1: Gastroenterology. 2005 Nov;129(5):1602-15. Silencing of Caspase-8 in Murine Hepatocellular Carcinomas Is Mediated via Methylation of an Essential Promoter Element. Liedtke C, Zschemisch NH, Cohrs A, Roskams T, Borlak J, Manns MP, Trautwein C. Department of Medicine III, University Hospital Aachen, Aachen University, Aachen, Germany. Background & Aims: Caspase-8 is the apical caspase essential for triggering Fas-induced apoptosis. In this study, we investigated caspase-8 expression in hepatocellular carcinomas (HCCs) using recently described HCC mouse models (c-myc and IgEGF transgenes). Methods: HCCs were isolated from c-myc and IgEGF transgenic animals. Expression of caspase-8 was monitored by reverse-transcription polymerase chain reaction. The murine caspase-8 promoter was characterized by luciferase-reporter analysis and the analysis of promoter methylation was performed by bisulfite genomic sequencing. Results: In HCCs investigated, we frequently found a lack of caspase-8 messenger RNA expression. Genomic deletions at the caspase-8 locus did not contribute to caspase-8 silencing. We examined tumor-derived promoter sequences and found significant hypermethylation at distinct CpG sites. In parallel, we characterized the murine caspase-8 promoter and identified a 30-bp promoter element that is indispensable for basal promoter activity. This minimal promoter element contained SP1 binding motifs that are colocalized with CpG sites and were methylated in tumor-derived promoter sequences. Electrophoretic mobility shift assay analysis showed that methylation of these SP1 sites is sufficient to prevent SP1 complex formation. To support our data, we mimicked the methylation pattern of a tumor-derived caspase-8 promoter in vitro using CpG methylase and found a strong reduction of promoter activity. Conclusions: We show that HCCs are correlated frequently with silencing of caspase-8 expression and provide data suggesting that caspase-8 silencing is a direct consequence of inhibiting SP1-dependent transactivation caused by CpG methylation at its essential binding sites in the promoter region. Our data support the hypothesis that inhibition of apoptosis triggers hepatocarcinogenesis. PMID: 16285959 [PubMed - in process] --------------------------------------------------------------- 2: Mol Pharmacol. 2005 Oct 7; [Epub ahead of print] Activation of ERK signaling by epidermal growth factor mediates c-Jun activation and p300 recruitment in keratin 16 gene expression. Wang YN, Chen YJ, Chang WC. National Cheng Kung University. In studies of gene regulation of keratin 16, we reported previously that Sp1 shows a functional cooperation with c-Jun and coactivators p300/CBP in driving the transcriptional regulation of EGF-induced keratin 16 gene expression. In the present study, we found that the stimulated expression of keratin 16 by EGF was mediated mainly through the MEK-ERK signaling pathway. Ser63 and Ser73 on the c-Jun NH2-terminal transactivation domain could be phosphorylated in cells treated with EGF, nevertheless, we found surprisingly that the c-Jun COOH-terminus played a pivotal role in EGF-induced expression of keratin 16. The activation of keratin 16 by EGF treatment could not be enhanced by overexpression of myc-c-JunK3R, in which three putative acetylation lysine residues on the c-Jun COOH-terminus were all mutated into arginines, suggesting that c-Jun acetylation on the COOH-terminus might partially play a functional role in this system. In addition, by using a chromatin immunoprecipitation assay and a DNA affinity precipitation assay, EGF treatment up-regulated the p300 recruitment through ERK signaling to the promoter region in regulating keratin 16 transcriptional activity. Furthermore, the enhancement of acetyl-histone H3 to the keratin 16 chromatin promoter induced by EGF was also mediated via ERK activation. In conclusion, these results strongly suggested both of c-Jun induction and p300 recruitment to gene promoter, mediated through ERK activation, played an essential role in regulating keratin 16 gene expression by EGF. p300 mediated and regulated EGF-induced keratin 16 gene expression, at least in part through multiple mechanisms including a selective acetylation of c-Jun and histone H3. PMID: 16214953 [PubMed - as supplied by publisher] --------------------------------------------------------------- 3: J Biol Chem. 2005 Oct 3; [Epub ahead of print] TIS7 regulation of the beta -catenin / TCF-4 target gene OPN is histone deacetylase dependent. Vietor I, Kurzbauer R, Brosch G, Huber LA. Department of Biocenter, Medical University Innsbruck, Innsbruck, Tirol A-6020. TPA Induced Sequence 7 (TIS7) acts as a transcriptional co-repressor interacting with SIN3, the histone deacetylase-containing complex. The overexpression of TIS7 down-regulates expression of a specific set of genes. Homozygous deletion of this gene in mice delays injury-induced muscle regeneration and inhibits muscle satellite cell differentiation and fusion of myoblasts in vitro. Osteopontin (OPN), a known ss-catenin / Tcf-4 downstream target gene is up-regulated in tumors and in cells with increased motility such as muscle cells. OPN promoter sequence contains binding sites for Sp1, glucocorticoid receptor, E-box-binding factors, octamer motif-binding protein, c-Myc and other transcription factors. Previously we have shown that TIS7 regulates the OPN expression through the inhibition of the Sp1-activating effects. Here we show that TIS7 has the capacity to inhibit OPN expression also through Lef-1, the second identified OPN regulatory element. TIS7 has the capacity to down-regulate ss-catenin / Tcf-4 transcriptional activity. TIS7 homologous deletion in mouse embryonic fibroblasts (MEFs) increased not only the TOPflash reporter gene transcriptional activity but also the expression of c-Myc and OPN. Furthermore, we show that TIS7 overexpression leads to the ss-catenin interaction with enzymatically active histone deacetylases. We propose that TIS7 down-regulates the ss-catenin / Tcf-4 transcriptional activity via its interaction with histone deacetylase-containing complex thereby inhibiting the expression of ss-catenin downstream target genes such as c-Myc and OPN. We hypothesize that TIS7 as a negative regulator of transcriptional activity represses expression of OPN and ss-catenin / Tcf-4 target genes, which are involved in myogenesis, muscle maintenance and regeneration in a histone deacetylase dependent manner. PMID: 16204248 [PubMed - as supplied by publisher] --------------------------------------------------------------- 4: Exp Mol Pathol. 2005 Oct;79(2):108-17. Retinoic acid-induced downmodulation of telomerase activity in human cancer cells. Xiao X, Sidorov IA, Gee J, Lempicki RA, Dimitrov DS. Protein Interactions Group, Laboratory of Experimental and Computational Biology, NCI-Frederick, NIH, Bldg. 469, Rm. 139, P.O. Box B, Miller Drive, Frederick, MD 21702-1201, USA. xiaox@ncifcrf.gov Most human cancers express telomerase but its activity is highly variable and regulated by complex mechanisms. Recently, several studies have suggested that retinoic acid (RA) downregulates telomerase activity and that this effect could be a major determinant of its therapeutic activity. To elucidate possible mechanisms of RA-mediated downmodulation of telomerase activity, we measured the kinetics of concentration changes of several transcription regulators by using standard biochemical techniques at low (10 muM) and high (100 muM) RA concentrations. We further evaluated the global impact of the RA treatment on gene expression profiles using microarray. It was found that the kinetics of c-Myc correlates most closely with the telomerase activity suggesting in agreement with previous studies that this protein is a major intermediate of the RA-induced downregulation of telomerase activity. Other telomerase regulators as Sp1 and Mad1 did not exhibit significant correlation. The dominant role of c-Myc in RA-induced telomerase downmodulation is confirmed by microarray data. Additionally, a number of proteins were found as possible correlates of telomerase activity by microarray analysis. These data suggest a complex interplay between c-Myc and other proteins that may be important determinants of the RA effects on telomerase activity in human cancer cells. The complex mechanism through which telomerase activity is controlled during differentiation and cancer transformation is also reflected. PMID: 16054129 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 5: Cell Cycle. 2005 Jul;4(7):881-2. Epub 2005 Jul 11. Genetic instability: the dark side of the hypoxic response. To KK, Koshiji M, Hammer S, Huang LE. Laboratory of Human Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-4255, USA. Under low oxygen tension, the activated transcription factor HIF-1alpha upregulates an array of hypoxia-inducible genes via heterodimerization with ARNT and binding to the hypoxia-responsive element in the promoter. Alternatively, HIF-1alpha regulates hypoxia-responsive genes by functionally antagonizing the oncoprotein Myc via protein-protein interactions. This so-called HIF-1alpha-Myc mechanism apparently not only accounts for the gene upregulation, but also for the gene downregulation during hypoxia, depending upon the activating and repressive nature of Myc in gene expression. Indeed, our recent study demonstrated that both mismatch repair genes, MSH2 and MSH6, are inhibited by this mechanism in a p53-dependent manner. In particular, the constitutively bound transcription factor Sp1 serves as a molecular switch by recruiting HIF-1alpha in hypoxia to displace the transcription activator Myc from the promoter. Therefore, our findings shed light on the mechanisms underlying hypoxia-induced genetic instability, an "adverse"effect of the hypoxic response, and yet a germane process to tumor survival and progression. PMID: 15970707 [PubMed - in process] --------------------------------------------------------------- 6: Carcinogenesis. 2005 Nov;26(11):1879-1889. Epub 2005 Jun 15. Differential transcriptional regulation of human telomerase in a cellular model representing important genetic alterations in esophageal squamous carcinogenesis. Quante M, Heeg S, von Werder A, Goessel G, Fulda C, Doebele M, Nakagawa H, Beijersbergen R, Blum HE, Opitz OG. Department of Medicine and Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany, Gastroenterology Division, University of Pennsylvania, Philadelphia, PA, USA and Netherlands Cancer Institute, Amsterdam, The Netherlands. Telomerase activity is observed in approximately 90% of human cancer including esophageal squamous cell cancer. Normal somatic cells do not display telomerase activity on a regular basis. The major mechanism to regulate telomerase activity in human cells is the transcriptional control of the catalytic subunit, the human reverse transcriptase gene hTERT. However, the manner in which telomerase activity is regulated during malignant transformation and whether this regulation is influenced by single genetic alterations important in this process are not well understood. In this study we investigated the transcriptional regulation and activity of human telomerase in a cellular model representing important known genetic alterations observed in esophageal cancer. We characterized the respective cells with regard to their telomere biology and telomerase expression, transcriptional regulation using promoter- as well as electrophoretic mobility shift assay-analyses and their promoter methylation status. We could demonstrate that telomerase expression and subsequent activity are differentially regulated in the progression from normal esophageal epithelial cells to genetically defined esophageal cells harboring a specific genetic alteration frequently found in esophageal cancer and compared those changes with esophageal cancer cells. Whereas primary esophageal cells are mainly regulated by Sp1, in cells harboring a genetic alteration as cyclin D1 overexpression other transcription factors like E2F and c-myc as well as promoter methylation influence hTERT transcription. This model demonstrates that the transcriptional regulation of telomerase is influenced by a given genetic alteration important in esophageal cancer, and therefore provides new insight in telomerase regulation during carcinogenesis. PMID: 15958520 [PubMed - as supplied by publisher] --------------------------------------------------------------- 7: J Biol Chem. 2005 Aug 12;280(32):28867-76. Epub 2005 Jun 10. Sp1/Sp3-dependent regulation of human telomerase reverse transcriptase promoter activity by the bioactive sphingolipid ceramide. Wooten LG, Ogretmen B. Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA. In this study, the roles of Sp1/Sp3 transcription factors in the regulation of the activity of human telomerase reverse transcriptase (hTERT) promoter in response to ceramide were examined in the A549 human lung adenocarcinoma cells. The activity of the N-terminal truncated hTERT promoter, lacking the c-Myc recognition (E-box) region but containing multiple Sp1/Sp3 sites, was also significantly inhibited by C6-ceramide, indicating a role for ceramide in the regulation of Sp1/Sp3 function. Partial inhibition of Sp1 expression using small interfering RNA resulted in a significant inhibition of the hTERT promoter. Treatment with C6-ceramide inhibited the trans-activation function of overexpressed Sp1, whereas it induced the repressor effects of exogenous Sp3 on the hTERT promoter. The interaction between Sp1 and hTERT promoter DNA was significantly reduced in response to ceramide as assessed by chromatin immunoprecipitation analysis. In contrast, the promoter DNA-binding activity of Sp3 was slightly increased in response to C6-ceramide, resulting in the increased ratio of Sp3/Sp1 on the hTERT promoter, which was concomitant with the reduced recruitment of RNA polymerase II to the promoter. Furthermore, mutations of various Sp1/Sp3 recognition sequences significantly attenuated the activity of the promoter in the presence or absence of ceramide, demonstrating the importance of multiple Sp1/Sp3 recognition sites for the promoter activity. Mechanistically, the data demonstrated that C6-ceramide reduced the acetylation of Sp3 protein and partially blocked the activation of the hTERT promoter by the histone deacetylase inhibitor trichostatin A. The roles of endogenous long chain ceramide generated in response to gemcitabine in the inhibition of hTERT promoter activity and the regulation of Sp3 acetylation were also demonstrated. PMID: 15951564 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 8: Gene. 2005 May 23;351:171-80. Regulation of the cyclin D1 and cyclin A1 promoters by B-Myb is mediated by Sp1 binding sites. Bartusel T, Schubert S, Klempnauer KH. Institut fur Biochemie, Westfalische-Wilhelms-Universitat Munster, Germany. B-Myb is a highly conserved member of the Myb family of transcription factors which plays an important role during the cell cycle. Previous work has shown that B-Myb is phosphorylated at several sites by cyclin A/Cdk2 in the early S-phase. These phosphorylations increase the transactivation potential of B-Myb by counteracting the repressive function of an inhibitory domain located at the carboxyl-terminus of B-Myb. As yet, only a few genes have been identified as B-Myb target genes. Previous work has suggested that the cyclin D1 gene might be regulated by B-Myb. Here, we have studied the effect of B-Myb on the promoter of the cyclin D1 gene. We show that B-Myb is a potent activator of the cyclin D1 promoter and that this activation is not mediated by Myb binding sites but rather by a group of Sp1 binding sites which have previously been shown to be crucial for cyclin D1 promoter activity. Our data show that the C-terminal domain of B-Myb is required for the activation of the cyclin D1 promoter and that this part of B-Myb interacts with Sp1. Finally, we have found that the promoter of the cyclin A1 gene is also activated by B-Myb by a Sp1 binding site-dependent mechanism. The effect of B-Myb on the promoters of the cyclin A1 and D1 genes is reminiscent of the mechanism that has been proposed for the autoregulation of the B-myb promoter by B-Myb, which also involves Sp1 binding sites. Taken together, our identification of two novel B-Myb responsive promoters whose activation by B-Myb does not involve Myb binding sites extends previous evidence for the existence of a distinct mechanism of transactivation by B-Myb which is dependent on Sp1 binding sites. The observation that this mechanism is not subject to the inhibitory effect of the C-terminal domain of B-Myb but rather requires this domain supports the notion that the Sp1 site-dependent mechanism is already active in the G1-phase prior to the phosphorylation of B-Myb by cyclin A/Cdk2. PMID: 15922873 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 9: Exp Cell Res. 2005 May 1;305(2):300-11. Survivin enhances telomerase activity via up-regulation of specificity protein 1- and c-Myc-mediated human telomerase reverse transcriptase gene transcription. Endoh T, Tsuji N, Asanuma K, Yagihashi A, Watanabe N. Department of Clinical Laboratory Medicine, Sapporo Medical University, School of Medicine, South-1, West-16, Sapporo 060-8543, Japan. Suppression of apoptosis is thought to contribute to carcinogenesis. Survivin, a member of the inhibitor-of-apoptosis family, blocks apoptotic signaling activated by various cellular stresses. Since elevated expression of survivin observed in human cancers of varied origin was associated with poor patient survival, survivin has attracted growing attention as a potential target for cancer treatment. Immortalization of cells also is required for carcinogenesis; telomere length maintenance by telomerase is required for cancer cells to proliferate indefinitely. Yet how cancer cells activate telomerase remains unclear. We therefore examined possible interrelationships between survivin expression and telomerase activity. Correlation between survivin and human telomerase reverse transcriptase (hTERT) expression was observed in colon cancer tissues, and overexpression of survivin enhanced telomerase activity by up-regulation of hTERT expression in LS180 human colon cancer cells. DNA-binding activities of specificity protein 1 (Sp1) and c-Myc to the hTERT core promoter were increased in survivin gene transfectant cells. Phosphorylation of Sp1 and c-Myc at serine and threonine residues was enhanced by survivin, while total amounts of these proteins were unchanged. Further, "knockdown" of survivin by a small inhibitory RNA decreased Sp1 and c-Myc phosphorylation. Thus survivin participates not only in inhibition of apoptosis, but also in prolonging cellular lifespan. PMID: 15817155 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 10: J Neurosci. 2005 Mar 30;25(13):3350-7. Retinoic acid-induced chromatin remodeling of mouse kappa opioid receptor gene. Park SW, Huq MD, Loh HH, Wei LN. Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA. The mouse kappa opioid receptor (KOR) gene is constitutively expressed in P19 embryonic stem cells but is first suppressed and reactivated during retinoic acid (RA)-induced neuronal differentiation. However, no RA response element (RARE) can be found in this gene regulatory region. The suppression and reactivation of the KOR gene in this neuronal differentiation model suggested chromatin remodeling occurred on this gene promoter triggered by RA induction. This study asks whether RA induces alteration in the nucleosomal structure of this gene promoter that has no apparent RARE and, if so, how RA remodels chromatin of this promoter. The results revealed two loose nucleosomes, N1 at -44 (3' boundary) from the transcription initiation site and N2 spanning the transcription initiation site, that are relevant to active transcription. RA formed a repressive chromatin configuration of this promoter by compacting nucleosome N1, followed by nucleosome N2 condensation. Chromatin immunoprecipitation assay demonstrated RA induced replacement of the c-Myc/Max complex with the Max/Mad1 complex on the E box located within nucleosome N1, coinciding with reduced Sp1 binding to GC boxes located within nucleosome N2 and recruitment of chromatin remodeling factor Brahma-related gene 1 (BRG-1) to this promoter. Consistently, histone deacetylation, Lys9 methylation, and hypophosphorylation of RNA polymerase II C-terminal domain were detected on this promoter after RA treatment. It is concluded that RA induces KOR gene suppression, as early neuronal differentiation marker, by inducing substitution of c-Myc/Max with Max/Mad on the E box and by BRG-1 involved nucleosome recruitment and chromatin condensation, thereby abolishing Sp1 binding. PMID: 15800190 [PubMed - in process] --------------------------------------------------------------- 11: Br J Dermatol. 2005 Mar;152(3):435-43. Retinoic acid suppresses telomerase activity in HSC-1 human cutaneous squamous cell carcinoma. Kunisada M, Budiyanto A, Bito T, Nishigori C, Ueda M. Division of Dermatology, Clinical Molecular Medicine, Faculty of Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan. kunisada@med.kobe-u.ac.jp BACKGROUND: Activation of telomerase is crucial for the continued growth and progression of cancer cells. In a previous study, we showed that telomerase is frequently activated in skin tumours. OBJECTIVE: Because retinoic acid (RA) plays an important role in the growth and differentiation of keratinocytes and as RA has some preventive and therapeutic effects on human skin cancers, we examined the effect of RA on the telomerase activity of HSC-1 human cutaneous squamous cell carcinoma cells. RESULTS: Treatment of HSC-1 cells with all-trans RA (ATRA) significantly suppressed their telomerase activity. The suppression of telomerase activity was obvious at day 4 and was maximal at day 5 after the start of treatment with RA. This suppression was reversible as removal of ATRA allowed the recovery of telomerase activity. The suppression of telomerase activity correlated with the decreased expression of mRNA of human telomerase catalytic subunit (hTERT), the rate-limiting determinant of enzyme activity. The production of c-myc and of Sp1 proteins, transcription factors regulating hTERT expression, was not suppressed in HSC-1 cells by ATRA, but phosphorylation of extracellular signal-regulated kinases (ERK)1/2 and of the serine/threonine kinase Akt was significantly suppressed. Phosphorylation of the epidermal growth factor receptor, which regulates hTERT expression in HSC-1 cells, was not altered by ATRA. CONCLUSIONS: These data indicate that RA is effective in inhibiting telomerase activity in HSC-1 cells. Suppression of ERK1/2 and Akt activation is presumed to be involved in the RA-induced suppression of hTERT. PMID: 15787811 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 12: Mol Cell. 2005 Mar 18;17(6):793-803. HIF-1alpha induces genetic instability by transcriptionally downregulating MutSalpha expression. Koshiji M, To KK, Hammer S, Kumamoto K, Harris AL, Modrich P, Huang LE. Laboratory of Human Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA. Hypoxia promotes genetic instability by undefined mechanisms. The transcription factor HIF-1alpha is crucial for the cellular response to hypoxia and is frequently overexpressed in human cancers, resulting in the activation of genes essential for cell survival. Here, we demonstrate that HIF-1alpha is responsible for genetic instability at the nucleotide level by inhibiting MSH2 and MSH6, thereby decreasing levels of the MSH2-MSH6 complex, MutSalpha, which recognizes base mismatches. HIF-1alpha displaces the transcriptional activator Myc from Sp1 binding to repress MutSalpha expression in a p53-dependent manner; Sp1 serves as a molecular switch by recruiting HIF-1alpha to the gene promoter under hypoxia. Furthermore, in human sporadic colon cancers, HIF-1alpha overexpression is statistically associated with the loss of MSH2 expression, especially when p53 is immunochemically undetectable. These findings indicate that the regulation of DNA repair is an integral part of the hypoxic response, providing molecular insights into the mechanisms underlying hypoxia-induced genetic instability. PMID: 15780936 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 13: Blood. 2005 Jul 1;106(1):304-10. Epub 2005 Mar 10. Arsenic suppresses gene expression in promyelocytic leukemia cells partly through Sp1 oxidation. Chou WC, Chen HY, Yu SL, Cheng L, Yang PC, Dang CV. Department of Laboratory Medicine, National Taiwan University Hospital, Taipei.wchou@ha.mc.ntu.edu.tw The mechanism by which arsenic dramatically affects gene expression remains poorly understood. Here we report that prolonged exposure of acute promyelocytic leukemia NB4 cells to low levels of arsenic trioxide increased the expression of a set of genes responsible for reactive oxygen species (ROS) production. We hypothesize that arsenic-induced ROS in turn contribute partially to altered gene expression. To identify genes responsive to arsenic-induced ROS, we used microarray gene expression analysis and identified genes that responded to arsenic and hydrogen peroxide but whose response to arsenic was reversed by an ROS scavenger, N-acetyl-L-cysteine. We found that 26% of the genes significantly responsive to arsenic might have been directly altered by ROS. We further explored the mechanisms by which ROS affects gene regulation and found that the Sp1 transcription factor was oxidized by arsenic treatment, with a corresponding decrease in its in situ binding on the promoters of 3 genes, hTERT, C17, and c-Myc, whose expressions were significantly suppressed. We conclude that ROS contributed partly to arsenic-mediated gene regulation and that Sp1 oxidation contributed to gene suppression by arsenic-induced ROS. PMID: 15761015 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 14: Hua Xi Kou Qiang Yi Xue Za Zhi. 2004 Dec;22(6):499-502. [Expression of telomerase activity and c-myc and stimulatory protein 1 in human ameloblastoma] [Article in Chinese] Zhong M, Li ZJ, Wang J, Zhang B, Hou L, Gong YB. Dept. of Pathology, College of Stomatology, China Medical University, Shenyang 110002, China. OBJECTIVE: To study the oncogene transcriptor c-myc, stimulatory protein 1 (SP1) expression in ameloblastoma (AB) and their relation with telomerase reverse transcripase (hTERT), and to investigate the clinical biological characteristics of AB. METHODS: The expression was observed in AB by in situ hybridization and SP method. RESULTS: The positive rates of c-myc mRNA, hTERT mRNA and SP1 protein were 81.5% (44/54), 94.4% (51/54) and 83.3% (45/54), respectively. Their positive rates increased as AB recurred and transformed malignantly. A strong correlation was found between hTERT and c-myc, hTERT and SP1 (rs = 0.853, P < 0.001; rs = 0.900, P < 0.001). CONCLUSION: Activity of telomerase plays an important role in the tumorigenesis development of AB. Increasing of hTERT expression may be related to c-myc and SP1. The expression of these three parameters has a significant correlation with the clinical biological characteristics of AB. PMID: 15656532 [PubMed - in process] --------------------------------------------------------------- 15: J Biol Chem. 2005 Jan 14;280(2):1112-22. Epub 2004 Nov 4. Myc antagonizes Ras-mediated growth arrest in leukemia cells through the inhibition of the Ras-ERK-p21Cip1 pathway. Vaque JP, Navascues J, Shiio Y, Laiho M, Ajenjo N, Mauleon I, Matallanas D, Crespo P, Leon J. Grupo de Biologia Molecular del Cancer, Departamento de Biologia Molecular, Unidad de Biomedicina del Consejo Superior de Investigaciones Cientiificas, Facultad de Medicina, Universidad de Cantabria, 39011 Santander, Spain. Even though RAS usually acts as a dominant transforming oncogene, in primary fibroblasts and some established cell lines Ras inhibits proliferation. This can explain the virtual absence of RAS mutations in some types of tumors, such as chronic myeloid leukemia (CML). We report that in the CML cell line K562 Ras induces p21Cip1 expression through the Raf-MEK-ERK pathway. Because K562 cells are deficient for p15INK4b, p16INK4a, p14ARF, and p53, this would be the main mechanism whereby Ras up-regulates p21 expression in these cells. Accordingly, we also found that Ras suppresses K562 growth by signaling through the Raf-ERK pathway. Because c-Myc and Ras cooperate in cell transformation and c-Myc is up-regulated in CML, we investigated the effect of c-Myc on Ras activity in K562 cells. c-Myc antagonized the induction of p21Cip1 mediated by oncogenic H-, K-, and N-Ras and by constitutively activated Raf and ERK2. Activation of the p21Cip1 promoter by Ras was dependent on Sp1/3 binding sites in K562. However, mutational analysis of the p21 promoter and the use of a Gal4-Sp1 chimeric protein strongly suggest that c-Myc affects Sp1 transcriptional activity but not the binding of Sp1 to the p21 promoter. c-Myc-mediated impairment of Ras activity on p21 expression required a transactivation domain, a DNA binding region, and a Max binding region. Moreover, the effect was independent of Miz1 binding to c-Myc. Consistent with its effect on p21Cip1 expression, c-Myc rescued cell growth inhibition induced by Ras. The data suggest that in particular tumor types, such as those associated with CML, c-Myc contributes to tumorigenesis by inhibiting Ras antiproliferative activity. PMID: 15528212 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 16: Mech Dev. 2004 Dec;121(12):1509-22. A role for nucleoprotein Zap3 in the reduction of telomerase activity during embryonic stem cell differentiation. Armstrong L, Lako M, van Herpe I, Evans J, Saretzki G, Hole N. School of Biological and Biomedical Sciences, University of Durham, South Road, Durham DH1 3LE, UK. Telomerase, the enzyme which maintains the ends of linear chromosomes in eukaryotic cells is found in murine embryonic stem cells; however, its activity is downregulated during in vitro differentiation. Previous work has indicated that this is due to the transcriptional downregulation of murine reverse transcriptase unit (mTert) of telomerase. To investigate the factors that cause the transcriptional repression of mTert we defined a 300 bp region which is essential for its transcription and performed site directed mutagenesis and electrophoretic mobility shift assays. This analysis indicated that Sp1, Sp3 and c-Myc bind to the GC-boxes and E-boxes, respectively, within the promoter and help activate the transcription of mTert gene. We also identified a novel binding sequence, found repeated within the mTert core region, which when mutated caused increased mTert expression. Yeast one hybrid screening combined with electrophoretic mobility shift assays indicated that the nuclear protein Zap3 binds to this site and its overexpression leads to the downregulation of mTert during differentiation. This suggests that regulation of mTert transcription is a complex process which depends on a quantitative balance between transcription factors that cause activation or repression of this gene. Overexpression of Zap3 in murine embryonic stem cells results in reduction in telomerase activity and telomere length as well as reduced proliferative capacity and limited ability to contribute to the development of haematopoietic cells upon differentiation. PMID: 15511642 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 17: Biochem Biophys Res Commun. 