. Thiomalic acid had no effect in the absence of gold(I), and other metal ions inhibited at higher concentrations, in a rank order correlating with their thiol binding affinities. Cysteine-to-serine mutants demonstrated that these effects of gold(I) and selenite require Cys272 and Cys154 in the DNA-binding domains of Jun and Fos, respectively. Gold(I) thiolates and selenite did not inhibit nonspecific protein binding to the AP-1 site and were at least an order of magnitude less potent as inhibitors of sequence-specific binding to the AP-2, TFIID, or NF1 sites compared with the AP-1 site. In addition, 10 microM gold(I) or 10 microM selenite inhibited expression of an AP-1-dependent reporter gene, but not an AP-2-dependent reporter gene. These data suggest a mechanism regulating transcription factor activity by inorganic ions which may contribute to the known antiarthritic action of gold and cancer chemoprevention by selenium. PMID: 7753832 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 12: J Biol Chem. 1994 Sep 30;269(39):24321-7. Activation of nuclear factor kappa B and oncogene expression by 12(R)-hydroxyeicosatrienoic acid, an angiogenic factor in microvessel endothelial cells. Laniado-Schwartzman M, Lavrovsky Y, Stoltz RA, Conners MS, Falck JR, Chauhan K, Abraham NG. Department of Pharmacology, New York Medical College, Valhalla 10595. 12(R)-Hydroxy-5,8,14(Z,Z,Z)-eicosatrienoic acid (12(R)-HETrE) is an arachidonic acid metabolite formed by the corneal epithelium of several species, porcine leukocytes, and human and rat epidermal cells. It is a potent, stereospecific proinflammatory and angiogenic factor and its synthesis is increased manyfold in inflamed tissues, e.g. cornea and skin. It is possible that the angiogenic activity of 12(R)-HETrE is due to a direct mitogenic effect on microvessel endothelial cells via yet to be elucidated cellular and molecular mechanisms. In the present study, we demonstrated the ability of 12(R)-HETrE to stimulate the growth of quiescent endothelial cells in a time- and concentration-dependent manner with a maximal effect at 0.1 nM. This effect was highly stereospecific since its enantiomer, 12(S)-HETrE, had no effect within the same concentration range. Northern blot analysis and transient transfection experiments with chloramphenicol acetyltransferase constructs of oncogene promoter regions demonstrated significant increases over control (0.5% fetal calf serum) in c-myc-, c-jun, and c-fos mRNA levels and expression in cells treated with 0.1 nM 12(R)-HETrE. Electrophoretic mobility shift assay of nuclear protein extracts from cells treated with 12(R)-HETrE with specific radiolabeled oligonucleotides corresponding to known transcriptional binding sites, including AP-1, AP-2, SP1, TRE, NF kappa B, TFIID, OKT1, CREB, CTF/NF1, and GRE demonstrated a markedly rapid and specific increase in the binding activity of NF kappa B and to a lesser extent, AP-1. No significant increase was observed in the binding of other transcription factors assayed as compared to control (untreated) cells. Since the protooncogenes (c-fos, c-jun, and c-myc) are immediate early response genes that are implicated in the process of cell proliferation and differentiation, and activation of certain transcription factors, in particular NF kappa B, is associated with the immediate response of the cell to an injury, we propose that 12(R)HETrE's mitogenic and angiogenic activities are mediated, in part, via the activation of NF kappa B and expression of these protooncogenes. PMID: 7523372 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 13: Bioessays. 1994 Jul;16(7):489-96. Regulation of the Ras signalling network. Maruta H, Burgess AW. Ludwig Institute for Cancer Research, Melbourne, Australia. The mitogenic action of cytokines such as epidermal growth factor (EGF) or platelet derived growth factor (PDGF) involves the stimulation of a signal cascade controlled by a small G protein called Ras. Mutations of Ras can cause its constitutive activation and, as a consequence, bypass the regulation of cell growth by cytokines. Both growth factor-induced and oncogenic activation of Ras involve the conversion of Ras from the GDP-bound (D-Ras) to the GTP-bound (T-Ras) forms. T-Ras activates a network of protein kinases