2004 Nov 12;324(2):860-7. Histone deacetylase inhibitor, Trichostatin A, activates p21WAF1/CIP1 expression through downregulation of c-myc and release of the repression of c-myc from the promoter in human cervical cancer cells. Li H, Wu X. Institute of Medical Virology, Wuhan University School of Medicine, Wuhan, Hubei 430071, PR China. huilee6@yahoo.com Histone deacetylase (HDAC) inhibitors have shown promise in clinical cancer therapy and to consistently induce p21WAF1/CIP1 expression in a p53-independent manner and via increased acetylation of the chromatin at the Sp1 sites in the p21WAF1/CIP1 promoter region. However, the exact mechanism by which HDAC inhibitors induce p21WAF1/CIP1 remains unclear. In this study, we observed that Trichostatin A (TSA), a HDAC inhibitor, induced strikingly p21WAF1/CIP1 expression in human cervical cancer (HeLa) cells, and this induction correlated with downregulation of c-myc expression. Coincident with this observation, knock down of c-myc with a c-myc specific small interfering RNA dramatically induced expression of p21WAF1/CIP1 in these cancer cells. These data suggest that c-myc may play a critical role in repression of p21WAF1/CIP1 expression in HeLa cells. More importantly, using chromatin immunoprecipitation assay, we observed for the first time that c-myc bound to the endogenous p21WAF1/CIP1 promoter in untreated HeLa cells, but not in TSA-treated cells. Taken together, TSA induced c-myc downregulation and release from the endogenous p21WAF1/CIP1 promoter contributes, at least partially, to transcriptional activation of the p21WAF1/CIP1 in HeLa cells. PMID: 15474507 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 18: J Cell Sci. 2004 Aug 1;117(Pt 17):3855-65. Epub 2004 Jul 20. pRb, Myc and p53 are critically involved in SV40 large T antigen repression of PDGF beta-receptor transcription. Uramoto H, Hackzell A, Wetterskog D, Ballagi A, Izumi H, Funa K. Department of Cell Biology, Institute of Anatomy and Cell Biology, Goteborg University, Box 420, SE-405 30 Gothenburg, Sweden. The expression of the PDGF beta-receptor is tightly regulated during a normal cell cycle. c-Myc and p73alpha repress transcription of the receptor through interaction with NF-Y. In ST15A cells which stably express the temperature-sensitive SV40 large T antigen (LT) the receptor expression and ligand binding decreased under the permissive condition. Transient expression of the LT, but not small t, decreased the endogenous receptor expression at mRNA and protein levels in NIH3T3 cells but not in the myc-null HO15.19 cells. The wild-type LT, but not the various pRb or p53 binding defective LT mutants, represses the PDGF beta-receptor promoter activity. Moreover, the inability of the LT-mediated repression in the myc-null cells, the Rb-null 3T3 cells, and the Saos-2 cells lacking pRb and p53, indicates that Myc, pRb and p53 are all necessary elements. PDGF beta-receptor promoter-luciferase assays revealed that the CCAAT motif is important for the repression. Furthermore, p53 was found to increase the promoter activity mainly via the upstream Sp1 binding sites together with the CCAAT motif in the NIH 3T3 cells. This was confirmed by Schneider's Drosophila line (SL2) cells deficient in both endogenous NF-Y and Sp1. Chromatin immunoprecipitation using ST15A cells revealed that both LT and p53 bound the PDGF beta-receptor promoter and the binding of p53 diminished when LT was expressed in the permissive condition. However, LT binds the promoter in the absence of pRb and p53 in Saos-2 cells stably expressing LT. These results suggest that LT binds the promoter and interferes with NF-Y and Sp1 to repress it in the presence of Myc, pRb and p53. PMID: 15265983 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 19: J Biol Chem. 2004 Sep 17;279(38):39574-83. Epub 2004 Jul 19. Transcriptional regulation of the human beta-1,4-galactosyltransferase V gene in cancer cells: essential role of transcription factor Sp1. Sato T, Furukawa K. Department of Biosignal Research, Tokyo Metropolitan Institute of Gerontology, Itabashi-ku, Tokyo 173-0015, Japan. taksato@tmig.or.jp Beta-1,4-galactosyltransferase (beta-1,4-GalT) V is a constitutively expressed enzyme that can effectively galactosylate the GlcNAcbeta1-->6Man group of the highly branched N-glycans that are characteristic of tumor cells. Upon malignant transformation of cells, the expression of the beta-1,4-GalT V gene increases in accordance with the increase in the amounts of highly branched N-glycans. Lectin blot analysis showed that the galactosylation of highly branched N-glycans is inhibited significantly in SH-SY5Y human neuroblastoma cells by the transfection of the antisense beta-1,4-GalT V cDNA, indicating the biological importance of the beta-1,4-GalT V for the functions of highly branched N-glycans. We cloned the 2.3-kb 5'-flanking region of the human beta-1,4-GalT V gene, and we identified the region -116/-18 relative to the transcription start site as that having promoter activity. The region was found to contain several putative binding sites for transcription factors, including AP2, AP4, N-Myc, Sp1, and upstream stimulatory factor. Electrophoretic mobility shift assay showed that Sp1 binds to nucleotide positions -81/-69 of the promoter region. Mutations induced in the Sp1-binding site showed that the promoter activity of the beta-1,4-GalT V gene is impaired completely in cancer cells. In contrast, the promoter activity increased significantly by the transfection of the Sp1 cDNA into A549 human lung carcinoma cells. Mithramycin A, which inhibits the binding of Sp1 to its binding site, reduced the promoter activation and expression of the beta-1,4-GalT V gene in A549 cells. These results indicate that Sp1 plays an essential role in the transcriptional activity of the beta-1,4-GalT V gene in cancer cells. PMID: 15263012 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 20: Blood. 2004 Oct 15;104(8):2523-31. Epub 2004 Jun 29. Transcriptional activation of hTERT through the NF-kappaB pathway in HTLV-I-transformed cells. Sinha-Datta U, Horikawa I, Michishita E, Datta A, Sigler-Nicot JC, Brown M, Kazanji M, Barrett JC, Nicot C. University of Kansas Medical Center, Department of Microbiology, Immunology, and Molecular Genetics, 3025 Wahl Hall West, 3901 Rainbow Blvd, Kansas City, KS 66160, USA. In immortal cells, the existence of a mechanism for the maintenance of telomere length is critical. In most cases this is achieved by the reactivation of telomerase, a cellular reverse transcriptase that prevents telomere shortening. Here we report that the telomerase gene (hTERT) promoter is up-regulated during transmission of human T-cell lymphotropic virus type-I (HTLV-I) to primary T cells in vitro and in ex vivo adult T-cell leukemia/lymphoma (ATLL) samples, but not asymptomatic carriers. Although Tax impaired induction of human telomerase reverse transcriptase (hTERT) mRNA in response to mitogenic stimulation, transduction of Tax into primary lymphocytes was sufficient to activate and maintain telomerase expression and telomere length when cultured in the absence of any exogenous stimulation. Transient transfection assays revealed that Tax stimulates the hTERT promoter through the nuclear factor kappaB (NF-kappaB) pathway. Consistently, Tax mutants inactive for NF-kappaB activation could not activate the hTERT or sustain telomere length in transduced primary lymphocytes. Analysis of the hTERT promoter occupancy in vivo using chromatin immunoprecipitation assays suggested that an increased binding of c-Myc and Sp1 is involved in the NF-kappaB-mediated activation of the hTERT promoter. This study establishes the role of Tax in regulation of telomerase expression, which may cooperate with other functions of Tax to promote HTLV-I-associated adult T-cell leukemia. PMID: 15226182 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 21: Hum Mol Genet. 2004 Aug 1;13(15):1611-21. Epub 2004 Jun 2. Occupancy and synergistic activation of the FMR1 promoter by Nrf-1 and Sp1 in vivo. Smith KT, Coffee B, Reines D. Department of Biochemistry and Graduate Program in Genetics and Molecular Biology, Emory University School of Medicine, Atlanta, GA 30322, USA. Fragile X syndrome is due to mutation of the FMR1 gene. The most common mutation is an expansion of a CGG repeat in the 5' UTR that triggers dense DNA methylation and formation of a heterochromatin-like structure which lead to transcriptional silencing. In vitro experiments have identified several transcription factors, including Sp1, Nrf-1 and USF1/2, as potential regulators of normal FMR1 promoter activity. Using CpG methylation-deficient Drosophila cells, we demonstrate in vivo that Nrf-1 and Sp1 are strong, synergistic activators of an unmethylated human FMR1-driven reporter, while USF1/2 and Max repress this activation. In addition, analyses of transcription factor activity upon DNA methylation of the reporter show that Sp1 activity was largely intact when the promoter was densely methylated, but Nrf-1 transactivation was very sensitive to dense methylation. Notably, Nrf-1 transactivation was relatively insensitive to methylation of cytosines only at its binding site. FMR1 reporter activity is also reduced in HeLa cells after expression of a short interfering RNA directed against endogenous Nrf-1. Using chromatin immunoprecipitation, we demonstrate directly that Sp1 and Nrf-1 occupy the human FMR1 promoter in vivo and these interactions are disrupted in fragile X patient cells. In addition, we discover that Max resides at the FMR1 promoter and show that USF1/2 but not c-Myc are present at endogenous FMR1. These findings provide the first direct in vivo evidence identifying the specific transcription factors that regulate FMR1. PMID: 15175277 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 22: Biochem Biophys Res Commun. 2004 May 14;317(4):1096-102. Activation of mouse RAG-2 promoter by Myc-associated zinc finger protein. Wu CX, Zhao WP, Kishi H, Dokan J, Jin ZX, Wei XC, Yokoyama KK, Muraguchi A. Department of Immunology, Faculty of Medicine, Toyama Medical and Pharmaceutical University, 2630, Sugitani, Toyama 930-0194, Japan. Recombination activating gene-1 (RAG-1) and RAG-2 are expressed specifically in lymphocytes undergoing the antigen receptor gene rearrangement during the lymphocyte development. Our previous study showed that the -41 to -17 nucleotides (nt) 5' -upstream region of mouse RAG-2 were pre-requisite for the core promoter activity and that Pax-5/c-Myb/LEF-1 protein-protein complex was responsible for its activity in immature B cells. In this study, we show that the -65/-42 sequence, the non-conserved sequence between human and mouse RAG-2 promoter, is necessary for the full promoter activity for mouse RAG-2. Electrophoresis mobility shift assay revealed that Myc-associated zinc finger protein (MAZ) as well as SP1/3 binds a GA box in this region. Using chromatin immunoprecipitation, we show that MAZ binds the RAG-2 promoter region in pre-B cells. Furthermore, we show that MAZ synergistically activates the murine RAG-2 promoter with Pax-5/c-Myb/LEF-1 complex. These results first demonstrate that MAZ participates in activation of mouse RAG-2 promoter. PMID: 15094381 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 23: Gene. 2004 Mar 3;327(2):233-8. The human RAP250 gene: genomic structure and promoter analysis. Antonson P, Al-Beidh F, Matthews J, Gustafsson JA. Department of Biosciences at Novum, Karolinska Institutet, Novum, S-14157 Huddinge, Sweden. per.antonson@cbt.ki.se Ligand-induced gene activation by nuclear receptors involves the recruitment of coactivators to hormone bound receptors. Recent results have shown that RAP250, also termed as ASC-2/PRIP/TRBP/NRC/AIB3, plays a critical role as a coactivator of nuclear receptors. In this study, we have determined the genomic organization of the human RAP250 gene in order to identify the promoter region. By searching the GenBank database for EST sequences, we could identify two previously unknown exons in the 5'-end of the gene. Our results show that the RAP250 gene consists of 15 exons spanning 111 kb. All sequences of the splice donor and acceptor sites fit the GT-AG role for splicing. We also show using linkage analysis that the mouse Rap250 gene is located on chromosome 2 close to the agouti gene. The RAP250 promoter is GC-rich and TATA-less, and contains multiple Sp1 binding sites and a MYC binding site. A reporter plasmid containing the 5' flanking region of the RAP250 gene showed significant activity compared to pGL3-basic and minimal thymidine kinase (TK) reporter plasmids in transfection experiments using luciferase reporter genes. Our data show that the RAP250 has a complex genomic structure with a promoter that is regulated by multiple transcription factors for its basal expression. PMID: 14980720 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 24: Cytogenet Genome Res. 2003;102(1-4):309-17. The chicken telomerase RNA gene: conservation of sequence, regulatory elements and synteny among viral, avian and mammalian genomes. Delany ME, Daniels LM. Department of Animal Science, University of California, Davis, CA 95616, USA. medelany@ucdavis.edu Telomerase RNA (TR) is essential for telomerase activity and the maintenance of telomere length in proliferating cell populations. The objective of the present research was to define the cytogenetic and molecular genomic organization of chicken TR (chTR). The chTR exists as a single copy gene (TERC, alias TR), mapping to chromosome 9 (GGA9). The loci on the q arm of GGA9 map to three chromosomes in human with five of the nine GGA9q loci mapping to HSA3q. Sequencing of the chTERC locus (3,763 bp) from the UCD 001 genome (Red Jungle Fowl) included: 604 bp 5', 465 coding, and 2,694 bp 3' (from -604 to +3159). Sequence analysis included homology searches conducted on several levels including comparisons among different chicken genotypes, Marek's disease virus (MDV) sequences, plus human and murine. We provide evidence for distal 5' and 3' sequence homology between chTERC and the MDV genome among other known regions of homology (promoter and coding), elaborate on 5' transcription factor binding motifs among the various genomes as well as show type and number of TERT-related motifs 3' of chicken TR (e.g., Sp1, c-Myb, c-Myc, AP2, among others). Surrounding the gene are more than 25 Sp1 sites, over 20 oncogene transcription factor binding motifs and numerous hormonal and other specialized binding motifs. Knowledge of 5' and 3' chTERC regulatory elements will be useful for investigating normal control mechanisms during growth and development as well as investigating the potential for dysregulation of this important gene during oncogenesis, especially among different genotypes. Copyright 2003 S. Karger AG, Basel PMID: 14970722 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 25: J Mol Endocrinol. 2004 Feb;32(1):99-113. Functional importance of Myc-associated zinc finger protein for the human parathyroid hormone (PTH)/PTH-related peptide receptor-1 P2 promoter constitutive activity. Leroy C, Manen D, Rizzoli R, Lombes M, Silve C. Inserm U 426 et Institut Federatif de Recherche 02, Faculte de Medecine Xavier Bichat, 16 rue Henri Huchard, 75018 Paris, France. The aim of the present study was to analyze the functional importance for the parathyroid hormone (PTH)/PTH-related peptide (PTHrP) receptor (PTHR1) gene P2 promoter activity of the putative proximal Myc-associated zinc finger protein (MAZ) site localized at position bp -45 to -39 bp, taking advantage of a G/A mutation identified at position -40 in the human sequence. Wild-type 'full-length' (1285P2) and truncated (760P2) promoter sequences were inserted upstream to the luciferase basic (pLucB) and enhancer (pLucE) reporter gene expression vectors. Transient transfections in osteoblast-like SaOS-2 cells and renal cells (RC.SV3A2) showed that the -40 G/A mutation significantly impaired transcriptional activity of wild-type 1285P2-pLucB and 760P2-pLucE promoter constructs. Further truncation of the P2 sequence demonstrated that the sequence -109/-37 bp was essential for promoter activity. Co-transfection with a MAZ expression vector did not modify the wild-type 1285P2-pLucB construct reporter activity but significantly increased 2-fold the mutated construction activity (P<0.05). Electrophoretic mobility shift assays using SaOS-2 nuclear extracts and a double-stranded DNA fragment encompassing the -45 to -39 putative MAZ site (ds-MAZ-oligo) disclosed two specific DNA-protein complexes. Complex II (fast moving) had a lower affinity for the mutated MAZ motif than for the wild-type MAZ motif while complex I (slow moving) had the same affinity for both wild-type or mutated MAZ sequences. Competition studies with Sp1 consensus oligonucleotide (ds-Sp1-oligo) markedly reduced complex I intensity, with a concomitant increase in that of complex II. Finally, ribonuclease protection assays showed that P2-specific PTHR1 mRNA transcript expression was significantly decreased in SaOS-2 cells transfected with ds-MAZ-oligo as compared with that for control (P<0.001) and ds-Sp1-oligo (P<0.05). Taken together, our studies suggest that the putative -45 to -39 MAZ-binding site regulates the constitutive activity of human PTHR1 P2 promoter. PMID: 14765995 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 26: Mol Cell Biol. 2004 Feb;24(4):1680-90. Ogt-dependent X-chromosome-linked protein glycosylation is a requisite modification in somatic cell function and embryo viability. O'Donnell N, Zachara NE, Hart GW, Marth JD. Department of Cellular and Molecular Medicine, Howard Hughes Medical Institute, University of California San Diego, La Jolla, California 92093-0625, USA. The Ogt gene encodes a glycosyltransferase that links N-acetylglucosamine to serine and threonine residues (O-GlcNAc) on nuclear and cytosolic proteins. Efforts to study a mammalian model of Ogt deficiency have been hindered by the requirement for this X-linked gene in embryonic stem cell viability, necessitating the use of conditional mutagenesis in vivo. We have extended these observations by segregating Ogt mutation to distinct somatic cell types, including neurons, thymocytes, and fibroblasts, the latter by an approach developed for inducible Ogt mutagenesis. We show that Ogt mutation results in the loss of O-GlcNAc and causes T-cell apoptosis, neuronal tau hyperphosphorylation, and fibroblast growth arrest with altered expression of c-Fos, c-Jun, c-Myc, Sp1, and p27. We further segregated the mutant Ogt allele to parental gametes by oocyte- and spermatid-specific Cre-loxP mutagenesis. By this we established an in vivo genetic approach that supports the ontogeny of female heterozygotes bearing mutant X-linked genes required during embryogenesis. Successful production and characterization of such female heterozygotes further indicates that mammalian cells commonly require a functional Ogt allele. We find that O-GlcNAc modulates protein phosphorylation and expression among essential and conserved cell signaling pathways. PMID: 14749383 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 27: J Invest Dermatol. 2003 Nov;121(5):1088-94. Inhibition of the epidermal growth factor receptor suppresses telomerase activity in HSC-1 human cutaneous squamous cell carcinoma cells. Budiyanto A, Bito T, Kunisada M, Ashida M, Ichihashi M, Ueda M. Division of Dermatology, Clinical Molecular Medicine, Faculty of Medicine, Kobe University Graduate School of Medicine, Japan. Activation of telomerase, which stabilizes the telomere length of chromosomes, is crucial for the continued growth or progression of cancer cells. In a previous study, we showed that telomerase is frequently activated in skin tumors. Because epidermal growth factor plays an important role during the tumorigenesis of epithelial tissue, we have now examined the role of epidermal growth factor signaling in regulating telomerase activity using HSC-1 human cutaneous squamous cell carcinoma cells. Treatment of HSC-1 cells with AG 1478, an inhibitor of the epidermal growth factor receptor, or with a neutralizing antibody to the epidermal growth factor receptor, significantly suppressed their telomerase activity, in association with inhibiting their growth. The suppression of telomerase activity was obvious at day 3 and was maximal at day 5 after treatment with AG 1478. The suppression of telomerase activity correlated with the decreased expression of human telomerase catalytic subunit (hTERT) mRNA, the rate-limiting determinant of its enzyme activity. The expression of c-Myc and of Sp1 proteins, transcription factors for hTERT, were also suppressed by AG 1478 in HSC-1 cells, but the expression of Ets-2 protein, another transcription factor, was not affected. The expression of Mad-1, a competitor of c-Myc, was increased. Inhibition of ERK, Src, or Akt suppressed telomerase activity in HSC-1 cells, but to a lesser extent than did treatment with AG 1478. Serum starvation suppressed telomerase activity, but addition of epidermal growth factor or transforming growth factor alpha did not increase it, indicating the involvement of other epidermal growth factor receptor ligands in the activation of telomerase in HSC-1 cells. These data indicate that blockade of the epidermal growth factor receptor might be effective in inhibiting telomerase activity of squamous cell carcinomas, which would lead to the suppression of tumor growth. PMID: 14708611 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 28: Thromb Haemost. 2003 Dec;90(6):1029-39. Characterization of the human protein S gene promoter: a possible role of transcription factors Sp1 and HNF3 in liver. Tatewaki H, Tsuda H, Kanaji T, Yokoyama K, Hamasaki N. Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-city 812-8582, Japan. Protein S is a vitamin-K-dependent plasma glycoprotein that serves as a cofactor for activated protein C in the protein C anticoagulation pathway, which regulates blood coagulation by inactivating factors Va and VIIIa. Mechanisms regulating the expression of the protein S gene remain unknown to date. The aim of this study was to characterize the cis-acting DNA elements of the human protein S gene in liver. We found that liver cell lines (HepG2 and PLC) transcribed the human protein S gene to mRNA, whereas non-liver cell lines (HEK293 and HeLa cells) either transcribed the gene weakly or not at all. Isolation and analysis of tissue-specific gene expression in HepG2 and HeLa cells of the 5'-flanking region from -6183 to +294 of the protein S gene indicated that the consensus binding motifs to HNF3 and Sp1 or MAZ transcription factors in the flanking region are essential for protein S gene expression. Exogenous expression of the Sp1 gene augmented the protein S-reporter gene expression in HepG2 or PLC cells but had no effect in HeLa cells.Taken together, we would conclude that transcription factors of HNF3, MAZ, and Sp1 are required for high-level expression of the protein S gene in hepatic cells, but in non-hepatic cells such as HeLa cells, an unknown factor(s) binds to the Sp1 region and disturbs the action of Sp1 and MAZ. PMID: 14652633 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 29: Int J Oncol. 2003 Dec;23(6):1703-8. Down-regulation of telomerase activity in malignant glioma cells by p27KIP1. Kanzawa T, Komata T, Kyo S, Germano IM, Kondo Y, Kondo S. Department of Neurosurgery, Mount Sinai School of Medicine, New York, NY 10029, USA. The cyclin-dependent kinase inhibitor p27KIP1 is considered not only a prognostic factor in cancer, but also a promising anti-cancer agent. However, the relationship between p27KIP1 and telomerase, that has potential as tumor-marker, remains to be elucidated. In this study, using the recombinant adenoviral vector expressing p27KIP1 (Adp27KIP1), we investigated whether p27KIP1 affects telomerase activity in malignant glioma U373-MG cells. Overexpression of p27KIP1 suppressed telomerase activity in tumor cells. The down-regulation of telomerase was due to inhibition of the human telomerase reverse transcriptase (hTERT) gene expression at the transcriptional level. This inhibitory effect was partially induced by interfering with binding sites of the hTERT core promoter for transcription factors Myc and Sp1. These findings identify a novel role for p27KIP1 in down-regulation of telomerase activity. PMID: 14612944 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 30: Exp Mol Pathol. 