including c-Mos, c-Raf-1 and MAP kinase. Eventually the activation of MAP kinase leads to the activation of the elongation factor 4E and several transcription factors such as c-Jun, c-Myc and c-Fos. There are several modulators of Ras activity, such as the GTPase activating proteins (GAP1 and NF1), which stimulate the conversion of T-Ras to D-Ras. A series of small NF1 fragments, which bind T-Ras, as well as truncated forms of derivatives of c-Raf-1, c-Jun and c-Myc, are capable of blocking the T-Ras-activated mitogenesis in a competitive manner. These agents offer a unique opportunity to control the proliferation of T-Ras-associated tumors, which represent more than 30% of total human carcinomas. Publication Types: Review Review, Tutorial PMID: 7945277 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 14: J Biol Chem. 1994 Jun 10;269(23):16512-7. Sphingosine 1-phosphate, a novel signaling molecule, stimulates DNA binding activity of AP-1 in quiescent Swiss 3T3 fibroblasts. Su Y, Rosenthal D, Smulson M, Spiegel S. Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, Washington, District of Columbia 20007. Sphingosine and sphingosine 1-phosphate, metabolites of sphingolipids, stimulate cell proliferation in quiescent Swiss 3T3 fibroblasts and induce transient increases in intracellular free calcium (Zhang, H., Desai, N. N., Olivera, A., Seki, T., Brooker, G., and Spiegel, S. (1991) J. Cell Biol. 114, 155-167). However, little is yet known of the nuclear events that follow the early responses induced by sphingolipid metabolites. Using a gel retardation assay, we found that specific DNA binding activity of activator protein-1 (AP-1) was markedly increased after treatment of quiescent Swiss 3T3 fibroblasts with sphingosine 1-phosphate and sphingosine. The DNA binding specificity of AP-1 was confirmed with competing probes containing consensus sequences of AP-1, AP-2, AP-3, SP-1, and NF1/CTF. The c-fos gene product was detected in the AP-1 complex using anti-c-Fos antibody. The dose response for stimulation of DNA binding activity of AP-1 by sphingosine 1-phosphate correlated closely with its effect on DNA synthesis. Furthermore, an inhibitor of sphingosine kinase, DL-threo-dihydrosphingosine, which inhibits sphingosine-induced DNA synthesis and the formation of sphingosine 1-phosphate, also inhibited sphingosine-stimulated AP-1 DNA binding activity. This result further supports our proposal that sphingosine 1-phosphate mediates the mitogenic effect of sphingosine. Our results indicate that sphingosine 1-phosphate-induced DNA synthesis and cell division may result from activation of AP-1 protein, linking signal transduction by sphingolipid metabolites to gene expression. PMID: 8206962 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 15: Mol Cell Biol. 1993 Apr;13(4):2497-503. Differential regulation of cellular activities by GTPase-activating protein and NF1. al-Alawi N, Xu G, White R, Clark R, McCormick F, Feramisco JR. Department of Medicine and Pharmacology, UCSD Cancer Center, La Jolla 92093. The regulation of the GTPase activity of the Ras proteins is thought to be a key element of signal transduction. Ras proteins have intrinsic GTPase activity and are active in signal transduction when bound to GTP but not following hydrolysis of GTP to GDP. Three cellular Ras GTPase-activating proteins (Ras-gaps) which increase the GTPase activity of wild-type (wt) Ras but not activated Ras in vitro have been identified: type I and type II GAP and type I NF1. Mutations of wt Ras resulting in lowered intrinsic GTPase activity or loss of response to cellular Ras-gap proteins are thought to be the primary reason for the transforming properties of the Ras proteins. In vitro assays show type I and type II GAP and the GAP-related domain of type I NF1 to have similar biochemical properties with respect to activation of the wt Ras GTPase, and it appears as though both type I GAP and NF1 can modulate the GTPase function of Ras in cells. Here we report the assembling of a full-length coding clone for type I NF1 and the biological effects of microinjection of Ras and Ras-gap proteins into fibroblasts. We have found that type I GAP, type II GAP, and type I NF1 show markedly different biological activities in vivo. Coinjection of type I GAP or type I NF1, but not type II GAP, with wt Ras abolished the ability of wt Ras to induce expression from an AP-1-controlled reporter gene. We also found that serum-stimulated DNA synthesis was reduced by prior injection of cells with type I GAP but not type II GAP or type I NF1. These results suggest that type I GAP, type II GAP, and type I NF1 may have different activities in vivo and support the hypothesis that while type I forms of GAP and NF1 may act as negative regulators of wt Ras, they may do so with differential efficiencies. PMID: 8455625 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 16: Oncogene. 1991 Aug;6(8):1417-22. Interactions between adenovirus E1A and members of the AP-1 family of cellular transcription factors. Maguire K, Shi XP, Horikoshi N, Rappaport J, Rosenberg M, Weinmann R. Wistar Institute, Philadelphia, Pennsylvania 19104. We have studied interactions between bacterially produced E1A linked to Sepharose and the various DNA-binding proteins present in HeLa cell nuclear extracts (NE). DNA-binding activities and cross-reactive polypeptides recognizing the cAMP-responsive element (CRE) and the activator protein 1 (AP1) sites were bound to the E1A column, whereas nuclear factor 1 (NF1) and the activator protein 2 (AP2) DNA-binding activities were not retained by E1A. The binding activities that were retained belonged to the CRE and JUN protein family, as judged by Western blot analysis. Authentic CRE-BP1, c-Jun and c-Fos proteins produced by in-vitro translation also bound to the E1A column. However, efficient binding of in-vitro-translated CRE-BP1 and c-Fos proteins to E1A required preincubation with NE. We show here that immobilized E1A sequesters several cellular upstream transcription activators, and suggest a role for members of the AP1 family of transcription factors in E1A-mediated gene regulation. PMID: 1832215 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 17: Oncogene. 1990 Sep;5(9):1285-90. Protein-binding elements in the promoter region of the mouse p53 gene. Ginsberg D, Oren M, Yaniv M, Piette J. Department of Chemical Immunology, Weizmann Institute of Science, Rehovot, Israel. p53 is a cellular protein whose expression plays a crucial role in the regulation of cell proliferation and of neoplastic processes. p53 mRNA levels in mouse fibroblasts can be elevated in response to TPA and to serum stimulation. The promoter region of the p53 gene contains a conserved element which is highly homologous to the consensus AP1 binding site (7/8 matching bases). This AP1-like site, denoted the PF1 site, confers upon a heterologous promoter ability to respond to elevated expression of c-jun. Furthermore, the PF1 site binds protein(s) in a specific and serum-induced manner. Unexpectedly, this factor is most probably not AP1, as evident from the inability of an authentic AP1 site to compete the binding efficiently, as well as from the failure of purified AP1 to bind to the PF1 site. Hence, PF1 may be a novel AP1-related transcription factor. In addition, the 5' region of the p53 gene also contains an NF1 binding site, whose location suggests a possible regulatory role. PMID: 2216454 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 18: Biochem Biophys Res Commun. 1990 Jul 16;170(1):140-6. Transcription factor levels in medullary thyroid carcinoma cells differentiated by Harvey ras oncogene: c-jun is increased. Nelkin BD, Borges M, Mabry M, Baylin SB. Oncology Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231. In the TT cell line of human medullary thyroid carcinoma, the viral Harvey ras (v-rasH) oncogene induces differentiation, marked by morphological changes, diminution of growth, and increased expression of the calcitonin gene. Here, we show that the transcriptional factor c-jun is increased during v-rasH induced differentiation of TT cells both at the mRNA and functional protein levels. In contrast, nuclear proteins with binding activities related to AP2, AP3, NF1/CTF, and Sp1 were unchanged in v-rasH differentiated TT cells. PMID: 2115330 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 19: J Virol. 