2003 Dec;75(3):238-47. Role of Ets/Id proteins for telomerase regulation in human cancer cells. Xiao X, Athanasiou M, Sidorov IA, Horikawa I, Cremona G, Blair D, Barret JC, Dimitrov DS. Laboratory of Experimental and Computational Biology, NCI-Frederick, NIH, Miller Drive, Frederick, MD 21702-1201, USA. xiaox@ncifcrf.gov Most human cancers express telomerase but its activity is highly variable and regulated by complex mechanisms. Recently, we have proposed that Ets proteins may be important for regulation of telomerase activity in leukemic cells. Here we provide further evidence for the role of Ets family members and related Id proteins in telomerase regulation and characterize the underlying molecular mechanisms. By using PCR-based and gel shift assays we demonstrated specific binding to a core hTERT promoter of Ets2, Fli1, Id2, c-Myc, Mad1, and Sp1 in lysates from subclones of U937 cells. Further analysis of binding of purified proteins and various mutants of the hTERT promoter suggested the existence of a trimolecular Ets-Id2-DNA complex, and Ets inhibitory activity mediated by c-Myc and the Ets binding site on the core hTERT promoter at -293 bp from the transcription initiation site as well as a positive Ets regulatory effect mediate through another Ets binding site at -36 bp. This analysis provided evidence for the existence of negative and positive Ets regulatory site and suggested a complex interplay between Ets/Id family members and c-Myc that may be an important determinant of the diversity of telomerase activity in leukemia and other cancers. PMID: 14611815 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 31: Hum Gene Ther. 2003 Oct 10;14(15):1415-28. Ad-mTERT-delta19, a conditional replication-competent adenovirus driven by the human telomerase promoter, selectively replicates in and elicits cytopathic effect in a cancer cell-specific manner. Kim E, Kim JH, Shin HY, Lee H, Yang JM, Kim J, Sohn JH, Kim H, Yun CO. BK21 Project for Medical Sciences, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, 120-752, Korea. Human telomerase reverse transcriptase (hTERT), the catalytic subunit of telomerase, functions to stabilize telomere length during chromosomal replication. Previous studies have shown that hTERT promoter is highly active in most tumor and immortal cell lines but inactive in normal somatic cell types. The use of wild-type hTERT promoter, however, may be limited by its inability to direct high level and cancer cell-specific expression necessary for effective targeted gene therapy. To improve cancer cell specificity and the strength of the hTERT promoter, a modified hTERT, m-hTERT promoter was generated in which additional copies of c-Myc and Sp1 binding sites were incorporated adjacent to the promoter. As assessed using relative lacZ expression, hTERT and m-hTERT promoter activity was significantly upregulated in cancer cells but not in normal cells, and within these upregulated cancer cells, m-hTERT promoter strength was substantially higher than that of the wild-type hTERT. Next, to restrict viral replication to tumor cells, a conditional replication-competent adenoviruses, Ad-TERT-Delta19 and Ad-mTERT-delta19 were generated in which the E1A gene, which is essential for viral replication, was placed under the control of the hTERT and m-hTERT promoter, respectively. While the wild-type Ad-TERT-delta19 replicated in and induced cytopathic effect in cancer and in some normal cell lines, Ad-mTERT-delta19 enhanced viral replication and cytopathic effect only in cancer cells. Furthermore, the growth of established human cervical carcinoma in nude mice was significantly suppressed by intratumoral injection of Ad-mTERT-delta19. Taken together, present results strongly suggest that the use of the m-hTERT promoter is not only useful in the regulation of therapeutic gene expression but also that replication-competent oncolytic adenovirus under the control of the m-hTERT promoter may be a new promising tool for the treatment of human malignancies. PMID: 14577922 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 32: Mol Pharmacol. 2003 Nov;64(5):1180-8. Regulation of the rat phenylethanolamine N-methyltransferase gene by transcription factors Sp1 and MAZ. Her S, Claycomb R, Tai TC, Wong DL. Department of Psychiatry, Harvard Medical School Laboratory of Molecular and Developmental Neurobiology, McLean Hospital, 115 Mill Street, Rm 116, Belmont, MA 02478, USA. The rat phenylethanolamine N-methyltransferase (PNMT) gene promoter contains 1-base pair (bp) overlapping consensus sequences for Sp1 and MAZ transcription factors at -48 and -38 bp, respectively. Gel mobility assays using PC-12-derived RS1 cell nuclear extracts or in vitro translated proteins showed that Sp1 and MAZ specifically bind to these elements, that MAZ displaces/prevents Sp1 binding, and that Sp1 and MAZ binding is mutually exclusive, with occupancy dependent on each factor's concentration and affinity for its consensus element. In transfection assays, PNMT promoter activation by Sp1 and MAZ depends on promoter length, with -893 bp of sequence yielding greatest activation. Although MAZ has higher affinity for its binding element, it is a less effective activator. Changes in PNMT promoter activity for the constructs pGL3RP60 or pGL3RP893 using a fixed amount of MAZ expression construct and a variable amount of Sp1 expression construct or vice versa confirmed the latter. Mutation of the MAZ or Sp1 sites in pGL3RP60 attenuated but did not eliminate PNMT promoter activity, even though the proteins no longer bind to their consensus elements. Phosphatase treatment of RS1 cell nuclear extracts prevented MAZ- and Sp1-DNA binding complex formation. Although MAZ and Sp1 elevate endogenous PNMT mRNA in RS1 cells, MAZ preferentially increases intron-retaining whereas Sp1 preferentially increases intronless mRNA. Thus, expression of the PNMT gene seems to be modulated through competitive binding of phosphorylated Sp1 and MAZ to their consensus elements in the promoter. In addition, post-transcriptional regulation seems to be another important mechanism controlling PNMT expression. PMID: 14573768 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 33: Mol Genet Metab. 2003 Sep-Oct;80(1-2):272-80. N-myc oncogene expression in neuroblastoma is driven by Sp1 and Sp3. Tuthill MC, Wada RK, Arimoto JM, Sugino CN, Kanemaru KK, Takeuchi KK, Sidell N. Molecular Carcinogenesis Section, Cancer Etiology Program, Cancer Research Center of Hawaii, The University of Hawaii at Manoa, 1236 Lauhala Street, Honolulu, HI 96813-2424, USA. Regulation of N-myc oncogene expression is an important determinant of the biological behavior of neuroblastoma. The N-myc promoter contains several potential binding sites for transcription factors of the Sp1 family. Mutation of a CT-box motif contained within a 26 bp region required for N-myc downregulation by retinoic acid decreased basal transcriptional activity and altered DNA-protein interactions of the promoter, while mutations flanking this motif did neither. On super-shift, this region was shown to recruit Sp1 and Sp3 transcription factor proteins, while a functionally significant CT-box mutation resulted in their replacement by NF-1 transcription factor. Lysates from Drosophila S2 cells expressing exogenous Sp1, Sp3, and NF-1 proteins were able to partially mimic gel shift complexes seen with neuroblastoma nuclear extract and either wild type or mutant probes. Transient transfections of S2 cells showed that both individually and together, Sp1 and Sp3 were able to trans-activate a wild type CT-box-driven luciferase reporter construct in a dose-dependent manner. Transfection of the wild type but not mutant CT-box oligonucleotide was able to decrease endogenous N-myc expression in neuroblastoma cells. Together these results suggest that the CT-box element serves a critically functional role, and in the basal state, allows for N-myc trans-activation by Sp1 and Sp3. Moreover when mutated, the CT-box may still function as a binding motif for alternate transcription factors such as NF-1 that can allow persistent N-myc expression. PMID: 14567977 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 34: Curr Med Chem Anti-Canc Agents. 2003 Nov;3(6):411-20. Chartreusin, elsamicin A and related anti-cancer antibiotics. Portugal J. Departamento de Biologia Molecular y Celular, Instituto de Biologia Molecular de Barcelona, CSIC, Barcelona, Spain. jpmbmc@cid.csic.es Chartreusin and elsamicin A are structurally related antibiotics that bind to GC-rich tracts in DNA, with a clear preference for B-DNA over Z-DNA. They inhibit RNA synthesis and cause single-strand scission of DNA via the formation of free radicals. Elsamicin A can also be regarded as the most potent inhibitor of topoisomerase II reported so far. It can inhibit the formation of several DNA-protein complexes. Elsamicin A binding to the P1 and P2 promoter regions of the c-myc oncogene inhibits the binding of the Sp1 transcription factor, thus inhibiting transcription. Despite the pharmacological interest in chartreusin, elsamicin A and their derivatives, there is no experimental data on the structure of their complexes with DNA. This shortcoming has been partially solved by a theoretical approach, which provided some details about the DNA-elsamicin A interaction, and the thermodynamic characterization of the binding of chartreusin and elsamicin A to DNA. Elsamicin A but not chartreusin is being developed clinically as an anti-cancer agent. IST-622 (6-O-(3-ethoxypropylonyl)-3',4'-O-exo-benzylidene-chartreusin), a novel semi-synthetic derivative of chartreusin, which has shown a promising anti-cancer activity in a phase II study, appears to be a pro-drug with a more suitable pharmacokinetic profile than chartreusin. Publication Types: Review Review, Tutorial PMID: 14529449 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 35: Mol Cancer Res. 2003 Aug;1(10):739-46. Telomerase reverse transcriptase promoter regulation during myogenic differentiation of human RD rhabdomyosarcoma cells. Ma H, Urquidi V, Wong J, Kleeman J, Goodison S. UCSD Cancer Center, University of California, San Diego, La Jolla, CA 92093-0064, USA. During terminal differentiation of human and murine cells, telomerase activity and parallel transcription of telomerase reverse transcriptase (hTERT) are inhibited. In this study, we used in vitro and in vivo analyses to determine the role of hTERT promoter elements and associated factors during differentiation-induced inhibition of telomerase expression in RD, a human rhabdomyosarcoma cell line. Assay of telomerase enzyme activity, hTERT mRNA, and reporter gene assays confirmed that the hTERT promoter was silenced during 12-O-tetradecanoylphorbol-13-acetate-induced myogenic differentiation of telomerase-positive RD cells. Promoter deletion and mutation analyses revealed that two E-boxes and an AP-2 site present in a 320-bp region of the promoter were essential for the transcriptional activity of the hTERT gene. Electrophoretic mobility shift assays identified several factors that interact with this region of DNA, including the muscle-specific transcription factors Myf5, Myf6, and myogenin and the ubiquitously expressed factors Sp1 and AP-2. Ectopic expression of the E-box binding factors c-Myc and Mad did influence promoter activity in these cells; indeed, the presence of endogenous c-Myc protein was altered after differentiation. Our findings suggest that the acute regulation of hTERT transcription is primarily controlled by E-box elements, which bind a series of factors during the phased phenotypic changes occurring during the differentiation of RD human muscle cells. PMID: 12939399 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 36: Prog Nucleic Acid Res Mol Biol. 2003;73:107-36. Dynamic interplay between O-glycosylation and O-phosphorylation of nucleocytoplasmic proteins: a new paradigm for metabolic control of signal transduction and transcription. Kamemura K, Hart GW. Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. The glycosylation of serine and threonine residues with beta-O-linked N-acetylglucosamine (O-GlcNAc) is an abundant posttranslational modification of nuclear and cytoplasmic proteins in multicellular eukaryotes. This highly dynamic glycosylation/deglycosylation of protein is catalyzed by the nucleocytoplasmic enzymes, UDP-G1cNAc: polypeptide O-beta-N-acetylglucosaminyltransferase (OGT)/O-beta-N-acetylglucosaminidase. OGT is required for embryonic stem cell viability and mouse ontogeny, thus O-GlcNAc is essential for the life of eukaryotes. The gene encoding O-GlcNAcase maps to a locus important to late-onset Alzheimer's disease. All known O-GlcNAc-modified proteins are also phosphoproteins that form reversible multimeric protein complexes. There is both a global and often site-specific reciprocal relationship between O-GlcNAc and O-phosphate in many cellular responses to stimuli. Thus, regulation of the protein-protein interaction(s) and/or protein function by dynamic glycosylation/phosphorylation has been hypothesized. In this chapter, we will review the current status of dynamic glycosylation/phosphorylation of several important regulatory proteins including c-Myc, estrogen receptors, Sp1, endothelial nitric oxide synthase, and beta-catenin. Various aspects of subcellular localization, association with binding partners, activity, and/or turnover of these proteins appear to be regulated by dynamic glycosylation/ phosphorylation in response to cellular signals or stages. Publication Types: Review PMID: 12882516 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 37: J Biol Chem. 2003 Sep 19;278(38):36017-26. Epub 2003 Jul 2. Characterization of the murine Nramp1 promoter: requirements for transactivation by Miz-1. Bowen H, Lapham A, Phillips E, Yeung I, Alter-Koltunoff M, Levi BZ, Perry VH, Mann DA, Barton CH. Division of Biochemistry and Molecular Biology, University of Southampton, Bassett Crescent East, Southampton SO16 7PX, United Kingdom. Murine Nramp1 encodes a divalent cation transporter that is expressed in late endosomes/lysosomes of macrophages, and the transported cations facilitate intracellular pathogen growth control. The Nramp1 promoter is TATA box-deficient, has two initiator elements, and is repressed by c-Myc, in accordance with the notion that genes that deplete the iron content of the cell cytosol antagonize cell growth. Repression via c-Myc occurs at the initiator elements, whereas a c-Myc-interacting protein (Miz-1) stimulates transcription. Here we demonstrate that a non-canonical E box (CAACTG) inhibits basal promoter activity and activation by Miz-1. A consensus Sp1-binding site or GC box is also necessary for Miz-1-dependent transactivation, but not repression. Repression occurs by c-Myc competing with p300/CBP for binding Miz-1. Our results show that an Sp1 site mutant inhibits coactivation by p300 and that the murine Nramp1 promoter is preferentially expressed within macrophages (relative to a beta-actin control) compared with non-macrophage cells. The effect of the Sp1 site mutation on promoter function shows cell-type specificity: stimulation in COS-1 and inhibition in RAW264.7 cells. Miz-1-directed RNA interference confirms a stimulatory role for Miz-1 in Nramp1 promoter function. c-Myc, Miz-1, and Sp1 were identified as binding to the Nramp1 core promoter in control cells and following acute stimulation with interferon-gamma and lipopolysaccharide. These results provide a description of sites that modulate the activity of the initiator-binding protein Miz-1 and indicate a stimulatory role for GC box-binding factors in macrophages and a inhibitory role for E box elements in proliferating cells. PMID: 12840021 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 38: Genomics. 2003 Jun;81(6):640-3. Organization of the human FK506-binding immunophilin FKBP52 protein gene (FKBP4). Scammell JG, Hubler TR, Denny WB, Valentine DL. Department of Pharmacology, University of South Alabama College of Medicine, Mobile, AL 36688, USA. jscammel@jaguar1.usouthal.edu FKBP52 is a widely expressed FK506-binding immunophilin that possesses peptidylprolyl isomerase activity and a tetratricopeptide repeat involved in protein-protein interaction. FKBP52 plays an important role in steroid receptor function and is implicated in other diverse processes, including regulation of transcription, cation channel activity, and gene transfer efficiency. Reported here is the genomic organization of the human FKBP52 gene (FKBP4), which shares all but one of the same exon-intron boundaries as the structurally related immunophilin FKBP51 gene (FKBP5). Approximately 3.5 kb of 5'-flanking DNA of FKBP4 was subcloned into a luciferase reporter vector and was found to exhibit robust activity in T-47D, MCF7, and COS-7 cells. Promoter constructs with only 143 bp of upstream sequence maintained high activity. This region contains a CAAT motif sequence and consensus binding sites for Sp1, heat-shock factor, and MYC-MAX, which are conserved in the rabbit FKBP4 promoter and, when deleted, dramatically reduced promoter activity in T-47D cells. PMID: 12782134 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 39: J Mol Biol. 2003 May 16;328(5):977-83. Repression of the TMEFF2 promoter by c-Myc. Gery S, Koeffler HP. Division of Hematology/Oncology, Cedars-Sinai Medical Center, UCLA School of Medicine, Los Angeles, CA 90048, USA. gerys@schs.org TMEFF2 is a novel transmembrane protein, containing two follistatin domains and an epidermal growth factor-like motif that is mainly expressed in the prostate and brain. Recently, we showed that expression of TMEFF2 could inhibit prostate cancer cell growth. In addition, the TMEFF2 gene is frequently hypermethylated in human tumor cells, suggesting that it might be a tumor suppressor gene. We cloned the 5'-flanking region of the human TMEFF2 gene and using a luciferase reporter assay showed that it contains a functional promoter. The 0.7 kb region upstream to the TMEFF2 transcription start site encompasses the minimal promoter required for TMEFF2 expression. Sequence analysis of the TMEFF2 promoter revealed potential binding sites for several transcription factors including Sp1 and an E-box that could be recognized by c-Myc. An inverse correlation between TMEFF2 and c-Myc expression was found in CWR22 prostate xenografts. Reporter gene and mobility shift assays demonstrated that c-Myc could repress TMEFF2 gene expression through its cognate site. In light of the probable role of TMEFF2 in inhibiting cell growth, its suppression may contribute to the oncogenic properties of c-Myc. PMID: 12729735 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 40: Int J Mol Med. 2003 May;11(5):547-53. Transcriptional regulation by zinc-finger proteins Sp1 and MAZ involves interactions with the same cis-elements. Song J, Ugai H, Nakata-Tsutsui H, Kishikawa S, Suzuki E, Murata T, Yokoyama KK. Gene Engineering Division, Department of Biological Systems, BioResource Center, Tsukuba Institute, RIKEN (The Institute of Physical and Chemical Research), Ibaraki 305-0074, Japan. Various transcription factors, such as Sp1 and MAZ, include C2H2-type zinc-finger motifs and are able to bind to GC-rich cis-elements that are distributed in the promoter regions of numerous mammalian genes. The consensus sequence of Sp1-binding sites is very similar to that of MAZ-binding sites. In fact, Sp1 and MAZ bind to the same cis-elements in the promoters of the genes for the receptor for serotonin 1A (HT1Ar), endothelial nitric-oxide synthase (eNOS), phenylethanolamine N-methyltransferase (PNMT), the receptor for parathyroid hormone (PTHr), MAZ and the major late promoter of adenovirus (AdMLP). It appears that two consecutive zinc-finger motifs of Sp1 and MAZ might be essential for the interaction of each protein with DNA. Sp1 and MAZ activated the expression of the genes for HT1Ar and PTHr, as well as AdMLP. Both Sp1 and MAZ inhibited the expression of the gene for MAZ, while expression of the gene for eNOS was enhanced by Sp1 and repressed by MAZ. These observations suggest that both Sp1 and MAZ might have dual functions in the regulation of gene expression. Our results suggest, furthermore, that histone deacetylases are involved in autorepression of the gene for MAZ, while expression of DNA methyltransferase I is associated with suppression of the expression of the gene for MAZ by Sp1. Thus, both deacetylation and methylation might be involved in the regulation of expression of individual genes, with different zinc-finger proteins binding to the same cis-elements but recruiting different proteins, such as methylases and acetylases, to the transcriptional complex. Publication Types: Review Review, Tutorial PMID: 12684688 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 41: Arterioscler Thromb Vasc Biol. 2003 May 1;23(5):748-54. Epub 2003 Apr 3. Fibroblast growth factor-2, but not vascular endothelial growth factor, upregulates telomerase activity in human endothelial cells. Kurz DJ, Hong Y, Trivier E, Huang HL, Decary S, Zang GH, Luscher TF, Erusalimsky JD. Cell Biology Group, British Heart Foundation Laboratories, Department of Medicine, University College London, London, UK. OBJECTIVE: Telomerase plays a major role in the control of replicative capacity, a critical property for successful angiogenesis and maintenance of endothelial integrity. In this study, we examined the relationship between telomerase activity and endothelial cell proliferation as well as the regulation of this enzyme by fibroblast growth factor-2 (FGF-2) and vascular endothelial growth factor-A (VEGF). METHODS AND RESULTS: Telomerase was repressed in endothelial cells freshly derived from intact endothelium, whereas activity was present during logarithmic growth in culture. In cultured human umbilical vein endothelial cells (HUVECs), mRNA levels of hTERT-the catalytic subunit of telomerase-and enzyme activity decreased reversibly on induction of quiescence. Treatment of quiescent HUVECs with FGF-2 restored telomerase activity in a time- and dose-dependent manner, whereas VEGF had no such effect, although both factors induced comparable mitogenic responses. FGF-2, but not VEGF, upregulated the mRNA levels for hTERT and for the hTERT gene transactivation factor Sp1. Serial passage in the presence of individual growth factors accelerated the accumulation of senescent cells in VEGF-treated cultures compared with cultures treated with FGF-2. CONCLUSIONS: FGF-2, but not VEGF, restores telomerase activity and maintains the replicative capacity of endothelial cells. PMID: 12676798 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 42: Clin Exp Metastasis. 2003;20(1):77-84. Transcriptional regulation of osteopontin and the metastatic phenotype: evidence for a Ras-activated enhancer in the human OPN promoter. Denhardt DT, Mistretta D, Chambers AF, Krishna S, Porter JF, Raghuram S, Rittling SR. Nelson Laboratories, Rutgers University, Piscataway, New Jersey 88854, USA. denhardt@biology.rutgers.edu Elevated osteopontin (OPN) transcription often correlates with increased metastatic potential of transformed cells, and in several model systems OPN--whether produced by the tumor cells or by stromal cells - has been shown to enhance metastatic ability. Sequence elements in the OPN promoter have been identified on the basis of their ability to interact with protein factors associated with the tumorigenic process in one or more cell lineages. One of these is a Ras-activated enhancer (RAE) that binds a protein, the Ras-response factor (RRF), whose ability to form a complex with the RAE is stimulated by Ras signaling in fibroblasts and epithelial cells. Another is the T cell factor-4 binding site, which in the OPN promoter can retard OPN transcription when bound by the Tcf-4 protein. In Rama 37 rat mammary epithelial cells Tcf-4 suppresses OPN transcription and the metastatic phenotype. A third promoter segment consists of two sequences in the -94 to -24 region of the human OPN promoter able to bind several known transcription factors, including Sp1, Myc and Oct-1, which may act synergistically to stimulate OPN transcription in malignant astrocytic cells. Although expression of other genes may also be regulated by these transcription factors, evidence suggests that often OPN alone can stimulate metastasis. In this communication we address two issues: (1) How does OPN facilitate the metastatic phenotype? (2) What mechanisms are responsible for the increase in OPN transcription in metastatic cells? Publication Types: Review Review, Tutorial PMID: 12650610 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 43: Genomics. 2003 Feb;81(2):221-33. Molecular cloning of the mouse AMY-1 gene and identification of the synergistic activation of the AMY-1 promoter by GATA-1 and Sp1. Furusawa M, Taira T, Iguchi-Ariga SM, Ariga H. CREST, Japan Science and Technology Corporation, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan. We have reported that a novel c-Myc binding protein, AMY-1, stimulated the transcription activity of c-Myc and was translocated from the cytoplasm to the nucleus in a c-Myc-dependent manner. AMY-1 works as an inducer of human K562 cell differentiation upon induction of AraC. To characterize the expression or functional importance of AMY-1, the genomic DNA of mouse AMY-1 was cloned and characterized. Both mouse and human genomic DNAs, the latter of which was retrieved from a human DNA database, comprise five exons spanning about 11 kb. To characterize the promoter of the mouse AMY-1 gene, a series of deletion constructs of the region upstream of the first ATG was linked to the luciferase gene, and their luciferase activities were measured in human HeLa and K562 cells. The results showed that Sp1 was essential for AMY-1 expression in both cell lines and that GATA-1 is also necessary in K562 cells. Sp1 in both cell lines and GATA-1 only in K562 cells were identified as proteins binding to these sites by a mobility shift assay. Furthermore, it was found that GATA-1 stimulated AMY-1 expression synergistically with Sp1 in ectopically expressed insect cells and that both proteins were associated in K562 cells. PMID: 12620400 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 44: Exp Cell Res. 2003 Feb 1;283(1):17-21. Mechanisms of c-myc-mediated transcriptional repression of growth arrest genes. Gartel AL, Shchors K. Department of Molecular Genetics, M/C 669, 900 S Ashland Avenue, University of Illinois at Chicago, Chicago, IL 60607, USA. agartel@uic.edu Constitutive expression of the proto-oncogene c-myc results in oncogenic activation and contributes to progression of a wide range of human and animal tumors. Myc executes its multiple activities mostly through transcriptional regulation of the target genes. The special interest of this review is the mechanism of transcriptional repression of cell cycle inhibitors by Myc. Myc suppresses expression of cell cycle/growth arrest genes gas1, p15, p21, p27, and gadd34, -45, and -153. It appears that Myc represses growth arrest gene transcription by at least two distinct mechanisms. One mechanism is limited to the binding of Myc-Max heterodimers to the Inr element in their promoters and inhibition of Miz-1 or other transcriptional activators via the C-terminal domain of c-Myc. This mechanism requires DNA binding of the Myc-Max complex to Inr sequences. The other mechanism is dependent on c-Myc binding to the Sp1 transcription factor via the c-Myc central region and inhibition of Sp1 transcriptional activity. At this time it is not entirely clear which Sp1-containing promoters will be repressed by c-Myc and what other modes of c-Myc transcriptional repression may exist. The ability of c-Myc to repress transcription of growth arrest genes may contribute to its potential to promote proliferation and oncogenesis. Copyright 2003 Elsevier Science (USA) Publication Types: Review Review, Tutorial PMID: 12565816 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 45: Nucleic Acids Res. 2003 Jan 15;31(2):562-9. Basal transcriptional regulation of human damage-specific DNA-binding protein genes DDB1 and DDB2 by Sp1, E2F, N-myc and NF1 elements. Nichols AF, Itoh T, Zolezzi F, Hutsell S, Linn S. Department of Molecular and Cell Biology, Barker Hall, University of California, Berkeley, CA 94720-3202, USA. The human DDB1 and DDB2 genes encode the 127 and 48 kDa subunits, respectively, of the damage-specific DNA-binding protein (DDB). Mutations in the DDB2 gene have been correlated with the hereditary disease xeroderma pigmentosum group E. We have investigated the proximal promoters of the DDB genes, both of which are G/C-rich and do not contain a TATA box. Transient expression analysis in HeLa cells using a luciferase reporter system indicated the presence of core promoters located within 292 bp (DDB1) and 220 bp (DDB2) upstream of the putative transcription initiation sites. Both core promoters contain multiple active Sp1 sites, with those of DDB1 at -123 to -115 and of DDB2 at -29 to -22 being critical determinants of promoter activity. In addition, an N-myc site at -56 to -51 for DDB1 is an essential transcription element, and mutations in a DDB1 NF-1 site at -104 to -92, a DDB2 NF-1 site at -68 to -56 and a DDB2 E2F site at +36 to +43 also reduce promoter activity. Taken together, these results suggest a regulation of basal transcription typical of cell cycle-regulated genes, and therefore support conjectures that the DDB heterodimer and/or its subunits have functions other than direct involvement in DNA repair. PMID: 12527763 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 46: Apoptosis. 2003 Jan;8(1):11-8. Roles of the stress-induced gene IEX-1 in regulation of cell death and oncogenesis. Wu MX. Department of Molecular and Cell Biology, Boston University Goldman School of Dental Medicine, Boston, MA 02118, USA. mxwu@bu.edu In response to changes in the external environment cells must initiate a coordinated program of gene expression for them to adapt. IEX-1 (immediate early response gene X-1) is precisely regulated by multiple transcription factors among which p53, NF-kappaB/rel, Sp1 and c-Myc play central roles, to ensure rapid and transient expression of IEX-1 in cells under a variety of stress conditions. Overexpression of IEX-1 renders some cells sensitive to apoptosis and accelerates cell cycle progression, but reduces proliferation of other cells, whereas disruption of IEX-1 expression is associated with decreases in both apoptosis and cell cycle progression. In sharp contrast to in vitro studies, in vivo constitutive expression of IEX-1 prevents activated T cells but not B cells from apoptosis, as shown using IEX-1-transgenic mice that target IEX-1 expression specifically to lymphocytes driven by the Emu enhancer. The animals developed a lupus-like disease and subsequently a high incidence of T cell lymphomas when they aged, due to insufficient apoptosis of T cells. These varied effects of IEX-1 on cell death and cell cycle progression in a cell-context dependent fashion implicate that IEX-1 is involved in more than one signaling pathway, understanding of which will certainly improve our knowledge with respect to cancer biology, cell death and cell cycle regulation. Publication Types: Review Review, Tutorial PMID: 12510147 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 47: Biochem Biophys Res Commun. 2002 Dec 6;299(3):360-5. Transcriptional activation of the c-fos gene by a LIM protein, Hic-5. Kim-Kaneyama J, Shibanuma M, Nose K. Department of Microbiology, Showa University School of Pharmaceutical Sciences, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, Japan. Hic-5 is a member of LIM family proteins with a striking similarity to paxillin and localizes primarily in the focal adhesion. We recently reported that Hic-5 translocated to the nucleus under oxidative stress and was involved in transcriptional regulation. In the present study, we extended these findings to show that transcription of c-fos gene was up-regulated by overexpression of Hic-5. In clonal stable transformants established from human immortalized fibroblasts by transfection of an expression vector of Hic-5, the constitutive level of c-fos mRNA was well correlated with that of Hic-5. In reporter assays using the luciferase gene under control of the human c-fos 5(')-upstream region from -2.2kb to +1, expression of Hic-5, that was engineered to accumulate in the nucleus, stimulated the transcriptional activity of the c-fos enhancer. From experiments using various deletions and point mutations, it was revealed that multiple sequences including GC/Sp1, Ets, and ERE/AP-1 elements found around the -1.3kb region were responsible for the activation by Hic-5. Hic-5 itself did not bind to these elements in a sequence specific manner, but p300 appeared to be involved in the induction of c-fos. These results suggest that Hic-5 participates in the transcriptional regulation of c-fos as a scaffold in transcriptional complexes. PMID: 12445807 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 48: Oncogene. 2002 Nov 14;21(52):7991-8000. Tumor-specific activation of hTERT-derived promoters by tumor suppressive E1A-mutants involves recruitment of p300/CBP/HAT and suppression of HDAC-1 and defines a combined tumor targeting and suppression system. Kirch HC, Ruschen S, Brockmann D, Esche H, Horikawa I, Barrett JC, Opalka B, Hengge UR. Department of Internal Medicine (Cancer Research), University of Essen, Germany. h-c.kirch@uni-essen.de Adenovirus (Ad) E1A proteins are transcriptional regulators with antioncogenic but also transforming properties. We have previously shown that transformation-defective Ad5 E1A-derivatives are excellent tumor suppressors. For tumor-specific expression of the E1A-derivatives we intend to use tumor specific human telomerase reverse transcriptase (hTERT) core promoters. Here, we show that Spm2 and other E1A proteins with an intact amino terminus activated all hTERT constructs 10-20-fold in malignant tumor cells but not in primary fibroblasts, without affecting the activity of endogenous telomerase. The transcription rate in tumor cells was in the range of transcription from the SV40 promoter, which qualifies an E1A-hTERT system as a putative tumor targeting/expression system. The activation of the hTERT promoter by E1A was enhanced upon deletion of the Wilms' tumor 1 negative regulatory element and maintained high after deletion of the adjacent c-Myc-responsive E-box, demonstrating an important role of the remaining sequences that contain several Sp1-motifs. E1A-mediated hTERT activation was independent from the presence of the conserved region 3 (CR3) of E1A but dependent on E1A's binding to p300/CBP and recruitment of its histone acetyltransferase activity. Moreover, E1A-Spm2 and histone deacetylase-1 behaved as antagonists with respect to the regulation of transcription from the hTERT promoter. Overall, hTERT promoter/E1A-Spm2 systems may turn out to be excellent tools for transcriptionally targeted anticancer gene therapy. PMID: 12439749 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 49: Brain Res Mol Brain Res. 2002 Nov 15;107(2):89-96. Effect of the ubiquitous transcription factors, SP1 and MAZ, on NMDA receptor subunit type 1 (NR1) expression during neuronal differentiation. Okamoto S, Sherman K, Bai G, Lipton SA. Center for Neuroscience and Aging, The Burnham Institute, La Jolla, CA 92037, USA. The silencer factor NRSF/REST has been reported to restrict expression to neurons of a variety of genes, including that encoding NMDA receptor subunit type 1 (NR1), by suppressing transcription in nonneuronal cells. However, we recently reported that in addition to the absence of NRSF/REST-binding activity, another neuron-specific mechanism is necessary for high level expression of the NR1 gene in neurons. In this study, we explored the mechanism of induction of NR1 promoter activity during neuronal differentiation of the P19 cell line. We identified a 27 base pair GC-rich region in the promoter as an important element responsible for induction of the NR1 gene after neuronal differentiation. We found that the ubiquitous transcription factors SP1 and MAZ bind to this GC-rich region. Surprisingly, the binding activities of SP1 and MAZ are not remarkably changed after neuronal differentiation. Mutations in the SP1 and MAZ sites impair binding of SP1 and MAZ proteins and also decrease NR1 promoter activity. These findings suggest that SP1 and MAZ mediate enhancement of NR1 promoter activity during neuronal differentiation despite the fact that their binding activity does not change. PMID: 12425938 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 50: Gene. 2002 Jul 24;295(1):117-23. Molecular cloning and characterization of the human budding uninhibited by benomyl (BUB3) promoter. Baek WK, Park JW, Lim JH, Suh SI, Suh MH, Gabrielson E, Kwon TK. Department of Microbiology, School of Medicine, Keimyung University, 194 DongSan-Dong Jung-Gu, Taegu 700-712, South Korea. Recently, cDNA corresponding to the human homologue of the BUB3 (budding uninhibited by benomyl) mitotic checkpoint protein has been identified and cloned. Previous studies from our laboratory and others have found this gene to localize to 10q26, a region that is frequently altered in various human cancers. We describe here a series of studies designed to understand the genomic structure of BUB3, particularly as it relates to regulation of gene expression. The human BUB3 gene has seven exons and six introns, and spans a genomic region of over 16 kb. The four WD repeat sequences in this gene are localized to exons 2, 4, and 6, and there is a major transcriptional start site located 554 nucleotides upstream of the ATG translation initiator codon. The promoter region lacks a TATA box but contains potential binding sites for the transcriptional factors including SP1, E2F, c-Myc, C/EBP and NFkappaB. To analyse the regulatory mechanisms controlling hBUB3 gene expression, we characterized the 5'-flanking region from nucleotide -1.3 to +0.58 kb by cloning various potions of this region in front of a luciferase reporter sequence. These experiments indicate that this region 5' region contains distinctive positive and negative regulatory elements. PMID: 12242018 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 51: Surgery. 2002 Aug;132(2):232-8. Importance of Sp1 consensus motifs in the MYCN promoter. Inge TH, Casson LK, Priebe W, Trent JO, Georgeson KE, Miller DM, Bates PJ. Children's Hospital Research Foundation and Department of Pediatric Surgery, Cincinnati Children's Hospital Medical Center, Ohio 45229,USA. BACKGROUND: MYCN (N-myc) amplification in neuroblastoma is associated with poor clinical outcome. Factors that regulate MYCN expression have not been elucidated. MYCN is considered a TATA-less promoter, whereas significant promoter activity resides within 160 bp 5' of the major transcription start site. This region contains two GC-rich motifs and a CT box, regions for potential transcription factor interaction. METHODS: To characterize DNA-protein interactions in this region of the MYCN promoter, electrophoretic mobility shift assays, and promoter-reporter were used. RESULTS: A MYCN promoter fragment was incubated with HeLa nuclear extract, with or without competitors. Three major protein/DNA complexes were formed. Formation of 2 complexes could be inhibited by unlabeled Sp1 consensus duplex and by the Sp1 site-specific drug WP631. Purified Sp1 protein produced a complex similar to that formed with HeLa extract. To determine whether these DNA/protein interactions could be blocked in a sequence-specific fashion, a triplex forming oligonucleotide (TFO) was used. This TFO was designed to bind in the major groove of the promoter, covering the CT-box (putative Sp1 binding) motif. When triplex formation was followed by addition of nuclear extract, protein binding was indeed inhibited. Functional significance of this inhibition was tested with pE/Bnmyc-luc, a promoter-reporter plasmid containing the human MYCN promoter driving luciferase expression. Incubation with TFO, but not control oligodeoxynucleotides, completely inhibited luciferase activity. CONCLUSIONS: These data suggest that protein binding does occur in regions of the MYCN promoter containing GC and CT box elements and that this interaction is important for MYCN promoter activity. By inference, these data also suggest that the proteins that bind in this region are Sp1 family members. PMID: 12219017 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 52: Cell Growth Differ. 2002 Aug;13(8):343-54. All-trans-retinoic acid induces nuclear factor kappaB activation and matrix metalloproteinase-9 expression and enhances basement membrane invasivity of differentiation-resistant human SK-N-BE 9N neuroblastoma Cells. Farina AR, Masciulli MP, Tacconelli A, Cappabianca L, De Santis G, Gulino A, Mackay AR. Section of Molecular Pathology, Department of Experimental Medicine, University of L'Aquila, 67100 L'Aquila, Italy. A comparison between retinoic acid (RA) differentiation-resistant and differentiation-sensitive SK-N-BE neuroblastoma (NB) cell lines revealed an association between resistance to differentiation, exhibited by N-myc stable transfected SK-N-BE 9N cells, with sensitivity to RA induction of p50/p65 nuclear factor kappaB (NF-kappaB) transcription factor activity and induction of matrix metalloproteinase (MMP)-9 expression leading to enhanced invasive behavior in vitro. These effects were not observed in differentiation-sensitive parental SK-N-BE or control-transfected SK-N-BE 2N counterparts. RA activated a MMP-9 promoter reporter gene construct in SK-N-BE 9N but not parental SK-N-BE or SK-N-BE 2N cells through a NF-kappaB element (-600) in association with enhanced p50 mRNA expression, reduced cytoplasmic inhibitor of nuclear factor kappaBalpha protein levels, and the induction of nuclear p50/p65 containing MMP-9 NF-kappaB site binding activity. RA activation of the MMP-9 promoter was inhibited by transient overexpression of a dominant-negative inhibitor of nuclear factor kappaBalpha protein and stimulated by transient p50 but not p65 overexpression in the absence of RA. A limited, nonessential function for activator protein 1 (-74), Ets (-540), and SP1 (-560) elements within the MMP-9 promoter was revealed by point mutation but was not associated with changes in the binding or position of complexes constitutive to differentiation-sensitive or -resistant cells. Our data indicates that in this model of NB resistance to differentiation that results from uncoupled RA regulation of N-myc expression, RA stimulates malignant NB cell behavior by inducing nuclear NF-kappaB transcription factor activity, which in turn induces MMP-9 expression and stimulation of basement membrane invasive capacity involving MMP-9 activity. PMID: 12193473 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 53: Mol Biol Rep. 2001;28(4):223-33. Sp1 is a key regulator of the PDGF beta-receptor transcription. Molander C, Hackzell A, Ohta M, Izumi H, Funa K. Institute of Anatomy and Cell Biology, Goteborg University, Gothenburg, Sweden. The mouse PDGF beta-receptor promoter is tightly controlled by NF-Y that binds to a CCAAT box located upstream of the initiation site [1, 2]. In this report, we show that Sp1 plays an essential role in the PDGF beta-receptor transcription. Within the upstream GC rich area there are two Sp1 binding sites located in close proximity to the CCAAT box. Deletion of the GC rich region resulted in a 50% decrease of the transcriptional activity of the promoter, and a complete loss of its responsiveness to over-expression of Sp1. There was an additive effect between NF-Y and Sp I in reporter activity when they were co-transfected together with the promoter-reporter construct. Furthermore, transfection of NF-Y failed to enhance transcriptional activity when the Sp1 binding sites were deleted from the promoter, suggesting an important role for Sp1 in this NF-Y controlled transcription. We have recently reported that c-Myc represses PDGF beta-receptor transcription through its interference with the transactivation activity of NF-Y [3]. In the case of p21(wafl/cip1) transcription, c-Myc was shown to repress its transcription by sequestering Sp1 [4]. However, we could not find any effect of Sp1 in the c-Myc-mediated repression on the PFDGF beta-receptor promoter, since the deletion of SpI binding sites could not attenuate the repression by c-Myc on the promoter activity. PMID: 12153142 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 54: Gastroenterology. 2002 Apr;122(4):1020-34. Interferon-alpha activates multiple STAT signals and down-regulates c-Met in primary human hepatocytes. Radaeva S, Jaruga B, Hong F, Kim WH, Fan S, Cai H, Strom S, Liu Y, El-Assal O, Gao B. Section on Liver Biology, Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland 20892, USA. BACKGROUND & AIMS: Interferon (IFN)-alpha therapy is currently the primary choice for viral hepatitis and a promising treatment for hepatocellular carcinoma (HCC). Primary mouse and rat hepatocytes respond poorly to IFN-alpha stimulation. Thus, it is very important to examine the IFN-alpha signal pathway in primary human hepatocytes. METHODS: The IFN-alpha-activated signals and genes in primary human hepatocytes and hepatoma cells were examined by Western blotting and microarray analyses. RESULTS: Primary human hepatocytes respond very well to IFN-alpha stimulation as shown by activation of multiple signal transducer and activator of transcription factor (STAT) 1, 2, 3, 5, and multiple genes. The differential response to IFN-alpha stimulation in primary human and mouse hepatocytes may be caused by expression of predominant functional IFN-alpha receptor 2c (IFNAR2c) in primary human hepatocytes vs. expression of predominant inhibitory IFNAR2a in mouse hepatocytes. Microarray analyses of primary human hepatocytes show that IFN-alpha up-regulates about 44 genes by over 2-fold and down-regulates about 9 genes by 50%. The up-regulated genes include a variety of antiviral and tumor suppressors/proapoptotic genes. The down-regulated genes include c-myc and c-Met, the hepatocyte growth factor (HGF) receptor. Down-regulation of c-Met is caused by IFN-alpha suppression of the c-Met promoter through down-regulation of Sp1 binding and results in attenuation of HGF-induced signals and cell proliferation. CONCLUSIONS: IFN-alpha directly targets human hepatocytes, followed by activation of multiple STATs and regulation of a wide variety of genes, which may contribute to the antiviral and antitumor activities of IFN-alpha in human liver. PMID: 11910354 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 55: Mol Cell. 2002 Jan;9(1):133-43. Direct interaction of c-Myc with Smad2 and Smad3 to inhibit TGF-beta-mediated induction of the CDK inhibitor p15(Ink4B). Feng XH, Liang YY, Liang M, Zhai W, Lin X. Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA. xialin@bcm.tmc.edu The c-Myc oncogene has been implicated in the genesis of diverse human tumors. Ectopic expression of the c-Myc gene in cultured epithelial cells causes resistance to the antiproliferative effects of TGF-beta. However, little is known about the precise mechanisms of c-Myc-mediated TGF-beta resistance. In this study, we reveal that c-Myc physically interacts with Smad2 and Smad3, two specific signal transducers involved in TGF-beta signaling. Through its direct interaction with Smads, c-Myc binds to the Sp1-Smad complex on the promoter of the p15(Ink4B) gene, thereby inhibiting the TGF-beta-induced transcriptional activity of Sp1 and Smad/Sp1-dependent transcription of the p15(Ink4B) gene. These results suggest that oncogenic c-Myc promotes cell growth and cancer development partly by inhibiting the growth inhibitory functions of Smads. PMID: 11804592 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 56: Biochemistry. 2002 Jan 29;41(4):1229-40. A polypyrimidine/polypurine tract within the Hmga2 minimal promoter: a common feature of many growth-related genes. Rustighi A, Tessari MA, Vascotto F, Sgarra R, Giancotti V, Manfioletti G. Dipartimento di Biochimica, Biofisica e Chimica delle Macromolecole, Universita di Trieste, Italy. HMGA2 is an architectural nuclear factor which plays an important role in development and tumorigenesis, but mechanisms regulating its expression are largely unknown. The proximal promoters of the mouse and human genes coding for HMGA2 contain a conserved polypyrimidine/polypurine (ppyr/ppur) element which constitutes a multiple binding site for Sp1 and Sp3 transcription factors. In the present study we report that this region can adopt a single-stranded DNA conformation, as demonstrated in vitro by S1 nuclease sensitivity on supercoiled plasmids, indicative of an intramolecular triple-helical H-DNA structure. Moreover, we find that PTB (polypyrimidine tract binding protein), a member of the hnRNP family, binds the pyrimidine strand of Hmga2 as well as similar ppyr/ppur elements of the c-Ki-ras (R.Y) and c-myc P1 promoters. Transfection experiments indicate that non-B-DNA conformers of the ppyr/ppur tract of the Hmga2 promoter contribute to positive transcriptional activity. We propose a transcriptional mechanism, acting on the Hmga2 non-B-DNA structure and functioning through interconversion between double-stranded and single-stranded DNA, that seems to be adopted by an increasing number of genes, mainly growth-related. PMID: 11802722 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 57: J Clin Invest. 2001 Nov;108(10):1541-7. Arsenic inhibition of telomerase transcription leads to genetic instability. Chou WC, Hawkins AL, Barrett JF, Griffin CA, Dang CV. Program in Human Genetics and Molecular Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA. Arsenic is effective in the treatment of acute promyelocytic leukemia. Paradoxically, it is also carcinogenic. In the process of elucidating a mechanism of arsenic resistance in a leukemia cell line, NB4, we discovered that arsenic exposure causes chromosomal abnormalities, with a preponderance of end-to-end fusions. These chromosomal end fusions suggested that telomerase activity may be inhibited by arsenic. We found that arsenic inhibits transcription of the hTERT gene, which encodes the reverse transcriptase subunit of human telomerase. This effect may in part be explained by decreased c-Myc and Sp1 transcription factor activities. Decreased telomerase activity leads to chromosomal end lesions, which promote either genomic instability and carcinogenesis or cancer cell death. These phenomena may explain the seemingly paradoxical carcinogenic and antitumor effects of arsenic. PMID: 11714746 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 58: J Clin Invest. 2001 Nov;108(9):1341-8. Comment in: J Clin Invest. 2001 Nov;108(10):1425-7. Hyperglycemia inhibits endothelial nitric oxide synthase activity by posttranslational modification at the Akt site. Du XL, Edelstein D, Dimmeler S, Ju Q, Sui C, Brownlee M. Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York 10461, USA. Endothelial nitric oxide synthase (eNOS) is activated by phosphorylation of serine 1177 by the protein kinase Akt/PKB. Since hyperglycemia-induced mitochondrial superoxide overproduction increases O-linked N-acetylglucosamine modification and decreases O-linked phosphorylation of the transcription factor Sp1, the effect of hyperglycemia and the hexosamine pathway on eNOS was evaluated. In bovine aortic endothelial cells, hyperglycemia inhibited eNOS activity 67%, and treatment with glucosamine had a similar effect. Hyperglycemia-associated inhibition of eNOS was accompanied by a twofold increase in O-linked N-acetylglucosamine modification of eNOS and a reciprocal decrease in O-linked serine phosphorylation at residue 1177. Both the inhibition of eNOS and the changes in its post-translational modifications were reversed by antisense inhibition of glutamine:fructose-6-phosphate amidotransferase, the rate-limiting enzyme of the hexosamine pathway, or by blocking mitochondrial superoxide overproduction with uncoupling protein-1 (UCP-1) or manganese superoxide dismutase (MnSOD). Immunoblot analysis of cells expressing myc-tagged wild-type human eNOS confirmed the reciprocal increase in O-linked N-acetylglucosamine and decrease in O-linked serine 1177 phosphorylation in response to hyperglycemia. In contrast, when myc-tagged human eNOS carried a mutation at the Akt phosphorylation site (Ser1177), O-linked N-acetylglucosamine modification was unchanged by hyperglycemia and phospho-eNOS was undetectable. Similar changes in eNOS activity and covalent modification were found in aortae from diabetic animals. Chronic impairment of eNOS activity by this mechanism may partly explain the accelerated atherosclerosis of diabetes. PMID: 11696579 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 59: Int J Oncol. 2001 Oct;19(4):755-61. c-Myc and Sp1/3 are required for transactivation of hamster telomerase catalytic subunit gene promoter. Park NH, Guo W, Kim HR, Kang MK, Park NH. Dental and Craniofacial Research Institute, University of California, Los Angeles 90095-1668, USA. npark@dent.ucla.edu The hamster and human TERT promoters share common critical protein binding sites, such as the GC-box or E-box, which is known to be a binding site for Sp1/Sp3 transcriptional factors and c-Myc, respectively. Our previous data demonstrated that Sp1/Sp3 synergistically transactivate the hamster TERT (hamTERT) promoter. In this study, we determined the role of c-Myc in the regulation of hamTERT, and analyzed the relative significance of GC-boxes and the E-box for transcriptional activation of hamster TERT. Wild-type, mutated E-box or mutated GC-box hamTERT core promoter reporter was introduced into 293T cells in combination with murine or human Myc expression vectors. The promoter activity was determined using the luciferase assay, and the transfection efficiency was normalized with CAT activity. The electrophoretic mobility shift assay (EMSA) was done to prove the nuclear protein binding activity of the GC-box (region II) or E-box. Overexpression of murine or human Myc transactivated hamTERT core promoter activity. Inversion mutation in the E-box or substitution mutation in the GC-boxes abrogated endogenous or Myc induced hamTERT transactivation. Region II is the single most important Sp1/3 binding site in transcriptional activation, and multiple combined mutations in the GC-boxes abolished the hamTERT promoter activity. These results indicate that c-Myc and Sp1/3 are the major regulatory determinants of the hamTERT transcriptional activation. The mechanism of TERT gene activation during immortalization and carcinogenesis may be conserved among species. PMID: 11562751 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 60: Biochem Biophys Res Commun. 2001 Sep 21;287(2):313-22. Parathyroid hormone (PTH) suppresses rat PTH/PTH-related protein receptor gene promoter. Kawane T, Mimura J, Fujii-Kuriyama Y, Horiuchi N. Department of Biochemistry, Ohu University School of Dentistry, Koriyama 963-8611, Japan. Parathyroid hormone (PTH) regulates osteoblasts via a G protein-linked PTH/PTH-related protein (PTHrP) receptor. PTH effects on PTH/PTHrP receptor gene expression were studied in UMR 106 osteoblast-like cells. In heterogeneous nuclear RNA and Northern analysis, PTH suppressed PTH/PTHrP receptor transcription. We cloned the 7-kb promoter region of the rat PTH/PTHrP receptor gene and transiently transfected chimeric deletion constructs containing the 5'-flanking region and the luciferase gene into UMR 106 cells. In transfected cells the minimal region for basal promoter activity was between positions -128 and +103. The 5'-flanking region of exon U1 contained several putative-binding sites for Sp1 and the myc-associated zinc finger protein (MAZ). The minimal PTH-suppressive region (PTHSR) was between +1 and +25 in exon U1, but the 5'-flanking region or Sp1 and MAZ-binding sites also were required for PTH-mediated repression. By gel mobility shift assay PTH markedly decreased binding of PTHSR-protein complex in UMR 106 cells. The mutation experiments showed that the most critical sequence for the repression of PTH was 5'-GGGGGAGGGGAG-3' (+1 to +12) of PTHSR. This represents the first characterization of a PTH-suppressive region of the PTH/PTHrP receptor gene in rat. Copyright 2001 Academic Press. PMID: 11554727 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 61: Gene. 2001 Jul 11;272(1-2):173-9. Necdin acts as a transcriptional repressor that interacts with multiple guanosine clusters. Matsumoto K, Taniura H, Uetsuki T, Yoshikawa K. Division of Regulation of Macromolecular Functions, Institute for Protein Research, Osaka University, Yamadaoka 3-2, Suita, Osaka 565-0871, Japan. Necdin is a growth suppressor expressed predominantly in postmitotic neurons, and ectopic expression of this protein suppresses cell growth. Here we report that Necdin directly binds to specific DNA sequences and serves as a transcriptional repressor. Polyhistidine-tagged Necdin was used for selection of random-sequence oligonucleotides by polymerase chain reaction-based amplification. Necdin recognized guanosine (G)-rich sequences that encompass multiple G clusters and intervening mono- or di-nucleotides of A, T and C. These sequences, termed GN boxes, resemble multiply aligned forms of the canonical GC box which is recognized by Sp family members. Necdin directly bound to a GN box consisting of contiguous two GC boxes with four G clusters, but not to a single GC box with two G clusters, whereas Sp1 bound to both. In a reporter system using Drosophila Schneider Line 2 cells, Necdin repressed Sp1-dependent activity of mouse c-myc P1 promoter that contains a typical GN box. Deletion of the GN box from the c-myc P1 promoter or its conversion to the single GC box abolished the Necdin-dependent repression. These results suggest that Necdin modulates gene transcription via the GN box that is potentially recognized by GC box-targeting Sp family members. PMID: 11470523 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 62: Nucleic Acids Res. 2001 Jul 15;29(14):3006-11. Telomerase activation by histone deacetylase inhibitor in normal cells. Takakura M, Kyo S, Sowa Y, Wang Z, Yatabe N, Maida Y, Tanaka M, Inoue M. Department of Obstetrics and Gynecology, Kanazawa University, School of Medicine, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8641, Japan. Although telomerase activity is known to be regulated mainly at the level of transcription of the human telomerase catalytic subunit (hTERT) gene, the molecular mechanism underlying tumor-specific expression of telomerase remains unclear. Emerging evidence suggests that reversible acetylation of nucleosomal histones and the resultant changes in the chromatin structure are important processes in gene transcription. In particular, histone deacetylase (HDAC) inhibitors activate the transcription of certain genes by altering the acetylation status of nucleosomal histones. The present study examines the effects of HDAC inhibitor on hTERT gene transcription. Treatment with tricostatin A (TSA) induced significant activation of hTERT mRNA expression and telomerase activity in normal cells, but not in cancer cells. Transient expression assays revealed that TSA activates the hTERT promoter. Furthermore, the proximal 181 bp core promoter of hTERT, which contains two c-Myc and five Sp1 sites, was determined to be the responsible element. Overexpression of Sp1 enhanced responsiveness to TSA, and mutation of Sp1 sites, but not c-Myc sites, of the core promoter of hTERT abrogated this activation. Introduction of the dominant-negative form of the Sp family inhibited TSA activation. These results indicate that HDAC inhibitor activates the hTERT promoter in normal cells, in which Sp1 plays a key role. This finding suggests one way whereby histone deacetylation may be involved in silencing the hTERT gene in normal cells. PMID: 11452025 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 63: J Biol Chem. 2001 Aug 10;276(32):30429-34. Epub 2001 Jun 6. Two consecutive zinc fingers in Sp1 and in MAZ are essential for interactions with cis-elements. Song J, Ugai H, Ogawa K, Wang Y, Sarai A, Obata Y, Kanazawa I, Sun K, Itakura K, Yokoyama KK. RIKEN (The Institute of Physical and Chemical Research), Tsukuba Institute, Bioresource Center, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan. The zinc finger proteins Sp1 and Myc-associated zinc finger protein (MAZ) are transcription factors that control the expression of various genes. Regulation of transcription by these factors is based on interactions between GC-rich DNA-binding sites (GGGCGG for Sp1 and GGGAGGG for MAZ) and the carboxyl-terminal zinc finger motifs of the two proteins. Sp1 and MAZ have three and six zinc fingers, respectively, and the details of their interactions with cis-elements remain to be clarified. We demonstrate here that Sp1 and MAZ interact with the same GC-rich DNA-binding sites, apparently sharing DNA-binding sites with each other. We found that the DNA binding activities of Sp1 and MAZ depended mainly on consecutive zinc fingers, namely the second and third zinc fingers in Sp1 and the third and fourth zinc fingers in MAZ. Furthermore, the interactions of the zinc finger proteins with the same cis-elements appear to play a critical role in the regulation of gene expression. It seems plausible that two consecutive zinc finger motifs in a zinc finger protein might be essential for interaction of the protein with DNA. PMID: 11395515 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 64: J Virol. 