1990 May;64(5):2004-13. Adenovirus early region 3 promoter regulation by E1A/E1B is independent of alterations in DNA binding and gene activation of CREB/ATF and AP1. Kornuc M, Kliewer S, Garcia J, Harrich D, Li C, Gaynor R. Department of Medicine, University of California, Los Angeles School of Medicine 90024. Transcription of the adenovirus early region 3 promoter is strongly induced by the adenovirus E1A protein. Previous DNase I footprinting has indicated that four regions in this promoter serve as binding sites for HeLa nuclear proteins. These include binding sites for NF-1 (site IV), AP1 (site III), CREB/activating transcription factor (ATF) (site II), and TATA (site I). To determine the relative importance of these sites in both the in vivo and in vitro transcriptional regulation of the E3 promoter, oligonucleotide-directed mutagenesis of these sites was performed. Each of these constructs was assayed by transfection onto HeLa cells in the presence of either dl434, an E1A/E1B deletion mutant, or wild-type adenovirus. Mutations of either the ATF- or AP1-binding sites but not the TATA- and NF1-binding sites resulted in severe decreases in both basal and E1A/E1B-induced transcriptional levels. These constructs were also assayed in in vitro transcription assays with cellular extracts prepared from dl434-infected or wild-type-adenovirus-infected HeLa cells. The wild-type E3 promoter was transcribed approximately 30 times more efficiently in extracts containing the E1A/E1B proteins compared with extracts lacking these proteins. Mutations of either the TATA element, the ATF site, or the AP1-binding site decreased both basal and E1A/E1B-induced transcriptional levels. Gel retardation analysis using these extracts indicated that the binding to ATF, AP1, or NF1 oligonucleotides was not altered in the presence of the E1A/E1B proteins compared with extracts lacking these proteins. Northern (RNA) blot analysis of c-jun and CREB RNA prepared from wild-type adenovirus and dl434-infected cells indicated that the levels of these RNAs were not altered by the E1A/E1B proteins. Immunoprecipitation of AP1 and CREB from both dl434- and wild-type-adenovirus-infected cells indicated that the amounts of these proteins were not significantly altered. These results suggest that E1A/E1B-induced activation of the E3 promoter does not involve activation of transcription factor genes nor a change in the DNA binding activity of important promoter-binding components. Our results are consistent with a model in which the E1A/E1B proteins either directly or indirectly alter the interactions of factors that bind to the basal E3 promoter transcription complex, thereby inducing transcription. PMID: 2139139 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 20: Nucleic Acids Res. 1990 Feb 11;18(3):465-70. Transcriptional activation of human papillomavirus 16 by nuclear factor I, AP1, steroid receptors and a possibly novel transcription factor, PVF: a model for the composition of genital papillomavirus enhancers. Chong T, Chan WK, Bernard HU. Institute of Molecular and Cell Biology, National University of Singapore, Kent Ridge. Human papillomavirus 16 (HPV-16), which is involved in genital carcinogenesis, contains an enhancer of transcription that is activated by cellular factors rather than by the viral E2 proteins. The activity resides on a 232 bp segment with 5 binding sites for nuclear factor 1 (NF1), 2 for AP1, and 1 for steroid receptors. Deletions and point mutations show that the constitutive enhancer and the steroid response depend on NF1 sites located 5' or 3' of a 65 bp fragment with AP1 sites that by itself shows little activity. Enhancement through a fragment with AP1 and NF1 sites is strongly reduced by mutation of the AP1 sites, or by mutation of the sequence AGGCACATAT. Sequence comparison and footprint analysis make it likely that this sequence binds a novel transcription factor which we call PVF. Fragments with one or several binding sites only for NF1, or AP1, or PVF exhibit little enhancement by themselves, suggesting the functional dependence of the HPV-16 enhancer on the cooperation of these factors. A comparison of our findings with the genomes and transcription factor binding sites of HPV-6, 11, 18, 31 and 33 lead us to propose a model of the composition of enhancers of genital papillomaviruses. PMID: 2155400 [PubMed - indexed for MEDLINE] ---------------------------------------------------------------