2001 Jun;75(12):5559-66. Telomerase activation by human papillomavirus type 16 E6 protein: induction of human telomerase reverse transcriptase expression through Myc and GC-rich Sp1 binding sites. Oh ST, Kyo S, Laimins LA. Department of Microbiology-Immunology, Northwestern University Medical School, Chicago, Illinois 60611, USA. High-risk human papillomaviruses (HPVs) immortalize keratinocytes by disrupting the retinoblastoma protein (Rb)/p16 pathway and activating telomerase. The E7 oncoprotein targets Rb, while the E6 oncoprotein induces telomerase activity in human keratinocytes. This study has examined the mechanism by which E6 activates telomerase. Expression of human telomerase reverse transcriptase (hTERT), the catalytic subunit of telomerase, was found to be increased in keratinocytes stably expressing HPV type 16 E6, suggesting that E6 acts to increase hTERT transcription. hTERT expression and telomerase activity were activated to significantly higher levels in cells expressing both E6 and E7 than in cells expressing E6 alone. This indicates that E7 may augment E6-mediated activation of hTERT transcription. In transient-transfection assays using hTERT reporters, the induction of hTERT expression by E6 was found to be mediated by a 258-bp fragment of the hTERT promoter, proximal to the ATG initiation codon. Previous studies have demonstrated that overexpression of Myc can activate hTERT expression, suggesting that Myc may be a mediator of E6-mediated hTERT induction. However, in cells stably expressing E6, no strict correlation between the level of Myc and the activation of hTERT was found. Consistent with this observation, mutation of the two Myc binding sites in the hTERT promoter only modestly reduced responsiveness to E6 in transient reporter assays. This indicates that activation of Myc-dependent transcription is not essential for E6-mediated upregulation of hTERT expression. The hTERT promoter also contains five GC-rich elements that can bind Sp1. Mutation of these sites within the 258-bp fragment partially reduced hTERT induction by E6. However, when mutations in the Sp1 sites were combined with the mutated Myc binding sites, all activation by E6 was lost. This indicates that it is the combinatorial binding of factors to Myc and Sp1 cis elements that is responsible for hTERT induction by E6. PMID: 11356963 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 65: J Biol Chem. 2001 Jun 22;276(25):22016-23. Epub 2001 Mar 28. Mechanism for the reduction of telomerase expression during muscle cell differentiation. Nozawa K, Maehara K, Isobe K. Department of Basic Gerontology, National Institute for Longevity Sciences, 36-3 Gengo, Morioka-cho, Obu, Aichi 474-8522, Japan. Telomerase, the reverse transcriptase that maintains telomere DNA, is usually undetectable in adult human tissues, but is positive in embryonic tissues and in cancers. However, in rodents, several organs of normal adult animals express substantial amounts of telomerase activity. To elucidate relevant control mechanisms operating on the tissue-specific expression of telomerase in rodents, we examined the transcriptional regulation of telomerase reverse transcriptase (mTERT) gene in muscle cell differentiation. Reverse transcriptase-polymerase chain reaction analysis showed that the reduction of telomerase activity was caused by the decrease of mTERT mRNA level during myogenesis. Transfections of mTERT promoter showed that the proximal 225-base pair region is the core promoter responsible for basal transcriptional activity and also participates in the reduced transcription after muscle differentiation. Electrophoretic mobility shift assays showed that this region contained the GC-boxes, which bind to Sp1 family proteins, and the E-box, which binds to c-Myc. Furthermore, DNA binding activities of Sp1, Sp3, and c-Myc were down-regulated during myogenesis. These data suggest that Sp1, Sp3, and c-Myc have critical roles of TERT transactivation in mouse, and the lack of these transcription factors cause down-regulation of mTERT gene expression in muscle cells differentiation. PMID: 11279234 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 66: Proc Natl Acad Sci U S A. 2001 Apr 10;98(8):4510-5. Epub 2001 Mar 27. Myc represses the p21(WAF1/CIP1) promoter and interacts with Sp1/Sp3. Gartel AL, Ye X, Goufman E, Shianov P, Hay N, Najmabadi F, Tyner AL. Department of Molecular Genetics, University of Illinois College of Medicine, Chicago, IL 60607, USA. agartel@uic.edu The cyclin-dependent kinase inhibitor p21((WAF1/CIP1)) inhibits proliferation both in vitro and in vivo, and overexpression of p21 in normal and tumor cell lines results in cell cycle arrest. In contrast, ectopic expression of Myc alleviates G(1) cell cycle arrest. Recent studies showed that Myc can repress p21 transcription, thereby overriding a p21-mediated cell cycle checkpoint. We found that activation of a Myc-estrogen receptor fusion protein by 4-hydroxytamoxifen in mouse cells resulted in suppression of endogenous p21 transcription. This effect was observed in the absence of de novo protein synthesis and was independent of histone deacetylase activity. In transient transfection studies, Myc effectively repressed p21 promoter constructs containing only 119 bp of sequence upstream of the transcription start site. This region contains multiple Sp1-binding sites and a potential initiator element, but no canonical Myc DNA-binding sites. Deletion of the potential initiator element does not affect repression of the p21 promoter by c-Myc. Coimmunoprecipitation and glutathione S-transferase pull-down experiments demonstrate that c-Myc may form complexes with Sp1/Sp3. We found that the central region of c-Myc interacts with the zinc finger domain of Sp1. Because Sp1 is required for p21 transcription, it is possible that Myc may down-regulate p21 transcription, at least in part, by sequestering Sp1. Repression of the p21 promoter may contribute to the ability of c-Myc to promote cell proliferation. PMID: 11274368 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 67: J Biol Chem. 2001 Jun 8;276(23):19897-904. Epub 2001 Mar 19. Independent repression of a GC-rich housekeeping gene by Sp1 and MAZ involves the same cis-elements. Song J, Ugai H, Kanazawa I, Sun K, Yokoyama KK. RIKEN (The Insitute of Physical & Chemical Research), Tsukuba Institute, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan. The transcription factors Sp1 and MAZ (Myc-associated zinc finger protein) contain several zinc finger motifs, and each functions as both a positive and a negative regulator of gene expression. In this study, we characterized the extremely GC-rich promoter of the human gene for MAZ, which is known as a housekeeping gene. Unique symmetrical motifs in the promoter region (nucleotides -383 to -334) were essential for the expression of the gene for MAZ, whereas an upstream silencer element (nucleotides -784 to -612) was found to act in a position-dependent but orientation-independent manner. Sp1 and MAZ bound to the same cis-elements in the GC-rich promoter, apparently sharing DNA-binding sites. The relative extent of binding of Sp1 and MAZ to these cis-elements corresponded to the extent of negative regulation of the expression of the gene for MAZ in various lines of cells. Furthermore, novel repressive domains in both Sp1 (amino acids 622-788) and MAZ (amino acids 127-292) were identified. Suppression by Sp1 and suppression by MAZ were independent phenomena; histone deacetylases were involved in the autorepression by MAZ itself, whereas DNA methyltransferase 1 was associated with suppression by Sp1. Our results indicate that both deacetylation and methylation might be involved in the regulation of expression of a single gene via the actions of different zinc finger proteins that bind to the same cis-elements. PMID: 11259406 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 68: Gene. 2001 Feb 7;264(1):19-27. Characterization and promoter analysis of the mouse gene for transcription factor Sp4. Song J, Mangold M, Suske G, Geltinger C, Kanazawa I, Sun K, Yokoyama KK. RIKEN (The Institute of Physical and Chemical Research), Tsukuba Institute, 3-1-1 Koyadai, Tsukuba, 305-0074, Ibaraki, Japan. Transcription factor Sp4 is a member of the Sp1 family. It functions differently from other members of this family, such as Sp1 and Sp3, and the gene for Sp4 is transcribed in a tissue-specific manner. Recent studies in mice suggest that Sp4 might play an important role in growth, viability, and male fertility. We report here the isolation and characterization of the gene for Sp4 from a mouse genomic library. The mouse gene for Sp4 was about 80 kb in length and it consisted of six exons and five introns. The promoter was found in a CpG island and had a high G+C content. The proximal promoter contained multiple putative binding sites for the transcription factors Sp1 and MAZ but lacked a consensus TATA box. Multiple sites for the initiation of transcription were mapped in a GC-rich region from 286 bp to 211 bp upstream of the ATG triplet at the site of initiation of translation, and all of the sites were either C or G. Transfection experiments and deletion analysis allowed us to localize the promoter to a region that was no more than 93 bp upstream from the first site of initiation of transcription. We also found that ectopic expression of Sp1 and of MAZ, but not of Sp3, suppressed expression of the Sp4 promoter in a dose-dependent manner. PMID: 11245974 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 69: Curr Med Chem. 2001 Jan;8(1):1-8. Analysis of the effects of daunorubicin and WP631 on transcription. Portugal J, Martin B, Vaquero A, Ferrer N, Villamarin S, Priebe W. Departamento de Biologia Molecular y Celular, Instituto de Biologia Molecular de Barcelona, CSIC, Jordi Girona, 18-26, Barcelona, 08034, Spain. jpmbmc@cid.csic.es The proficiency with which anthracyclines and other DNA-binding drugs target certain sequences in eukaryotic promoters offers a potential approach to interfere with the mechanisms that regulate gene expression in tumor cells. An in vitro transcription assay has been used to compare the ability of the bisintercalating anthracycline WP631 and the monointercalating anthracycline daunorubicin in terms of their ability to inhibit initiation of transcription of the adenovirus major late promoter linked to a G-less transcribed DNA template. Both drugs inhibit basal transcription by RNA polymerase II. However, WP631 is approximately 15 times more efficient at inhibiting transcription initiation from an adenovirus promoter containing an upstream Sp1-protein binding site. The differences in the ability of each drug to inhibit transcription initiation appear to be related to the competition between Sp1 and the anthracyclines for binding to the same site. To see whether WP631's strong effect on transcription can also be observed in cells, we compared the effects of WP631 and other anthracyclines on the transcription of the c-myc gene, which promoter contains Sp1 binding sites. The resulting data suggest that WP631 might circumvent some kinds of tumor resistance at rather low drug concentrations, inhibit c-myc expression in some cell lines, and exert its antitumoral effect by inducing apoptosis. Publication Types: Review Review, Tutorial PMID: 11172687 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 70: Biochem Biophys Res Commun. 2001 Jan 26;280(3):684-92. Identification and regulation of human PDE5A gene promoter. Lin CS, Chow S, Lau A, Tu R, Lue TF. Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California 94143-1695, USA. PDE5A gene encodes type 5 phosphodiesterase (PDE5), the principal cGMP-catalyzing enzyme in the penis and the primary target of sildenafil (Viagra). We have isolated a 3.7-kb DNA fragment that contains the human PDE5A gene promoter. The DNA fragment contains a sequence of 234 nucleotides at its 3' end that corresponds to most of the untranslated region of the PDE5A1 mRNA. The remaining 3.5-kb upstream flanking sequence contains no apparent TATA box but has several sequences that resemble binding sequences for transcription factors such as androgen receptor (AR), AP1, AP2, AP4, Sp1, MyoD, Myc, and CArG. In search of promoter activities, we used a luciferase reporter system to examine 10 DNA fragments that cover various regions of the 3.7-kb fragment. We found that a full basal promoter activity was confined to a 139-bp region that includes 78 bp of the PDE5A1-specific first exon. A lesser basal promoter activity was still detectable in a 94-bp fragment that contains the same 78-bp PDE5A1-specific exon plus 16 bp of upstream sequence. Either the 139-bp or the 94-bp promoter fragment responded minimally to cAMP or cGMP (2 mM) stimulation. Longer fragments that contain either a 308-bp 5' extension (from the 138-bp fragment) or a 156-bp 3' extension (all exon sequence) responded at higher levels to cAMP and cGMP stimulation. The 5' and 3' extensions cooperated with each other to provide the highest level of responses to cAMP and cGMP stimulation. DNase I footprint analysis identified four AP2- and two Sp1-binding sites in the 5' extension and four Sp1-binding sites in the 3' extensions. Cyclic AMP and cGMP had similar stimulatory effects on the PDE5A promoter. Copyright 2001 Academic Press. PMID: 11162575 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 71: Cancer Res. 2000 Dec 15;60(24):7170-6. Anti-CD20- and B-cell receptor-mediated apoptosis: evidence for shared intracellular signaling pathways. Mathas S, Rickers A, Bommert K, Dorken B, Mapara MY. University Medical Center Charite, Humboldt University, Robert-Rossle-Klinik, Department of Hematology, Oncology, and Tumor Immunology, Berlin, Germany. Clinical administration of the anti-CD20 antibody IDEC-C2B8 can induce remission of low-grade B-cell lymphoma. Whereas it has been suggested that the main mechanisms of action are complement-mediated and antibody-dependent cell-mediated cytotoxicity, we demonstrate that monoclonal antibody IDEC-C2B8 is a strong inducer of apoptosis in CD20-positive B-cell lymphoma cell lines reflecting different stages of lymphomagenesis. Thus, CD20-dependent apoptosis was inducible in human surface IgM-positive Burkitt's lymphoma cell lines as well as in more mature surface IgM-negative B-cell lymphoma cell lines carrying the t(14;18) translocation. Furthermore, in Burkitt's lymphoma cell lines, we observed a striking correlation between anti-CD20- and B-cell receptor-mediated apoptosis with regard to sensitivity toward the apoptotic stimuli and the execution of the apoptotic pathway. Thus, induction of anti-CD20- or B-cell receptor-mediated apoptosis involved rapid up-regulation of the proapoptotic protein Bax. In addition, we show similar changes in the mRNA expression level of two early response genes, c-myc and Berg36, as well as activation of the mitogen-activated protein kinase family members p44 (extracellular signal-regulated kinase 1) and p42 (extracellular signal-regulated kinase 2) and activation of activator protein 1 (AP-1) DNA binding activity. These data support our hypothesis that both pathways are mediated in part by the same signal-transducing molecules. These results might help explain the resistance and regression of lymphomas to IDEC-C2B8 and give new insights in the signaling cascade after CD20 ligation. PMID: 11156427 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 72: Oncogene. 2000 Nov 23;19(50):5801-9. Transcriptional regulation of the human osteopontin promoter: functional analysis and DNA-protein interactions. Wang D, Yamamoto S, Hijiya N, Benveniste EN, Gladson CL. Department of Pathology, The University of Alabama at Birmingham, 35294, USA. Synthesis of cell attachment proteins and cytokines, such as osteopontin (OPN), can promote tumor cell remodeling of the extracellular matrix into an environment that promotes tumor cell attachment and migration. We investigated the transcriptional regulation of OPN in the U-251MG and U-87MG human malignant astrocytoma cell lines. Deletion and mutagenesis analyses of the OPN promoter region identified a proximal promoter element (-24 to -94 relative to the transcription initiation site) that is essential for maintaining high levels of OPN expression in the tumor cells. This element, designated RE-1, consists of two cis-acting elements, RE-1a (-55 to -86) and RE-1b (-22 to -45), which act synergistically to regulate the activity of the OPN promoter. Gel shift assays using nuclear extracts of U-251MG cells demonstrated that RE-1a contains binding sites for transcription factors Sp1, the glucocorticoid receptor, and the E-box-binding factors, whereas RE-1b contains a binding site for the octamer motif-binding protein (OCT-1/OCT-2). Inclusion of antibodies directed toward Myc and OCT-1 in the gel shift assays indicated that Myc and OCT-1 participate in forming DNA-protein complexes on the RE-1a and RE-1b elements, respectively. Our results identify two previously unrecognized elements in the OPN promoter that act synergistically to promote upregulation of OPN synthesis by tumor cells but are regulated by different transcription factors. PMID: 11126367 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 73: J Mol Endocrinol. 2000 Dec;25(3):309-19. The transcription factors SP1 and MAZ regulate expression of the parathyroid hormone/parathyroid hormone-related peptide receptor gene. Williams LJ, Abou-Samra AB. Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA. The parathyroid hormone (PTH)/PTH-related peptide (PTHrP) receptor regulates extracellular calcium concentrations and is therefore important for mineral homeostasis. ROS 17/2.8 cells, a rat osteoblast-like osteosarcoma cell line, express the PTH/PTHrP receptor and provide a good model for examining the transcriptional regulation of its gene. The rat PTH/PTHrP receptor gene has two promoters, U1 and U3, which were shown to be important for its expression. Using extracts from ROS 17/2.8 cells, we have demonstrated two regions (termed FP1 and FP2) of nuclear protein/DNA interaction within promoter sequences previously shown to be important for the activity of the U3 promoter. Nuclear extracts from rat 2 fibroblasts, which do not express the PTH/PTHrP receptor, produced one site of protein/DNA interaction which was found at a position on the promoter identical to the position of FP1 produced by a ROS 17/2.8 nuclear extract. Mutation of these two sites of protein/DNA interaction resulted in reduced U3 promoter activity. Furthermore, we have demonstrated that the transcription factors SP1 and MAZ regulate U3 promoter expression and have shown their functional significance using mutational analysis. These data demonstrate that SP1 and MAZ bind to the PTH/PTHrP receptor promoter and that they are involved in cell-specific expression of its gene product. PMID: 11116210 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 74: Mol Cells. 2000 Oct 31;10(5):566-74. 5'-Flanking sequence and promoter activity of the rabbit neuronal nitric oxide synthase (nNOS) gene. Jeong Y, Won J, Kim C, Yim J. National Creative Research Initiative Center for Genetic Reprogramming, Institute for Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Korea. We have isolated a rabbit neuronal nitric oxide synthase (nNOS) cDNA encoding a protein of 1,435 amino acids. Using the cDNA clones as probes, the 5'-flanking region of the nNOS gene was isolated from a rabbit genomic DNA library. 5'RACE and primer extension analysis of rabbit brain total RNA mapped multiple transcription initiation sites localized 474-487 bp upstream from the translation start codon. Analysis of 5,197 bp of the 5'-flanking sequence revealed that the rabbit nNOS gene promoter lacks canonical TATA or CCAAT boxes and, instead, contains a GC-rich region and multiple Sp1 sites. Farther from the +1start, various putative cis-elements including AP-1, AP-4, NF-kappaB, STAT, CREB, C/EBP and c-Myc were observed. The functional promoter activity of the 5'-flanking region was demonstrated by its ability to drive the expression of a beta-galactosidase reporter gene in several cell types. Serial deletion analysis of the promoter region revealed that the -291 to -172 region, which contains two Sp1 sites, is essential for basal transcriptional activity. These results suggest that the rabbit nNOS promoter contains characteristics of inducible genes. PMID: 11101149 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 75: Biochem Biophys Res Commun. 2000 Oct 5;276(3):1203-9. Molecular cloning and functional analysis of the promoter region of rat nonmuscle myosin heavy chain-B gene. Yam JW, Chan KW, Li N, Hsiao WL. Department of Biology, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, Kowloon, China. Rat nonmuscle myosin heavy chain-B (r-nmMHC-B) mRNA was previously found downregulated in Rat 6 fibroblasts transformed by mutant p53(val135) [J. W. P. Yam, J. Y. Zheng, and W. L. W. Hsiao (1987) Biochem. Biophys. Res. Commun. 266, 472-480]. Overexpression of exogenous r-nmMHC-B could partially reverse the transforming phenotypes both in vitro and in vivo. The downregulation of r-nmMHC-B was also observed in Rat 6 transformed by c-H-ras and v-myc oncogenes. We cloned a 5.2-kb r-nmMHC-B promoter region. Sequence analysis of -1248 to +1 revealed no TATA box, but did show that it contained CAAT boxes, E12/E47, MyoD, MEF, E2F, CREB, and SP1 binding sites. Based on transient reporter assays, the promoter/enhancer activities were unusually extended to the entire 5.2 kb region in normal Rat 6 cultures, but markedly suppressed in p53(val135)-, and c-H-ras-transformed cells. The activity detected by the reporter assay corresponded to levels of mRNA as analyzed previously by Northern blots in each respective cell line. Thus, the switch-off of the r-nmMHC-B in the transformed cells is very likely controlled by upstream transcriptional factors, which might have been altered in the course of neoplastic transformation. Copyright 2000 Academic Press. PMID: 11027611 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 76: J Autoimmun. 2000 Aug;15(1):75-80. Identification of human autoantibodies to the transcriptional repressor ZF5. Yanagidani A, Matsuoka M, Yokoro K, Tanaka H, Numoto M. Department of Molecular Pathology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Hiroshima, Minami-ku, 734, Japan. ZF5 was originally cloned as a transcriptional repressor on the mouse c-myc promoter. It contains the Kruppel-type zinc fingers and a conserved POZ domain, which is found in a growing number of zinc finger proteins and mediate protein-protein interactions. Autoantibodies against transcription factors are sometimes found in sera from patients with high levels of anti-nuclear antibody (ANA). Using Western blotting with ZF5 and sera of autoimmune disease, we detected one serum, named M6 serum, which contains the antibody against a transcriptional repressor ZF5. The confirmed epitope was specific to ZF5 and was not reactive to the other analogous factors: BCL-6, ZID, and Sp1. This epitope also has a molecular mimicry of the viral proteins. From these results, we predict that viral protein which mimics host ZF5 antigen triggers self-reactive T cell clones and induces the autoantibody in M6 serum after the destruction of host tissues. Copyright 2000 Academic Press. PMID: 10936031 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 77: Cardiovasc Res. 2000 Jun;46(3):511-22. Characterization of the rat connexin40 promoter: two Sp1/Sp3 binding sites contribute to transcriptional activation. Bierhuizen MF, van Amersfoorth SC, Groenewegen WA, Vliex S, Jongsma HJ. University Medical Center Utrecht, Department of Medical Physiology, The Netherlands. m.f.a.bierhuizen@med.uu.nl OBJECTIVES: The gap junction protein connexin40 (Cx40) is differentially expressed during embryonic development and in adult tissues, for which the molecular basis is unknown. In order to elucidate the molecular mechanisms controlling Cx40 expression, we set out to map and characterize its promoter. METHODS: The transcriptional activity of individual rat Cx40 (rCx40)-derived promoter fragments fused to the luciferase reporter gene was determined by transfection/reporter assays in Cx40-expressing (A7r5, rat smooth muscle embryonic thoracic aorta cells, and BWEM, v-myc transformed rat fetal cardiomyocytes) and Cx40-nonexpressing cells (N2A, mouse neuroblastoma cells). The nature of DNA-protein interactions was investigated by a combination of standard electrophoretic-mobility-shift assays (EMSA) and EMSA/antibody supershift assays. RESULTS: Quantification of luciferase activity in cell lysates revealed that a 235-base-pair fragment, in between map positions -150 and +85 relative to the transcription initiation site, is able to provide for a significant level of transcription in both Cx40-expressing (A7r5, BWEM) and -nonexpressing (N2A) cells. These results indicate that this region contains the basal promoter but is not sufficient to completely determine the endogenous Cx40-expression pattern within these cell types. In search for the responsible transcriptional regulatory element(s), additional segments of the (-150, +85) region were deleted and the remaining fragments were tested for transcriptional activity. These studies established that the regions in between map positions (-96, -71) and (+58, +85) contribute to promoter activity. EMSA with these regions revealed that predominantly two DNA-protein complexes are formed upon incubation with either A7r5, BWEM or N2A nuclear extracts, which could be both inhibited by including excess oligonucleotide containing the Sp1 consensus binding site in the binding reaction. Purified recombinant human Sp1 provided also for a shift in the EMSA using these promoter regions as target fragments. When the DNA-protein complexes formed with nuclear extract were subsequently incubated with either an anti-Sp1 or an anti-Sp3 antibody clear supershifts in the EMSA were obtained, indicating Sp1 and Sp3 binding to both the (-98, -64) and (+53, +87) regions. The introduction of mutations within the core sequence of the putative Sp1/Sp3 binding sites present in these regulatory elements reduced the level of transcriptional activity and abrogated Sp1/Sp3 binding to these sites. CONCLUSION: The results indicate that at least two Sp1/Sp3 binding sites in the rCx40 promoter contribute to the transcriptional activation of its gene in cultured cells. PMID: 10912461 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 78: J Biol Chem. 2000 Sep 15;275(37):28539-48. Erratum in: J Biol Chem 2000 Dec 15;275(50):39801. Mitogen-induced expression of the fibroblast growth factor-binding protein is transcriptionally repressed through a non-canonical E-box element. Harris VK, Coticchia CM, List HJ, Wellstein A, Riegel AT. Department of Oncology, Vincent T. Lombardi Cancer Center, Georgetown University, Washington, D. C. 20007, USA. The fibroblast growth factor-binding protein (FGF-BP) stimulates FGF-2-mediated angiogenesis and is thought to play an important role in the progression of squamous cell, colon, and breast carcinomas. 12-O-Tetradecanoylphorbol-13-acetate (TPA) induction of the FGF-BP gene occurs through transcriptional mechanisms involving Sp1, AP-1, and CCAATT/enhancer-binding protein sites in the proximal FGF-BP gene promoter. The level of TPA induction, however, is limited due to the presence of a repressor element that shows similarity to a non-canonical E-box (AACGTG). Mutation or deletion of the repressor element led to enhanced induction by TPA or epidermal growth factor in cervical squamous cell and breast carcinoma cell lines. Repression was dependent on the adjacent AP-1 site, without discernible alteration in the binding affinity or composition of AP-1. We investigated the following two possible mechanisms for E-box-mediated repression: 1) CpG methylation of the core of the E-box element, and 2) binding of a distinct protein complex to this site. Point mutation of the CpG methylation site in the E-box showed loss of repressor activity. Conversely, in vitro methylation of this site significantly reduced TPA induction. In vitro gel shift analysis revealed distinct and TPA-dependent binding of USF1 and USF2 to the repressor element that required nucleotides within the E-box. Furthermore, chromatin immunoprecipitation assay showed that USF, c-Myc, and Max proteins were associated with the FGF-BP promoter in vivo. Overall, these findings suggested that the balance between trans-activation by AP-1 and repression through the E-box is an important control mechanism for fine-tuning the angiogenic response to growth factor-activated pathways. PMID: 10871606 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 79: Biochem J. 2000 Jun 15;348 Pt 3:675-86. Sequence of the 5'-flanking region and promoter activity of the human mucin gene MUC5B in different phenotypes of colon cancer cells. Van Seuningen I, Perrais M, Pigny P, Porchet N, Aubert JP. Unite INSERM 377, Place de Verdun, 59045 Lille Cedex, France. vanseuni@lille.inserm.fr Control of gene expression in intestinal cells is poorly understood. Molecular mechanisms that regulate transcription of cellular genes are the foundation for understanding developmental and differentiation events. Mucin gene expression has been shown to be altered in many intestinal diseases and especially cancers of the gastrointestinal tract. Towards understanding the transcriptional regulation of a member of the 11p15.5 human mucin gene cluster, we have characterized 3.55 kb of the 5'-flanking region of the human mucin gene MUC5B, including the promoter, the first two exons and the first intron. We report here the promoter activity of successively 5'-truncated sections of 956 bases of this region by fusing it to the coding region of a luciferase reporter gene. The transcription start site was determined by primer-extension analysis. The region upstream of the transcription start site is characterized by the presence of a TATA box at bases -32/-26, DNA-binding elements for transcription factors c-Myc, N-Myc, Sp1 and nuclear factor kappaB as well as putative activator protein (AP)-1-, cAMP-response-element-binding protein (CREB)-, hepatocyte nuclear factor (HNF)-1-, HNF-3-, TGT3-, gut-enriched Kruppel factor (GKLF)-, thyroid transcription factor (TTF)-1- and glucocorticoid receptor element (GRE)-binding sites. Intron 1 of MUC5B was also characterized, it is 2511 nucleotides long and contains a DNA segment of 259 bp in which are clustered eight tandemly repeated GA boxes and a CACCC box that bind Sp1. AP-2alpha and GATA-1 nuclear factors were also shown to bind to their respective cognate elements in intron 1. In transfection studies the MUC5B promoter showed a cell-specific activity as it is very active in mucus-secreting LS174T cells, whereas it is inactive in Caco-2 enterocytes and HT-29 STD (standard) undifferentiated cells. Within the promoter, maximal transcription activity was found in a segment covering the first 223 bp upstream of the transcription start site. Finally, in co-transfection experiments a transactivating effect of Sp1 on to MUC5B promoter was seen in LS174T and Caco-2 cells. PMID: 10840001 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 80: Biochim Biophys Acta. 2000 Apr 25;1491(1-3):57-64. Promoter analysis of the human ubiquitin-conjugating enzyme gene family UBE2L1-4, including UBE2L3 which encodes UbcH7. Ardley HC, Moynihan TP, Markham AF, Robinson PA. Molecular Medicine Unit, University of Leeds, Clinical Sciences Building, St. James's University Hospital, Leeds, UK. rmrhca@stjames.leeds.ac.uk UbcH7 is a ubiquitin-conjugating enzyme mediating c-fos degradation, transcription factor NF-kappaB maturation, human papilloma virus-mediated p53 and Myc protein degradation, in vitro. Previously, we characterised a highly dispersed gene family, UBE2L1-UBE2L4, whose members could potentially encode different isoforms of the UbcH7 protein. UBE2L3, located at chromosome 22q11.2, is the only identified family member with introns and encodes a polypeptide sequence identical to that of UbcH7. Promoter characterisation of UBE2L1, UBE2L3 and UBE2L4 5'-upstream regions was performed to establish which are transcribed under normal physiological conditions and after heat shock. Promoter activity was observed only with the UBE2L3 construct, the minimal promoter lying within a region 100 bp upstream of the transcriptional start site. No evidence for the presence of UBE2L1 or UBE2L4 transcripts was observed in human or murine tissues and cell lines. These data strongly suggest that UBE2L1 and UBE2L4 are likely to encode pseudogenes. Sequencing revealed that the UBE2L3 promoter contained no TATA or CCAAT boxes. Protein:DNA interaction studies confirmed the presence of binding sites for the transcription factors AP2 and Sp1 in the UBE2L3 minimal promoter. Deletion of these binding sites indicated that these factors are crucial for transcription of this gene. PMID: 10760570 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 81: Nucleic Acids Res. 2000 Feb 15;28(4):932-9. Structure and expression of the human p68 RNA helicase gene. Rossler OG, Hloch P, Schutz N, Weitzenegger T, Stahl H. Medizinische Biochemie und Molekularbiologie, Universitat des Saarlandes, 66421 Homburg, Germany. Nuclear DEAD box protein p68 is immunologically related to SV40 large tumour antigen and both proteins possess RNA helicase activity. In this report, we describe the structural organisation of the human p68 gene and aspects of the regulation of its expression. Northern blot and primer extension analyses indicate that, although its level is variable, the p68 RNA helicase appears to be expressed from a single transcription start site in all tissues tested. Sequence analysis revealed that the p68 promoter harbours a 'TATA', a 'CAAT' and an initiator element and contains high affinity binding sites for Sp1, AP-2, CRE and Myc. This and functional promoter analyses in transient expression assays suggest that transcriptional regulation of the p68 gene is complex. Furthermore, there are indications that p68 expression is also regulated post-transcriptionally. Steady-state pools of poly(A)(+)RNA from human cells contain completely spliced p68 mRNA and alternatively spliced forms that contain introns 8-11 or 8-12 (from a total of 12 introns) and are not translated. Analysis of a conditionally p68-overproducing HeLa cell line points to negative autoregulation at the level of splicing, which is confirmed by a recently reported association of p68 with spliceosomes in human cells. PMID: 10648785 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 82: Nucleic Acids Res. 2000 Feb 1;28(3):800-8. Activation of c-myc promoter P1 by immunoglobulin kappa gene enhancers in Burkitt lymphoma: functional characterization of the intron enhancer motifs kappaB, E box 1 and E box 2, and of the 3' enhancer motif PU. Wittekindt NE, Hortnagel K, Geltinger C, Polack A. GSF-National Research Center for Environment and Health, Institute of Clinical Molecular Biology and Tumor Genetics, Marchioninistrasse 25, D-81377 Munich, Germany. wittekin@biochem.mpg.de Deregulated expression of the proto-oncogene c- myc in Burkitt lymphoma (BL) cells carrying a t(2;8) translocation is mediated by a synergistic interaction of the translocated immunoglobulin (Ig) kappa gene intron (kappaEi) and 3' (kappaE3') enhancers and characterized by a strong activation of the promoter P1. We have investigated the functional role of distinct kappa enhancer sequence motifs in P1 activation on both mini-chromosomes and reporter gene constructs. Stable and transient transfections of BL cells revealed critical roles of the kappaEi and kappaE3' elements kappaB and PU, respectively. Joint mutation of kappaB and PU completely abolished P1 activity, implying that an interaction of kappaB- and PU-binding factors is essential for the enhancer synergism. Mutation of the E box 1 and E box 2 motifs markedly decreased P1 activity in transient but not in stable transfection experiments. Co-expression of the NF-kappaB subunit p65(RelA) and Sp1, an essential factor for P1 transcription, in Drosophila melanogaster SL2 cells synergistically enhanced promoter activity. Our results support a model which proposes cross-talk between promoter and enhancer binding factors as the basic mechanism for kappa enhancer-mediated c- myc activation in BL cells. PMID: 10637333 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 83: Nucleic Acids Res. 2000 Feb 1;28(3):669-77. Sp1 cooperates with c-Myc to activate transcription of the human telomerase reverse transcriptase gene (hTERT). Kyo S, Takakura M, Taira T, Kanaya T, Itoh H, Yutsudo M, Ariga H, Inoue M. Department of Obstetrics and Gynecology, Kanazawa University, School of Medicine, 13-1 Takaramachi, Kanazawa, Ishikawa 920-0934, Japan, satoruky@med.kanazawa-u.ac.jp Telomerase activation is thought to be a critical step in cellular immortalization and carcinogenesis. The human telomerase catalytic subunit (hTERT) is a rate limiting determinant of the enzymatic activity of human telomerase. In the previous study, we identified the proximal 181 bp core promoter responsible for transcriptional activity of the hTERT gene. To identify the regulatory factors of transcription, transient expression assays were performed using hTERT promoter reporter plasmids. Serial deletion assays of the core promoter revealed that the 5'-region containing the E-box, which binds Myc/Max, as well as the 3'-region containing the GC-box, which binds Sp1, are essential for transactivation. The mutations introduced in the E-box or GC-box significantly decreased transcriptional activity of the promoter. Overexpression of Myc/Max or Sp1 led to significant activation of transcription in a cell type-specific manner, while Mad/Max introduction repressed it. However, the effects of Myc/Max on transactivation were marginal when Sp1 sites were mutated. Western blot analysis using various cell lines revealed a positive correlation between c-Myc and Sp1 expression and transcriptional activity of hTERT. Using fibroblast lineages in different stages of transformation, we found that c-Myc and Sp1 were induced to a dramatic extent when cells overcame replicative senescence and obtained immortal characteristics, in association with telomerase activation. These findings suggest that c-Myc and Sp1 cooperatively function as the major determinants of hTERT expression, and that the switching functions of Myc/Max and Mad/Max might also play roles in telomerase regulation. PMID: 10637317 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 84: Proc Assoc Am Physicians. 1999 Sep-Oct;111(5):438-47. Heme oxygenase: recent advances in understanding its regulation and role. Elbirt KK, Bonkovsky HL. Department of Medicine, University of Massachusetts Medical School, Worcester, USA. Heme oxygenase (HO) is responsible for the physiological breakdown of heme into equimolar amounts of biliverdin, carbon monoxide, and iron. Three isoforms (HO-1, HO-2, and HO-3) have been identified. HO-1 is ubiquitous and its mRNA and activity can be increased several-fold by heme, other metalloporphyrins, transition metals, and stimuli that induce cellular stress. HO-1 is recognized as a major heat shock/stress response protein. Recent work from our laboratory has demonstrated several potential consensus regulatory elements in the 5'-untranslated region (UTR) of HO-1, including activator protein 1 (AP-1), metal responsive element (MRE), oncogene c-myc/max heterodimer binding site (Myc/Max), antioxidant response element (ARE), and GC box binding (Sp1) sites. Using deletion-reporter gene constructs, we have mapped sites that mediate the arsenite-dependent induction of HO-1, and we have shown that components of the extracellular signal-regulated kinase (ERK) and p38 (a homologue of the yeast HOG1 kinase), but not c-jun N-terminal kinase (JNK), mitogen-activated protein (MAP) kinase pathways are involved in arsenite-dependent upregulation. In contrast, HO-2 is present chiefly in the brain and testes and is virtually uninducible. HO-3 has very low activity; its physiological function probably involves heme binding. Products of the HO reaction have important effects: carbon monoxide is a potent vasodilator, which is thought to play a key role in the modulation of vascular tone, especially in the liver under physiological conditions, and in many organs under "stressful" conditions associated with HO-1 induction. Biliverdin and its product bilirubin, formed in most mammals, are potent antioxidants. In contrast, "free" iron increases oxidative stress and regulates the expression of many mRNAs (e.g., DCT-1, ferritin, and transferrin receptor) by affecting the conformation of iron regulatory protein (IRP)-1 and its binding to iron regulatory elements (IREs) in the 5'- or 3'-UTRs of the mRNAs. Publication Types: Review Review, Tutorial PMID: 10519165 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 85: Thromb Res. 1999 Sep 1;95(5):255-62. The promoter of human tissue factor pathway inhibitor gene: identification of potential regulatory elements. Petit L, Lesnik P, Dachet C, Hugou I, Moreau M, Chapman J, Rouis M. Institut National de la Sante et de la Recherche Medicale, Unite 321, Lipoproteins and Atherogenesis, Hopital de la Pitie-Salpetriere, Paris, France. Tissue factor pathway inhibitor is the major potent physiologic inhibitor of tissue factor-induced coagulation. Several potential binding sites for transcription factors have been described in the 750 bp of the 5' flanking region of the human tissue factor pathway inhibitor gene reported earlier. To identify elements that regulate the expression of tissue factor pathway inhibitor in endothelial, hepatocyte, and monocyte cells, the sequence of an additional 770 bp of tissue factor pathway inhibitor was determined. Comparison of this new sequence as well as that reported earlier with consensus sequences for transcription factor binding sites provided matches for GATA-2, SP1, and c-Myc sequences. Moreover, plasmids containing deletion mutants of the 5' tissue factor pathway inhibitor promoter region and the luciferase reporter gene were transfected into HepG2, ECV304, and THP1 cells. Three negative regulatory elements were localized between -548 to -390, - 390 to -75, and -1158 to -796 relative to the transcriptional start, respectively, in HepG2, ECV304 and THP-1 cells. PMID: 10515290 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 86: J Immunol. 1999 Oct 15;163(8):4292-9. Inhibition of CD28 expression by oligonucleotide decoys to the regulatory element in exon 1 of the CD28 gene. Tam RC, Lin CJ, Lim C, Pai B, Stoisavljevic V. Immunology Laboratory, ICN Research Center, Costa Mesa, CA 92626, USA. rctam@icnpharm.com Ligation of CD28 provides a costimulatory signal essential for Ag-mediated T cell activation via the TCR. Previously we demonstrated that inhibition of human and murine CD28 expression by a guanosine (G)-rich oligonucleotide (ODN), GR1, led to immunosuppression in vitro and in vivo. The bioactivity of GR1 was dependent on a G-rich DNA sequence motif consisting of two G tetrads separated by four nucleotides, (G4N4G4). We have shown recently that a G-rich region, designated CD28GR, in exon 1 of the CD28 gene is such a motif and is a positive regulatory element that binds the transcription factors Sp1 and EGR-1. Here we showed that the bioactivity of GR1 and the related GR2 correlated with the sequence-specific formation of distinct nuclear protein complexes and a high degree of ODN secondary structure. In addition, these ODN blocked transcription factor binding to CD28GR (also in a sequence-specific manner) and prevented CD28GR from driving transcription of a reporter gene. Interestingly, GR1 potently inhibited CD28, but not the expression of other Sp1- and EGR-1-regulated genes, an effect associated with lower Sp1 protein binding affinity of GR1 and GR2 compared with that of canonical Sp1 sites. These data show that DNA sequences that contain the G-rich sequence motif, G4N4G4, such as GR1 and GR2, can functionally mimic the regulatory protein binding ability of CD28GR. Thus, GR1 and GR2 act as molecular decoys to selectively interfere with transcriptional regulation of the CD28 gene. PMID: 10510368 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 87: Anticancer Drug Des. 1999 Jun;14(3):179-86. Effect of ecteinascidin-743 on the interaction between DNA binding proteins and DNA. Bonfanti M, La Valle E, Fernandez Sousa Faro JM, Faircloth G, Caretti G, Mantovani R, D'Incalci M. Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy. Ecteinascidin-743 (ET-743) is a tetrahydroisoquinoline alkaloid isolated from Ecteinascidia turbinata, a tunicate growing in mangrove roots in Caribbean. It has been shown to bind in the minor groove of DNA forming covalent adducts by reaction of the N2 of guanine with the carbinolamine moiety. We investigated ET-743 ability to inhibit the binding of different transcription factors to their consensus sequences by using gel shift assays. We have selected three types of factors: (i) oncogene products such as MYC, c-MYB and Maf; (ii) transcriptional activators regulated during the cell cycle as E2F and SRF; and (iii) general transcription factors such as TATA binding protein (TBP), Sp1 and NF-Y. We observed no inhibition of the binding of Sp1, Maf, MYB and MYC. Inhibition of DNA binding was observed for TBP, E2F, SRF at ET-743 concentrations ranging from 50 to 300 microM. The inhibition of binding of NF-Y occurs at even lower concentrations (i.e. 10-30 microM) when the recombinant subunits of NF-Y are preincubated with the drug, indicating that the inhibition of NF-Y binding does not require previous ET-743 DNA binding. Since NF-Y is a trimer containing two subunits with high resemblance to histones H2B and H2A, we have investigated the effect of ET-743 on nucleosome reconstitution. ET-743 caused a decrease of the nucleosomal band at 100 nM, with the complete disappearance of the band at 3-10 microM. These data suggest that the mode of action of this novel anticancer drug is related to its ability to modify the interaction between some DNA binding proteins and DNA. PMID: 10500494 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 88: J Biol Chem. 1999 Oct 1;274(40):28794-802. Expression of MXI1, a Myc antagonist, is regulated by Sp1 and AP2. Benson LQ, Coon MR, Krueger LM, Han GC, Sarnaik AA, Wechsler DS. Division of Pediatric Hematology, Department of Pediatrics, University of Michigan School of Medicine, Ann Arbor, Michigan 48109, USA. MXI1, a member of the MAD family of Myc antagonists, encodes a transcription factor whose expression must be tightly regulated to maintain normal cell growth and differentiation. To more closely investigate the transcriptional regulation of the human MXI1 gene, we have cloned and characterized the MXI1 promoter. After clarification of the 5'- and 3'-untranslated regions of the cDNA (indicating that the true length of the MXI1 transcript is 2643 base pairs), we identified two transcription initiation sites. We subsequently isolated the MXI1 promoter, which is GC-rich and lacks a TATA box. Although it contains at least six potential initiator sequences, functional studies indicate the proximal two initiator sequences in combination with nearby Sp1 and MED-1 sites together account for virtually all promoter activity. We also demonstrate that MXI1 promoter activity is repressed by high levels of AP2. These studies provide further insight into the complex regulatory mechanisms governing MXI1 gene expression and its role in cellular differentiation and tumor suppression. PMID: 10497252 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 89: Genes Cells. 1999 May;4(5):277-89. The G10BP-1 gene encoding a GC box binding protein, is a target of Myc and Jun/Fos. Shirasuna K, Takeuchi A, Bando T, Nakajima T, Oda K. Department of Biological Science and Technology, Science University of Tokyo, 2641 Yamazaki, Noda 278, Japan. BACKGROUND: G10BP, a serum-inducible factor, represses the transcription of the fibronectin gene through binding to the G-rich sequences in the promoter excluding Sp1 from binding to these sequences. RESULTS: The 5' flanking sequence of the G10BP-1 gene was isolated by polymerase chain reaction of the adaptor-ligated genomic DNA library using the adaptor primer and the G10BP-1 cDNA primer. The elements required for activation of the G10BP-1 promoter following serum stimulation were analysed by transfection of quiescent rat 3Y1 cells with G10BP-1 promoter-luciferase cDNA constructs containing 5' sequential deletions or base substitutions. The results showed that the promoter was activated by Myc and Jun through the E box and AP1 sites. The formation of DNA-protein complexes with 32P-labelled oligonucleotides containing the E box or AP1 site with cell extracts prepared during G1 progression was correlated with the promoter activation and greatly reduced by immunodepletion of Myc or c-Jun from the extracts. CONCLUSION: These results indicate that the G10BP-1 gene is a target of Myc and Jun/Fos and that these factors repress the fibronectin gene expression through induction of G10BP-1 during G1-to-S phase progression. PMID: 10421838 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 90: Gene. 1999 Jul 8;234(2):337-44. Characterization of the murine gene encoding 1-Cys peroxiredoxin and identification of highly homologous genes. Lee TH, Yu SL, Kim SU, Kim YM, Choi I, Kang SW, Rhee SG, Yu DY. Korea Research Institute of Bioscience and Biotechnology, PO Box 115, Yusong, Taejon 305-600, South Korea. A new type of peroxiredoxin, named 1-Cys peroxiredoxin (1-Cys Prx), reduces hydrogen peroxide with the use of electrons from unidentified electron donor(s). We have isolated the mouse gene encoding 1-Cys Prx (CP-3) and shown that it is comprised of five exons and four introns. Analysis of 5' flanking regions revealed binding sequences of several putative transcription factors such as Sp1, Pit-1a, c-Jun, c-Myc and YY1. It is noticeable that several potential Sp1 binding sites assigned the -60 through -96bp from putative transcription initiation site. The gel shift assays showed that Sp1 and Pit-1a bind specifically to each binding site in 1-Cys Prx promoter. We also isolated two highly related genes such as CP-2 and CP-5. These genes are encoded by single exons, and show 85% of nucleotide sequence homology with the CP-3. The structural features of these genes suggest that they might be intronless genes derived from the CP-3 by the mechanism involving retrotransposition. In addition, our data suggest that they are inserted to a specific site of the mouse L1 repetitive element. The 1-Cys Prx was actively transcribed in a variety of adult tissues as well as in the developing embryos. These results suggest that only the 1-Cys Prx gene might be relevant for studying the function of the 1-Cys Prx in the murine system. PMID: 10395907 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 91: Gene. 1999 May 17;232(1):97-106. Genomic organization and promoter characterization of the gene encoding the human telomerase reverse transcriptase (hTERT). Wick M, Zubov D, Hagen G. Bayer AG, Central Research Division, Department of Molecular Biology, D-51368, Leverkusen, Germany. maresa.wick.mw@bayer.ag.de The enzyme telomerase plays a crucial role in cellular proliferation and tumorigenesis. By adding hexameric repeats to chromosome ends, it prevents telomeric loss and, thus, entry into senescence. Recent data suggest that expression of the human telomerase reverse transcriptase subunit (hTERT) represents the limiting factor for telomerase activity. To gain an insight into the mechanisms regulating hTERT expression, we have determined the complete genomic organization of the hTERT gene and isolated the 5'- and 3'- flanking region. The hTERT gene encompasses more than 37kb and consists of 16 exons. We show that all hTERT insertion and deletion variants described so far most likely result from the usage of alternative splice consensus sequences in intron or exon regions. Furthermore, we identified a new hTERT splice variant. Analysis of the DNA sequence surrounding the putative transcriptional start region revealed a TATA-less promoter located in a CpG island. A promoter fragment spanning the first 1100bp upstream of the initiating ATG start codon exhibited high-level activity in HEK-293 cells. Several consensus binding sites for the transcription factor Sp1 as well as a c-Myc binding site were identified in this promoter region. Altogether, these results provide the basis for more detailed studies on the regulation of telomerase activity in normal and cancer cells, and may lead to the development of new cancer therapies. PMID: 10333526 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 92: Eur J Biochem. 1999 Feb;259(3):676-83. Structural organization and expression of the mouse gene for Pur-1, a highly conserved homolog of the human MAZ gene. Song J, Murakami H, Tsutsui H, Ugai H, Geltinger C, Murata T, Matsumura M, Itakura K, Kanazawa I, Sun K, Yokoyama KK. Tsukuba Life Science Center, Ibaraki, Japan. We have characterized the genomic structure and expression of the mouse gene for Pur-1. The cloned Pur-1 gene spans a 5-kb region encompassing the promoter, five exons, four introns and the 3'-untranslated region. All exon-intron junction sequences conform to the GT/AG rule. The promoter region has typical features of a housekeeping gene: a high G + C content (77.5%); a high frequency of CpG dinucleotides, in particular within the region 0.5 kb upstream of the site of initiation of translation; and the absence of canonical TATA and CAAT boxes. S1 nuclease protection assay demonstrated the presence of multiple sites for initiation of transcription around a site 108 nucleotides upstream of the ATG codon. Comparison of Pur-1 with the human gene for MAZ (Myc-associated zinc finger protein) revealed a striking homology of both their nucleotide and deduced protein sequences, an identical genomic organization and high similarity in promoter architecture and mRNA expression pattern. Sequence analysis of the 5'-flanking region of Pur-1 revealed numerous potential binding sites for transcription factors Sp1, AP-2 and Pur-1/MAZ itself. An element required for basal Pur-1 expression was mapped from nucleotide -258 to +43. This region also mediated stimulation of basal transcription by ectopically expressed MAZ protein. We conclude that the Pur-1 gene is the murine homolog of human MAZ and, like it, belongs to the family of housekeeping genes. PMID: 10092852 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 93: J Biol Chem. 1999 Mar 26;274(13):8698-707. Phenylethanolamine N-methyltransferase gene expression. Sp1 and MAZ potential for tissue-specific expression. Her S, Bell RA, Bloom AK, Siddall BJ, Wong DL. Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California 94305-5485, USA. Phenylethanolamine N-methyltransferase (PNMT) promoter-luciferase reporter gene constructs (pGL3RP863, pGL3RP444, and pGL3RP392) transfected into COS1, RS1, PC12, NIH/3T3, or Neuro2A cells showed the highest basal luciferase activity in the Neuro2A cells. DNase I footprinting with Neuro2A cell nuclear extract identified protected PNMT promoter regions spanning the -168/-165 and -48/-45 base pair Sp1/Egr-1 binding sites. Gel mobility shift assays and transient transfection assays using site-directed mutant PNMT promoter-luciferase reporter gene constructs indicated that the elevated basal luciferase activity in the Neuro2A cells was mediated by Sp-1. Furthermore, activation of the PNMT promoter by Sp1 depends on both its binding affinity for its cognate target sequences and its intracellular concentrations. When Sp1 levels were increased through an expression plasmid, luciferase reporter gene expression rose well beyond basal wild-type levels, even with either Sp1 binding element mutated. Finally, another transcription factor expressed in the Neuro2A cells competes with Sp1 by interacting with DNA sequences 3' to the -48 base pair Sp1 site to prevent Sp1 binding and induction of the PNMT promoter. The DNA consensus sequence, Southwestern analysis, and gel mobility shift assays with antibodies identify MAZ as the competitive factor. These findings suggest that Sp1 may potentially contribute to the tissue-specific expression of the PNMT gene, with the competition between Sp1 and MAZ conferring additional tissue-specific control. PMID: 10085109 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 94: Cancer Res. 1999 Feb 1;59(3):551-7. Cloning of human telomerase catalytic subunit (hTERT) gene promoter and identification of proximal core promoter sequences essential for transcriptional activation in immortalized and cancer cells. Takakura M, Kyo S, Kanaya T, Hirano H, Takeda J, Yutsudo M, Inoue M. Department of Obstetrics and Gynecology, Kanazawa University, School of Medicine, Ishikawa, Japan. Telomerase activation is thought to be a critical step in cellular immortalization and carcinogenesis. Of the three major subunits comprising human telomerase, human telomerase catalytic subunit (hTERT) has been shown to be a rate-limiting determinant of the enzymatic activity of human telomerase. However, little is known concerning how expression of hTERT is regulated in human cells. To identify the regulatory elements controlling hTERT gene expression, approximately 3.5 kb of the 5'-flanking sequence of hTERT was cloned and characterized. The promoter of hTERT was GC rich and lacked both TATA and CAAT boxes. The CapSite Hunting method identified transcription start site 19 bp upstream of the first nucleotide of the published cDNA sequence. Transient expression assays revealed that transcription of hTERT was significantly activated in cancer cell lines but repressed in normal primary cells. Using the fibroblast lineage at various stages of transformation, we found that transcription occurred in strains that had overcome replicative senescence and expressed telomerase activity. Deletion analysis of hTERT promoter identified the 181-bp core promoter region upstream of the transcription start site. Gel shift analysis revealed two major factors binding to core promoter, an E box (CACGTG) binding factor and Sp1. Overexpression of c-Myc resulted in a significant increase in transcriptional activity of the core promoter. These findings suggest that hTERT expression is strictly regulated at the transcription machinery, and that the proximal core promoter containing an E box and Sp1 sites is required for transactivation of hTERT. PMID: 9973199 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 95: J Exp Zool. 1998 Sep-Oct 1;282(1-2):188-95. Regulation of LDH-A gene expression by transcriptional and posttranscriptional signal transduction mechanisms. Jungmann RA, Huang D, Tian D. Cancer Center, Northwestern University Medical School, Chicago, Illinois 60611, USA. The lactate dehydrogenase-A (LDH-A) gene, whose product plays a pivotal role in normal anaerobic glycolysis and is frequently increased in human cancers, is highly regulated at the transcriptional and posttranscriptional levels. Our laboratory has carried out extensive studies concerning the regulation of LDH-A subunit expression. We have elucidated complex regulatory mechanisms by identifying multiple cis-acting promoter elements including functional sites for Sp1 and c-Myc interactions as well as sites that interact with the protein kinase A and protein kinase C substrates, CREB and AP1, respectively. Furthermore, we have reported the existence of a CRE-dependent silencer element in the LDH-A promoter. LDH-A expression is additionally regulated through the protein kinase A and C signal pathways at the posttranscriptional level, specifically mRNA stability. PMID: 9723176 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 96: J Cell Biochem. 1998 Sep 1;70(3):297-303. Magnetic field activation of protein-DNA binding. Lin H, Han L, Blank M, Head M, Goodman R. Department of Pathology, Columbia University Health Sciences, New York, New York 10032, USA. The mechanisms involved in sensing, signaling, and coordinating changes resulting from magnetic field-induced stress show substantial similarities to those of heat shock, e.g., magnetic field-induced heat shock 70 gene (HSP70) expression involves heat shock factor (HSF) activation and heat shock element binding. However, an additional requirement for transactivation of HSP70 expression by magnetic fields is the binding of Myc protein, indicating that additional elements and/or pathways are involved in the induction of HSP70 expression by magnetic fields. To investigate the possible participation of additional genetic elements in magnetic field-induced HSP70 expression, we examined both magnetic field exposure and heat shock on protein-DNA binding of the transcription factors HSF, AP-1, AP-2, and SP-1 in four human cell lines. The binding sites for these transcription factors are present in the HSP70 promoter. AP-1 binding activity, normally not increased by heat shock, was increased by magnetic fields; heat shock induced an increase only in HSF binding. Although intersecting and converging signaling pathways could account for the multiplicity of elements involved in magnetic field-induced HSP70 transcription, direct interaction of magnetic fields with DNA is also a possible mechanism. Because magnetic fields penetrate the cell, they could well react with conducting electrons present in the stacked bases of the DNA. PMID: 9706866 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 97: Biochem Biophys Res Commun. 1998 May 29;246(3):668-74. Genomic cloning and characterization of the mouse POZ/zinc-finger protein ZF5. Yokoro K, Yanagidani A, Obata T, Yamamoto S, Numoto M. Department of Dermatology, Hiroshima University School of Medicine, Japan. We isolated genomic DNA containing the entire sequence of ZF5, which was originally identified by its ability to repress the mouse c-myc promoter and which was characterized as one of the POZ (Poxvirus and zinc finger) proteins. The POZ motif is a protein-protein interaction interface found at the N-terminal region of zinc finger proteins. Sequence analysis demonstrated that the ATG translation initiation codon was separately located from the remainder of the coding sequence. Using both RNase protection and primer extension assay, a single major transcription start site was determined. Promoter analysis by transient transfection assay suggested positive autoregulation by ZF5 itself. The ZF5 N-terminal region, including the POZ domain, was required for this regulation. Sp1 also activated the ZF5 promoter and this activity was repressed by addition of ZF5. ZF5 expression was stronger in mouse ovary, lung and brain than in other organs. PMID: 9618270 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 98: J Biol Chem. 1998 May 29;273(22):13982-94. Structure of the human sarco/endoplasmic reticulum Ca2+-ATPase 3 gene. Promoter analysis and alternative splicing of the SERCA3 pre-mRNA. Dode L, De Greef C, Mountian I, Attard M, Town MM, Casteels R, Wuytack F. Laboratorium voor Fysiologie, Katholieke Universiteit Leuven, Campus Gasthuisberg, Herestraat 49, B-3000, Leuven, Belgium. Leo.Dode@med.kuleuven.ac.be Human chromosome 17-specific genomic clones extending over 90 kilobases (kb) of DNA and coding for sarco/endoplasmic reticulum Ca2+-ATPase 3 (SERCA3) were isolated. The presence of the D17S1828 genetic marker in the cosmid contig enabled us to map the SERCA3 gene (ATP2A3) 11 centimorgans from the top of the short arm p of chromosome 17, in the vicinity of the cystinosis gene locus. The SERCA3 gene contains 22 exons spread over 50 kb of genomic DNA. The exon/intron boundaries are well conserved between human SERCA3 and SERCA1 genes, except for the junction between exons 8 and 9 which is found in the SERCA1 gene but not in SERCA3 and SERCA2 genes. The transcription start site (+1) is located 152 nucleotides (nt) upstream of the AUG codon. The 5'-flanking region, including exon 1, is embedded in a 1.5-kb CpG island and is characterized by the absence of a TATA box and by the presence of 14 putative Sp1 sites, 11 CACCC boxes, 5 AP-2-binding motifs, 3 GGCTGGGG motifs, 3 CANNTG boxes, a GATA motif, as well as single sites for Ets-1, c-Myc, and TFIIIc. Functional promoter analysis indicated that the GC-rich region (87% G + C) from -135 to -31 is of critical importance in initiating SERCA3 gene transcription in Jurkat cells. Exon 21 (human, 101 base pairs; mouse, 86 base pairs) can be alternatively excluded, partially included, or totally included, thus generating, respectively, SERCA3a (human and mouse, 999 amino acids (aa)), SERCA3b (human, 1043 aa; mouse, 1038 aa), or SERCA3c (human, 1024 aa; mouse, 1021 aa) isoforms with different C termini. Expression of the mouse SERCA3 isoforms in COS-1 cells demonstrated their ability to function as active pumps, although with different apparent affinities for Ca2+. PMID: 9593748 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 99: Nucleic Acids Res. 1998 Mar 15;26(6):1449-57. Regulation of expression of nuclear and mitochondrial forms of human uracil-DNA glycosylase. Haug T, Skorpen F, Aas PA, Malm V, Skjelbred C, Krokan HE. UNIGEN Center for Molecular Biology, The Medical Faculty, Norwegian University of Science and Technology, N-7005 Trondheim, Norway. Promoters PA and PBin the UNG gene and alternative splicing are utilized to generate nuclear (UNG2) and mitochondrial (UNG1) forms of human uracil-DNA glycosylase. We have found the highest levels of UNG1 mRNA in skeletal muscle, heart and testis and the highest UNG2 mRNA levels in testis, placenta, colon, small intestine and thymus, all of which contain proliferating cells. In synchronized HaCaT cells mRNAs for both forms increased in late G1/early S phase, accompanied by a 4- to 5-fold increase in enzyme activity. A combination of mutational analysis and transient transfection demonstrated that an E2F-1/DP-1-Rb complex is a strong negative regulator of both promoters, whereas 'free' E2F-1/DP-1 is a weak positive regulator, although a consensus element for E2F binding is only present in PB. These results indicate a central role for an E2F-DP-1-Rb complex in cell cycle regulation of UNG proteins. Sp1 and c-Myc binding elements close to transcription start areas were positive regulators of both promoters, however, whereas overexpression in HeLa cells of Sp1 stimulated both promoters, c-Myc and c-Myc/Max overexpression had a suppressive effect. CCAAT elements were negative regulators of PB, but positive regulators of PA. These results demonstrate differential expression of mRNAs for UNG1 and UNG2 in human tissues. PMID: 9490791 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 100: Gene. 1998 Jan 12;206(2):237-45. Structural organization and chromosomal localization of the mouse tesk1 (testis-specific protein kinase 1) gene. Toshima J, Nakagawara K, Mori M, Noda T, Mizuno K. Department of Biology, Faculty of Science, Kyushu University, Hakozaki, Fukuoka 812-81, Japan. TESK1 (testis-specific protein kinase 1) is a protein serine-threonine kinase, containing characteristic structural features composed of an N-terminal kinase domain and a C-terminal proline-rich domain. Tesk1 mRNA is predominantly expressed in testicular germ cells, and developmental changes of expression in mouse testis suggest a role for this kinase in spermatogenesis. In the present study, we isolated and determined the overall sequence of the mouse Tesk1 gene, which spans 6.1 kilobases (kb) and contains 10 exons and 9 introns. The protein kinase domain is located in exons 1-9, while the proline-rich domain is in exons 9 and 10. The deduced 627 amino acid sequence of mouse TESK1 shows 97% and 94% identity with the rat and human TESK1, respectively. Sequence of the 5'-flanking and -untranslated region is devoid of a TATA box, but does contain several potential binding sites for transcription factors, including Sp1, AP-1, c-Myc, SRY and CREM (cyclic AMP-responsive element modulator). As CREM is implicated in the activation of several male germ cell-specific genes, it is suggested that the expression of the Tesk1 gene is under the control of CREM transcription activity. The Tesk1 gene was mapped to mouse chromosome 4A5-C1 by fluorescence in situ hybridization. PMID: 9469938 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 101: Mutat Res. 1997 Dec;385(3):159-72. Comparison of the promoters of the mouse (APEX) and human (APE) apurinic endonuclease genes. Harrison L, Ascione AG, Takiguchi Y, Wilson DM 3rd, Chen DJ, Demple B. Department of Cancer Cell Biology, Harvard School of Public Health, Boston, MA 02115, USA. We investigated the minimal promoter of APEX, which encodes mouse apurinic DNA repair endonuclease. A 1.85-kb fragment with APEX upstream sequences and approximately 290 bp of the transcribed region linked to a chloramphenicol acetyltransferase (CAT) reporter gene was assayed by transient transfection in NIH-3T3 cells. The minimal APEX promoter was comprised of approximately 190 bp of upstream and approximately 170 bp of transcribed DNA (exon 1 and most of intron 1). This approximately 360-bp region contains two CCAAT boxes and other consensus protein binding sites, but no TATA box. Deletion of the 5'-most CCAAT box decreased activity approximately 5-fold. The second CCAAT box (situated in exon 1) may play an independent role in APEX expression. Transcription start sites have been identified downstream of the second CCAAT box, and DNase I footprinting demonstrated NIH-3T3 nuclear proteins binding this region, including an Spl site located between the CCAAT boxes. Electrophoretic mobility-shift assays indicated binding by purified Sp1. Mouse proteins did not bind three myc-like (USF) sites in the APEX promoter, in contrast to the APE promoter. The APEX and APE promoter had similar activity in Hela cells, but in mouse cells, the murine promoter had approximately 5-fold higher activity than did the human promoter. Both the APEX and APE promoters exhibited bidirectional activity in their cognate cells. PMID: 9506886 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 102: Eur J Biochem. 1998 Jan 15;251(1-2):435-42. Modulation of DNA-protein interactions in the P1 and P2 c-myc promoters by two intercalating drugs. Vaquero A, Portugal J. Departamento de Biologia Molecular y Celular, Centro de Investigacion y Desarrollo, CSIC, Barcelona, Spain. Regulation of transcription from the oncogene c-myc has an important role in the genesis of various tumors. Therefore, c-myc is a potential target for chemotherapy by drugs which are able to modify its activity directly. In this article, we identify the binding sites in the P1 and P2 promoter regions of c-myc for the intercalating antibiotics actinomycin D and elsamicin A. Gel retardation experiments indicate that actinomycin D or elsamicin A binding can inhibit the formation of several DNA-protein complexes. However, relatively low concentrations of elsamicin A, but not actinomycin D, appear to increase the level of binding to the P1 promoter of a protein factor. Using pure Sp1 transcription factor and an oligonucleotide containing the Sp1 putative binding site, we determined that the binding enhancement induced by small amounts of elsamicin was on the Sp1-DNA complex. Run-off transcription experiments in vitro showed that the effect of elsamicin A on Sp1 binding is followed by the maintenance or a relative rise in transcription levels from the P1 promoter of c-myc, while actinomycin D always inhibited the transcription from the P1 c-myc promoter in a concentration-dependent manner. Higher concentrations of elsamicin acted as an inhibitor of the transcription from the P1 start site but not from the P2. PMID: 9492315 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 103: Biochem Biophys Res Commun. 1997 Dec 18;241(2):258-63. Identification of regulatory elements of human alpha 6 integrin subunit gene. Nishida K, Kitazawa R, Mizuno K, Maeda S, Kitazawa S. Second Department of Pathology, Kobe University School of Medicine, Japan. The integrin alpha 6 subunit associates with either the beta 1 or beta 4 subunit to form receptors for laminin, a major component of the basement membrane. Here, we characterized basal promoter of the human integrin alpha 6 subunit gene. The transcription start site, mapped by primer extension analysis, was 208 bp upstream of the translation start site. The promoter region lacked canonical TATA and GC boxes, but did contain a TATA-like sequence (GATAAA) 23 bp upstream of the major transcription start site. Consensus binding sites for Sp1 and the NF-kappa B complex were also present in the promoter region. A putative glucocorticoid/progesterone receptor responsive element (GRE/PRE), together with the Ap1 and c-myc binding sites located around 350-360 bp upstream of the transcription start site, represented positive regulatory sequences. Our current study showed a molecular model by which progesterone promotes tumor cell invasion through the basement membrane by up-regulating the laminin receptor. PMID: 9425259 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 104: J Virol. 1997 Dec;71(12):9600-7. Activation of the adenovirus major late promoter by transcription factors MAZ and Sp1. Parks CL, Shenk T. Department of Molecular Biology, Howard Hughes Medical Institute, Princeton University, New Jersey 08544-1014, USA. Multiple binding sites for the transcription factors MAZ and Sp1 within the adenovirus type 5 major late promoter have been identified by DNase I protection studies. In the proximal region of the promoter, both MAZ and Sp1 interact with GC-rich sequences flanking the TATA box. Two MAZ binding sites are centered at -18 and -36 relative to the transcriptional initiation site. Sp1 bound only to the -18 GC-rich sequence. Several sites of interaction were also evident in the distal region of the promoter. Both MAZ and Sp1 interacted with a sequence centered at -166, and MAZ bound weakly to an additional site centered at -130. Overexpression of MAZ or Sp1 activated expression from the major late promoter in transient expression assays. Mutational analysis of the GC-rich sequences in the major late promoter suggested that a primary target of MAZ activation is the GC-rich sequences flanking the TATA sequence, whereas Sp1 requires the distal GC-rich sequence elements to stimulate gene expression. This activation is enhanced by the adenovirus E1A protein, and evidence for interaction between E1A and both transcription factors was obtained by using an immunoprecipitation assay. Activation by MAZ and Sp1 also was observed in transfection studies using the complete adenovirus type 5 genome as the target. Increased levels of late mRNA from both the L1 and L5 regions were observed when MAZ or Sp1 expression plasmids were transfected with viral DNA. Unexpectedly, activation of the major late promoter by MAZ and Sp1 was detected irrespective of whether the viral DNA could replicate. PMID: 9371624 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 105: Hum Mol Genet. 1997 Nov;6(12):2051-60. Structural and functional characterization of the human FMR1 promoter reveals similarities with the hnRNP-A2 promoter region. Drouin R, Angers M, Dallaire N, Rose TM, Khandjian EW, Rousseau F. Unite de Recherche en Genetique Humaine et Moleculaire,Centre de Recherche, Pavillon Saint-Francois d'Assise, Centre Hospitalier Universitaire de Quebec, Canada. Fragile X mental retardation syndrome is associated with an expansion of a CGG repeat within the 5'UTR of the first exon of the FMR1 gene, abnormal methylation of the CpG island in the promoter region, and a transcriptional silencing of this gene. We studied transcriptional regulation of the FMR1 gene using protein footprint analysis of the active and inactive gene in vivo . We identified four footprints within the FMR1 promoter region which correspond to consensus binding sites of known transcription factors, alpha-PAL/NRF1, Sp1, H4TF1/Sp1-like and c-myc. These footprints were present in normal cells with a transcriptionally active FMR1 gene. The same footprints were present in different cell types: primary fibroblasts, lymphoblastoid cells and peripheral lymphocytes. However, for the 1.1 kb region analyzed, no footprints were detected in a variety of cell types derived from patients with fragile X syndrome which have a transcriptionally inactive FMR1 gene. A BLAST nucleotide search identified sequence similarities between the region of the FMR1 gene containing the footprints and an analogous region within the promoter region of the gene for the heterogeneous nuclear ribonucleoprotein (hnRNP) A2, a member of a family of ribonucleoproteins implicated in mRNA processing and nuclear-cytoplasm transport. The nucleotide sequences identified in the hnRNP-A2 promoter region correspond to the same consensus binding sites showing DNA-protein interactions in the FMR1 gene. Our previous functional studies and the studies of others demonstrate that FMR proteins, like hnRNP-A2, are also ribonucleoproteins which appear to be involved in mRNA transport. The results from our footprint studies suggest that the expression of the FMR1 gene is regulated by the binding of specific transcription factors to sequence elements in the 5' region of the gene and that this expression may be regulated by elements in common with the hnRNP-A2 gene. Common regulation of these two genes might play an important role in the cooperative processing and transport of mRNA from the nucleus to the translation machinery. PMID: 9328468 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 106: Mol Cells. 1997 Aug 31;7(4):537-43. Characterization of the murine cyclin D2 gene: exon/intron organization and promoter activity. Jun DY, Kim MK, Kim IG, Kim YH. Department of Microbiology, College of Natural Sciences, Kyungpook National University, Taegu, Korea. Cyclin D2 is normally expressed in G1 and promotes progression through G1 of the cell cycle. From a murine genomic library constructed with spleen DNA, two overlapping genomic clones of cyclin D2 were isolated. These clones contain most of the exon of cyclin D2 except exon 5. Characterization of these clones revealed that murine cyclin D2 mRNA spans over 18 kb and 5 exons ranging from 149 to approximately 462 bp in length, and suggested that exon 5 may be at least >5 kb downstream from exon 4. Primer extension analysis of cyclin D2 mRNA isolated from murine activated T cells detected 5 putative sites of transcription initiation. These are located at - 499, - 417, - 391, - 373, and - 349 relative to the translation start site, which is given as + 1. No consensus sequence for TATA box existed at an appropriate position within the promotor region. Instead, several putative transcriptional factor binding sites for C/EBP, PEA3, AP2, NF-Y, Sp1, c-Myc, GATA-1, AP1, v-Myb, and CREB were detected. The 5'-flanking region of the cyclin D2 gene up to nucleotide - 945 shared about 61% sequence homology between mouse and human. Functional analysis of promoter activity of the 5'-flanking region of cyclin D2 suggested that the region - 1,100 to - 805 including C/EBP, PEA3, AP2, NF-Y, c-Myc, and Sp1 may have a major positive regulatory activity for expression of cyclin D2. PMID: 9339900 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 107: Eur J Biochem. 1997 Aug 1;247(3):1166-73. Phosphorylation of human general transcription factors TATA-binding protein and transcription factor IIB by DNA-dependent protein kinase--synergistic stimulation of RNA polymerase II basal transcription in vitro. Chibazakura T, Watanabe F, Kitajima S, Tsukada K, Yasukochi Y, Teraoka H. Department of Molecular Genetics, Medical Research Institute, Tokyo Medical and Dental University, Japan. DNA-dependent protein kinase (DNA-PK) has been known to catalyze phosphorylation of a number of regulatory factors involved in DNA replication and transcription such as simian virus 40 T antigen, p53, c-Myc, Sp1, and RNA polymerase II (Pol II). We examined the possibility that DNA-PK phosphorylates the general transcription factors TATA-binding protein (TBP) and transcription factor (TF) IIB, which play key roles in the formation of transcription initiation complex with Pol II. By using a highly purified preparation of DNA-PK from Raji cells, both TBP and TFIIB were shown to be phosphorylated in vitro by DNA-PK. We then investigated the effect of the phosphorylation of these factors on Pol II basal transcription. Stepwise analysis of preinitiation complex formation by electrophoretic mobility shift assay revealed that the phosphorylation of TBP and TFIIB by DNA-PK did not affect the formation of promoter (P)-TBP and P-TBP-TFIIB complexes but synergistically stimulated the formation of P-TBP-TFIIB-TFIIF-Pol II complex. Similarly, combination of the phosphorylated TBP and TFIIB synergistically stimulated Pol II basal transcription from adenovirus major late promoter. These observations suggest that DNA-PK could positively regulate the Pol II basal transcription by phosphorylating TBP and TFIIB. PMID: 9288944 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 108: J Cell Biochem. 1997 Mar 15;64(4):651-60. Aberrant expression and regulation of hepatic epidermal growth factor receptor in a c-myc transgenic mouse model. Woitach JT, Conner EA, Wirth PJ, Thorgeirsson SS. Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-4255, USA. In an attempt to elucidate the mechanism by which c-myc and transforming growth factor-alpha (TGF-alpha) cooperate in hepatocyte tumor development, we have analyzed signaling by the epidermal growth factor (EGF) receptor and the consequent regulation of receptor number in transgenic mice bearing the c-myc transgene under the control of the albumin enhancer/promoter. 125I-EGF binding and Scatchard analysis indicated a single class of high affinity receptors with the total number of binding sites of 1.2 X 10(4) +/- 600 and 2.5 X 10(5) +/- 1000 sites/cell in the normal and c-myc hepatocytes in primary culture, respectively. After 72 h of EGF exposure in culture, the number of detectable EGF receptors on the cell surface of the c-myc hepatocytes was not reduced, whereas the number of EGF receptors on normal hepatocytes was reduced to 32% that of untreated hepatocytes. Nuclear run-on experiments done with nuclei isolated from intact livers demonstrated that transcription of the EGF receptor was 4.9-fold higher in c-myc mice. Increased levels of the transcriptional factor SP1 in the c-myc hepatocytes in vivo and in primary culture, suggest a mechanism for the increased transcription of the EGF receptor. c-myc also increases the expression of TGF-alpha; a consequent increase in tyrosine phosphorylation is also detected in vivo. Thus, the increased number of EGF receptors in c-myc expressing hepatocytes, even after prolonged exposure to EGF, or TGF-alpha in vivo, may allow greater triggering of the EGF receptor signaling cascade. PMID: 9093914 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 109: J Cell Biochem. 1997 Mar 15;64(4):565-72. Inhibition of human immunodeficiency virus type 1 replication by a cellular transcriptional factor MBP-1. Ray RB, Srinivas RV. Department of Internal Medicine, St. Louis University, Missouri 63110-0250, USA. Rayrb@sluvca.slu.edu A cellular transcriptional factor initially identified as the c-myc promoter binding protein (MBP-1) was subsequently characterized as a cell regulatory protein with multifunctional activities. In this study, the role of MBP-1 on human immunodeficiency virus type-1 (HIV-1) transcriptional activity was investigated. MBP-1 showed inhibition of HIV-1 long terminal repeat (LTR)-directed chloramphenicol acetyl transferase (CAT) activity in a transient cotransfection assay. Deletion of upstream elements of the HIV-1 LTR, including the nuclear factor kappa B (NF-kappa B) and Sp1 binding sites, did not affect the MBP-1 mediated suppression of HIV-1 LTR. The core promoter of the HIV-1 appeared to be the primary sequence involved in MBP-1 mediated inhibition. In the presence of HIV-1 TAR sequence and Tat protein, MBP-1 did not inhibit the viral promoter activity. In addition, cotransfection experiments with HIV-1 LTR and deletion mutants of MBP-1 suggested that the carboxyl terminal half of MBP-1 suppresses the HIV-1 promoter activity. Exogenous expression of MBP-1 showed suppression of HIV-1 replication in acutely infected cells and in cells cotransfected with a molecular clone of HIV-1. These results suggest that exogenous expression of MBP-1 plays an important role in the regulation of HIV-1 replication in infected cells. PMID: 9093905 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 110: Mol Cell Biol. 1997 Mar;17(3):1037-48. A minimal regulatory region maintains constitutive expression of the max gene. Peters MA, Sollenberger KG, Kao TL, Taparowsky EJ. Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907-1392, USA. Max is a basic helix-loop-helix/leucine zipper protein that forms heterodimers with the Myc family of proteins to promote cell growth and with the Mad/Mxi1 family of proteins to inhibit cell growth. The role of Max as the obligate binding partner for these two protein families necessitates the observed constitutive expression and relatively long half-life of the max mRNA under a variety of growth conditions. In this study, we have used the chicken max gene to map DNA elements maintaining max gene expression in vertebrate cells. We have identified a minimal regulatory region (MRR) that resides within 115 bp of the max translation initiation site and that possesses an overall structure typical of TATA-less promoters. Within the MRR are two consensus binding sites for Sp1, a ubiquitously expressed transcription factor that plays a role in the expression of many constitutive genes. Interestingly, we show that direct binding by Sp1 to these sites is not required for MRR-mediated transcription. Instead, the integrity of a 20-bp DNA element in the MRR is required for transcriptional activity, as is the interaction of this DNA element with a 90-kDa cellular protein. Our data suggest that it is the persistence of this 90-kDa protein in vertebrate cells which drives max gene expression, insulates the max promoter from the dramatic changes in transcription that accompany cell growth and development, and ensures that adequate levels of Max will be available to facilitate the function of the Myc, Mad, and Mxi1 families of proteins. PMID: 9032230 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 111: J Biol Chem. 1997 Feb 14;272(7):4021-6. Sp3 is a bifunctional transcription regulator with modular independent activation and repression domains. Majello B, De Luca P, Lania L. Department of Genetics, Molecular and General Biology, University of Naples "Federico II," via Mezzocannone 8, 80134 Naples 10, Italy. Sp3 is a member of the Sp family of transcription factors and binds to DNA with affinity and specificity comparable to that of Sp1. We demonstrate that Sp3 is a bifunctional transcription factor that can both activate and repress transcription. Gene fusion experiments in mammalian cells demonstrate that the Sp3 activation potential is distributed over an extensive glutamine-rich N-terminal region, whereas the repressor activity has been mapped in a 72-amino acid region located at the 5' of the zinc finger DNA-binding domain. We demonstrated that the repression activity is strictly dependent on the context of the DNA-binding sites bound by Sp3. We found that Sp3 represses transcription of promoters bearing multiple GAL4 DNA-binding sites, whereas it activates isogenic reporters containing a single GAL4-binding site. Transfection experiments in Drosophila cells that lack endogenous Sp activity demonstrated that Sp3 does not possess an active repression domain that can function in insect cells, rather it is a weak transcriptional activator of the c-myc promoter. Our results strongly suggest that Sp3 is a dual-function regulator whose activity is dependent upon both the promoter and the cellular context. PMID: 9020109 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 112: Nucleic Acids Res. 1996 Oct 1;24(19):3846-57. Cloning and characterization of the genomic DNA of the human MSSP genes. Haigermoser C, Fujimoto M, Iguchi-Ariga SM, Ariga H. Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan. MSSP proteins have been identified by their binding to an upstream element of c-myc. Independently, two different approaches yielded two cDNA clones highly homologous to the MSSP cDNAs, suggesting an involvement of MSSP in the regulation of the cell cycle (scr2) and in the repression of HIV-1 and ILR2 alpha-promoter transcription (human YC1). Screening human genomic libraries, we have isolated clones belonging to two different gene loci. Whereas the human MSSP gene 1 turned out to be intronless, the organization of the coding sequence within gene 2 is more complex. It spans more than 60 kb and contains 16 exons (including two alternative first exons), ranging from 48 to 287 bp, respectively. The intron sizes vary from 0.1 to more than 13 kb. Gene 1 has been completely sequenced. A deletion series of its upstream region was conjugated to the luciferase gene, but the transfection of the constructs did not display any promoter activity. Moreover, compared with gene 2 and the cDNA sequences known so far, about 20 point mutations as well as flanking direct repeats have been detected in the MSSP gene 1, showing that it possesses all the characteristics of processed retropseudogenes. Sequence analysis of a 1.7 kb fragment of the 5' flanking region of the MSSP gene 2 revealed that the promoter of gene 2 lacks consensus sequences for TATA and CCAAT boxes, is GC-rich, and contains numerous potential transcription factor binding elements including an Sp1 binding site. DNase I footprinting experiments showed that the putative Sp1 site was bound by proteins. The results of primer extension and S1 mapping analyses suggested the transcription of the gene starts at multiple positions upstream from the initiator methionine codon. Luciferase assays employing progressive deletions of the 1.7 kb promoter region allowed us to define the minimal promoter region of 428 bp (-488/+) and revealed a complex pattern of the transcriptional regulation the human MSSP gene 2. Furthermore, it can be concluded that the MSSP gene 2 encodes both MSSP-1 and MSSP-2, and moreover scr2 and human YC1. PMID: 8871567 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 113: DNA Cell Biol. 1996 Jul;15(7):581-8. Isolation and analysis of inducibility of the rat N-methylpurine-DNA glycosylase promoter. Grombacher T, Kaina B. Research Center Karlsruhe, Institute of Genetics, Germany. Alkylations at base nitrogens in DNA are removed by excision repair, the first step of which is catalyzed by the repair enzyme N-methylpurine-DNA glycosylase (MPG). To study regulation of MPG expression, we have cloned the rat MPG promoter. A cosmid clone containing the rat MPG gene was isolated from a library using rat MPG cDNA as a probe. The 5' part of the MPG gene and the nontranscribed 5'-flanking region were isolated and characterized. Transcription start sites of the rat MPG gene were identified by primer extension and S1 nuclease protection analysis of RNA from primary rat hepatocytes. Promoter activity of the 5'-flanking noncoding region was shown by transfection in H4IIE rat hepatoma cells of various genomic MPG fragments cloned in front of the reporter gene chloramphenicol acetyltransferase. The rat MPG promoter does not contain a TATA box, but has a CCAAT sequence element and putative binding sites for the transcription factors Sp1, AP-2, AP-3, Ets-1, PEA3, NF-1, p53, c-Myc, NF-kappa B, and the glucocorticoid receptor. The activity of the rat MPG promoter was found to be inducible by the tumor promoter TPA and UV light, but not to a significant extent by methylating agents and ionizing radiation. PMID: 8756339 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 114: DNA Cell Biol. 1996 Jul;15(7):543-8. Novel DNase I hypersensitive sites in the 3'-flanking region of the human c-myc gene. Murphy LC, Huzel N, Davie JR. Department of Biochemistry and Molecular Biology, University of Manitoba, Winnipeg, Canada. DNase I hypersensitivity regions correlate with genetic regulatory loci and binding sites for sequence-specific DNA-binding proteins. We present data supporting the presence of novel DNase 1 hypersensitive sites (which we have designated sites VI-IX) in both the body of the human c-myc gene downstream from exon 2 and the 3'-flanking region of the c-myc gene in HL-60 cells. All of these novel DH sites are markedly decreased when HL-60 cells are treated with either dimethyl sulfoxide (DMSO) or retinoic acid. Moreover, a similar pattern of DNase I hypersensitive sites in this region of c-myc was present in MCF-7 human breast cancer cells growing in culture. Our results suggest a potential role for these sites in transcriptional regulation of the human c-myc gene. PMID: 8756335 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 115: Mol Cell Biol. 1996 May;16(5):2350-60. Heterogeneous nuclear ribonucleoprotein K is a transcription factor. Michelotti EF, Michelotti GA, Aronsohn AI, Levens D. Laboratory of Pathology, National Cancer Institute, National Institute of Health, Bethesda, Maryland 20892, USA. The CT element is a positively acting homopyrimidine tract upstream of the c-myc gene to which the well-characterized transcription factor Spl and heterogeneous nuclear ribonucleoprotein (hnRNP) K, a less well-characterized protein associated with hnRNP complexes, have previously been shown to bind. The present work demonstrates that both of these molecules contribute to CT element-activated transcription in vitro. The pyrimidine-rich strand of the CT element both bound to hnRNP K and competitively inhibited transcription in vitro, suggesting a role for hnRNP K in activating transcription through this single-stranded sequence. Direct addition of recombinant hnRNP K to reaction mixtures programmed with templates bearing single-stranded CT elements increased specific RNA synthesis. If hnRNP K is a transcription factor, then interactions with the RNA polymerase II transcription apparatus are predicted. Affinity columns charged with recombinant hnRNP K specifically bind a component(s) necessary for transcription activation. The depleted factors were biochemically complemented by a crude TFIID phosphocellulose fraction, indicating that hnRNP K might interact with the TATA-binding protein (TBP)-TBP-associated factor complex. Coimmunoprecipitation of a complex formed in vivo between hnRNP K and epitope-tagged TBP as well as binding in vitro between recombinant proteins demonstrated a protein-protein interaction between TBP and hnRNP K. Furthermore, when the two proteins were overexpressed in vivo, transcription from a CT element-dependent reporter was synergistically activated. These data indicate that hnRNP K binds to a specific cis element, interacts with the RNA polymerase II transcription machinery, and stimulates transcription and thus has all of the properties of a transcription factor. PMID: 8628302 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 116: J Cell Biochem. 1996 Mar 15;60(4):560-71. Analysis of human breast cancer nuclear proteins binding to the promoter elements of the c-myc gene. Miller TL, Jin Y, Sun JM, Coutts AS, Murphy LC, Davie JR. Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Manitoba, Winnipeg, Canada. The expression of the c-myc gene is essential for the proliferation of both hormone-dependent and -independent human breast cancer cells. The regulation of c-myc gene expression in MCF-7 (hormone-dependent, estrogen-receptor (ER)-positive) and MDA MB 231 (hormone-independent, ER-negative) human breast cancer cells differs, with the c-myc gene of MCF-7 but not MDA MB 231 cells being regulated at the transcriptional level by estrogen. We have shown previously that the DNAase I hypersensitive (DH) sites in the c-myc chromatin of hormone-dependent and -independent human breast cancer cells were similar, with the exception of DH site II2. DH site II2, which maps near the P0 promoter, was less sensitive in hormone-dependent than in hormone-independent cells. As DH sites generally indicate the presence of sequence-specific DNA-binding proteins, we undertook a study to identify the nuclear proteins isolated from MCF-7 and MDA MB 231 cells that bound to the P0 and P2 promoter regions of the c-myc gene in vitro. The studies presented here provide evidence that Sp1 and/or Sp1-like proteins bind to the P0 and P2 promoter regions of the c-myc gene of MCF-7 and MDA MB 231 cells. Furthermore, evidence is presented for the presence of several previously unidentified sequence-specific DNA-binding proteins binding to these promoters. The DNA-binding activities of these latter proteins differed in the nuclear extracts of the MCF-7 and MDA MB 231 human breast cancer cells. PMID: 8707895 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 117: J Biol Chem. 1996 Feb 23;271(8):4417-30. The serotonin 1a receptor gene contains a TATA-less promoter that responds to MAZ and Sp1. Parks CL, Shenk T. Department of Molecular Biology, Howard Hughes Medical Institute, Princeton University, Princeton, New Jersey 08544-1014, USA. The structure and function of the 5'-flanking region of the mouse and human serotonin 1a receptor gene have been analyzed by RNA 5' end mapping, DNA-protein interaction, and transient expression assays. A large number of mRNA 5' termini, detected by mapping 5' ends from mouse brain RNA, were found dispersed over a region of about 700 base pairs flanking the receptor coding sequence. Consistent with the apparently heterogeneous pattern of transcription initiation, the flanking DNA sequence lacked typical TATA box elements and was rich in guanine and cytosine. The mouse and human 5'-flanking sequences were 63% homologus and similarly organized. A guanine-cytosine-rich DNA sequence motif related to the sequence 5'-GGGG(C/A)GGGG-3' was repeated within the 5'-flanking region and located at or near several mRNA 5' ends. This DNA sequence motif bound to proteins in a crude HeLa cell nuclear extract. A cDNA encoding a protein that interacts with this sequence was cloned and found to be the MAZ (Pur-1, Zif87) protein. The interaction between MAZ and the receptor gene 5'-flanking region proximal to the protein coding sequence was examined by DNase I footprinting, and four sites of MAZ interaction were identified. Three of the four MAZ binding sites also were shown to interact with transcription factor Sp1. Overproduction of MAZ or Sp1 in transient transfection assays increased expression directed by the human 5'-flanking sequence, although MAZ was substantially more effective. This result suggests that MAZ and Sp1 both participate in regulating expression from the serotonin 1a receptor gene promoter, and it raises the possibility that MAZ may act at a variety of promoters through the guanosine-cytosine-rich sequences generally thought to serve as binding sites for the Sp1 family of transcription factors. Analysis of one of the guanosine-cytosine-rich DNA sequences also revealed that it can serve as a transcription initiator sequence in vitro. This initiator sequence differs from previously characterized initiators and may represent a new class of this transcriptional control sequence. PMID: 8626793 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 118: Mol Cell Biol. 1996 Feb;16(2):634-47. Protein-DNA interactions at the major and minor promoters of the divergently transcribed dhfr and rep3 genes during the Chinese hamster ovary cell cycle. Wells J, Held P, Illenye S, Heintz NH. Program in Cell and Molecular Biology, University of Vermont College of Medicine, Burlington 05405, USA. In mammals, two TATA-less bidirectional promoters regulate expression of the divergently transcribed dihydrofolate reductase (dhfr) and rep3 genes. In CHOC 400 cells, dhfr mRNA levels increase about fourfold during the G1-to-S phase transition of the cell cycle, whereas the levels of rep3 transcripts vary less than twofold during this time. To assess the role of DNA-binding proteins in transcriptional regulation of the dhfr and rep3 genes, the major and minor dhfr-rep3 promoter regions were analyzed by high-resolution genomic footprinting during the cell cycle. At the major dhfr promoter, prominent DNase I footprints over four upstream Sp1 binding sites did not vary throughout G1 and entry into the S phase. Genomic footprinting revealed that a protein is constitutively bound to the overlapping E2F sites throughout the G1-to-S phase transition, an interaction that is most evident on the transcribed template strand. On the nontranscribed strand, multiple changes in the DNase I cleavage pattern are observed during transit through G1 and entry into the S phase. By using gel mobility shift assays and a series of sequence-specific probes, two different species of E2F were shown to interact with the dhfr promoter during the cell cycle. The DNA binding activity of one E2F species, which preferentially recognizes the sequence TTTGGCGC, did not vary significantly during the cell cycle. The DNA binding activity of the second E2F species, which preferentially recognizes the sequence TTTCGCGC, increased during the G1-to-S phase transition. Together, these results indicate that Sp1 and the species of E2F that binds TTTGGCGC participate in the formation of a basal transcription complex, while the species of E2F that binds TTTCGCGC regulates dhfr gene expression during the G1-to-S phase transition. At the minor promoter, DNase I footprints at a consensus c-Myc binding site and three Sp1 binding sites showed little variation during the G1-to-S phase transition. In addition to protein binding at sequences known to be involved in the regulation of transcription, genomic footprinting of the entire promoter region also showed that a protein factor is constitutively bound to the first intron of the rep3 gene. PMID: 8552092 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 119: Gene Expr. 1996;6(2):113-27. TATA box and Sp1 sites mediate the activation of c-myc promoter P1 by immunoglobulin kappa enhancers. Geltinger C, Hortnagel K, Polack A. GSF-National Research Center for Environment and Health, Institute of Clinical Molecular Biology and Tumour Genetics, Munchen, Germany. In Burkitt's lymphoma (BL) cells the proto-oncogene c-myc is transcriptionally activated by chromosomal translocation to the immunoglobulin (Ig) gene loci. This activation is characterized by preferential transcription from the c-myc promoter P1 and accomplished by juxtaposed Ig enhancer elements. To identify promoter elements required for enhancer-activated P1 transcription, we studied the activation of c-myc reporter gene constructs by the Ig kappa intron and 3' enhancers. Deletion analysis defined the core promoter with a TATA box and two adjacent GC/GT boxes upstream sufficient for basal and enhancer-activated transcription. Gel retardation assays revealed Sp1's binding affinity to the GC/GT box proximal to the TATA box to be higher than to the distal one. This difference correlated well with the resulting levels of transcription mediated by Sp1 in contransfection experiments in BL and Sp1-deficient SL2 cells. Sp3 also bound to the core promoter in vitro, but failed to transactivate in vivo. Mutation of the distal Sp1 site moderately affected basal transcription concomitant with a modest decrease in enhancer stimulation. Mutation of the proximal Sp1 site almost entirely abolished basal as well as enhanced transcription. A considerable level of basal transcription was maintained upon mutation of the TATA box, whereas enhancer-activated transcription largely was abolished. Stable transfection of the BL cell line Raji with constructs containing core promoter mutations confirmed that the proximal Sp1 site and the TATA box are essential for the activation of promoter P1 by the Ig kappa enhancers. PMID: 8979089 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 120: Virology. 1995 Nov 10;213(2):624-38. Induction by herpes simplex virus of free and heteromeric forms of E2F transcription factor. Hilton MJ, Mounghane D, McLean T, Contractor NV, O'Neil J, Carpenter K, Bachenheimer SL. Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill 27599-7290, USA. We have determined that HSV causes rapid and large increases in cell-cycle-regulated free E2F and S-phase p107/E2F DNA binding activities in asynchronous cultures of C33A cells. Induction occurred by 4 hr postinfection and coincided with the appearance of viral encoded immediate-early and delayed-early proteins, i.e., when viral DNA replication normally commences. No increase in E2F activities occurred when cells were infected with viruses expressing mutant regulatory proteins ICP4 or ICP27, or mutant replication proteins ICP8, pol or helicase, or when cells were infected with wild-type virus in the presence of inhibitors of DNA synthesis. In contrast, ICP8 mutant-infected cells contained elevated amounts of NF kappa B activity equivalent to WT virus, no induction of Sp1 relative to WT virus, and reduced ATF/CREB activity relative to WT virus. Results of transient expression assays with E2F-responsive reporters indicated that the net effect of induction of both active (free E2F) and repressive (p107/E2F) complexes was a decrease in AdE2 promoter activity and an increase in c-myc promoter activity. Taken together these results suggest that HSV can cause unscheduled changes in the amount and functional status of a cell-cycle-regulated transcription factor. These results are discussed in light of possible roles for viral-induced alterations in E2F, especially as related to imposing or overriding cell-cycle checkpoints. PMID: 7491786 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 121: Mol Endocrinol. 1995 Nov;9(11):1477-87. Control of insulin-like growth factor-II/mannose 6-phosphate receptor gene transcription by proximal promoter elements. Liu Z, Mittanck DW, Kim S, Rotwein P. Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, Saint Louis, Missouri 63110, USA. The insulin-like growth factor-II/cation-independent mannose 6-phosphate receptor (IGF-II/MPR) is a multifunctional protein that binds IGF-II and ligands containing a mannose 6-phosphate recognition marker. Recent studies have shown that this receptor plays a critical role in mammalian development, and that its expression is controlled by both epigenetic and tissue-specific factors. Our laboratory has cloned the 93-kilobase mouse gene and characterized its 48 exons. In this report we describe the structure and function of the IGF-II/MPR gene promoter. To study promoter function, a series of chimeric plasmids linking different segments of IGF-II/MPR 5' flanking DNA to the reporter gene, firefly luciferase, were transiently transfected into HepG2 and C3H 10T1/2 cells. Promoter activity was orientation-specific and was maximal (550- to 4250-fold above promoterless control) with a plasmid containing 266 base pairs (bp) of IGF-II/MPR DNA. The fusion gene accurately directed transcription as measured by ribonuclease protection assay using RNA extracted from transfected cells. DNA-protein binding studies by in vitro DNase I footprinting revealed an extended 54-bp footprint within the proximal promoter that contained two E-boxes and potential binding sites for transcription factors Sp1, NGF-IA, and related proteins. Gel mobility shift experiments with double-stranded oligonucleotides containing this region gave rise to several specific DNA-protein complexes, and the addition of specific antibodies indicated that proteins antigenically related to Sp1 and c-Myc were components of one or more of these bands. Deletion of this 54-bp segment led to an 8-fold decline in promoter activity, and its transfer to a heterologous promoter stimulated gene expression by nearly 7-fold. Mutational analyses indicated that each E box contributed to more than half of the enhancer's activity. These results define a strong minimal IGF-II/MPR promoter of no more than 266 bp and identify a 54-bp enhancer within this promoter fragment. Our observations thus represent a first step toward characterizing the developmental, epigenetic, and tissue-specific factors that control IGF-II/MPR gene expression. PMID: 8584025 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 122: Genomics. 1995 Jul 20;28(2):261-72. Structure and chromosomal localization of the human gene of the phosphotyrosyl phosphatase activator (PTPA) of protein phosphatase 2A. Van Hoof C, Aly MS, Garcia A, Cayla X, Cassiman JJ, Merlevede W, Goris J. Afdeling Biochemie, Faculteit Geneeskunde, Katholieke Universiteit Leuven, Belgium. The PTPA gene encodes a specific phosphotyrosyl phosphatase activator of the dimeric form of protein phosphatase 2A. PTPA, cloned from human genomic libraries, is encoded by one single-copy gene, composed of 10 exons and 9 introns with a total length of about 60 kb. The transcription start site was determined, and the 5' flanking sequence was analyzed for its potential as a promotor. This region lacks a TATA sequence in the appropriate position relative to the transcription start, is very GC-rich, and contains upstream of the transcription start four Sp1 sites, a feature common to many TATA-less promotors. Based on the homology with DNA binding consensus sequences of transcription factors, we identified in this promotor region several putative DNA binding sites for transcription factors, such as NF-kappa B, Myb, Ets-1, Myc, and ATF. Transfection experiments with a construct containing the PTPA promotor region inserted 5' of a luciferase reporter gene revealed that the 5' flanking sequence of the PTPA gene indeed displayed promotor activity that seems to be cell-line dependent. By fluorescence in situ hybridization and G-banding, the PTPA gene was localized to the 9q34 region. The PTPA gene is positioned centromeric of c-abl in a region embracing several genes implicated in oncogenesis. PMID: 8530035 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 123: Blood. 1995 May 15;85(10):2711-9. The cloning and characterization of the human transcobalamin II gene. Regec A, Quadros EV, Platica O, Rothenberg SP. Division of Hematology/Oncology, State University of New York (SUNY)-Health Science Center, Brooklyn 11203, USA. Transcobalamin II (TCII) is a plasma protein that binds vitamin B12 (cobalamin; Cbl) and facilitates the cellular uptake of the vitamin by receptor-mediated endocytosis. In genetic disorders that are characterized by congenital deficiency of TCII, intracellular Cbl deficiency occurs, resulting in an early onset of megaloblastic anemia that is sometimes accompanied by a neurologic disorder. To define the genetic basis for TCII deficiency, we have cloned and characterized the human gene that encodes this protein. The gene spans a minimum of 18 kbp and contains nine exons and eight introns, with a polyadenylation signal sequence located 509 bp downstream from the termination codon and a transcription initiation site beginning 158 bp upstream from the ATG translation start site. The 5' flanking DNA does not have a TATA or CCAAT regulatory element, but a 34-nucleotide stretch beginning just upstream of the CAP site contains four tandemly organized 5'-CCCC-3' tetramers. This sequence is a motif for a trans-active transcription factor (ETF) that regulates expression of the epidermal growth factor receptor gene (EGFR), which also lacks TATA and CCAAT regulatory elements. A GC-rich sequence that binds the SP1 protein is located 356 nucleotides upstream from the first of the series of CCCC tetramers. Although this GC sequence is at an unusual location with respect to the CAP site, a 507-bp fragment containing this GC box drives the chloramphenicol acetyltransferase (CAT) reporter gene after transient transfection into NIH 3T3 cells. No CAT activity was observed when a 420-bp fragment lacking this GC box but containing the ETF-binding domains was similarly transfected into this cell line. One consensus and two atypical motifs for the c-myc ligand are located downstream and upstream, respectively, of the GC box, and this could explain the elevated plasma TCII observed in some patients with multiple myeloma, as the c-myc product is overexpressed in some myeloma cells. Restriction endonuclease digestion of genomic DNA from eight normal subjects with Taq I, Hinfl, Msp I, and Bgl I identified three patterns of restriction fragment length polymorphism (RFLP). A number of the exon/intron splice junctions of human TCII, TCI, and IF genes are located in homologous regions of these proteins, providing evidence that these genes have evolved by duplication of an ancestral gene. This characterization of the TCII gene and the RFLP should facilitate the identification of the mutation(s) responsible for the genetic abnormalities of TCII expression. PMID: 7742531 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 124: Oncogene. 1995 May 4;10(9):1841-8. Differential transcriptional regulation of c-myc promoter through the same DNA binding sites targeted by Sp1-like proteins. Majello B, De Luca P, Suske G, Lania L. Dipartimento di Genetica, Biologia Generale e Molecolare, Universita Federico II, Naples, Italy. Sp1 and Sp3 are closely related members of a gene family encoding proteins with very similar structural features. The zinc finger DNA binding domains of Sp1 and Sp3 are highly conserved and they bind to GC and GT box with comparable affinities. To begin to delineate the specific roles of these two members of the Sp1-like gene family, here we have analysed the DNA binding specificity and their effects on activation of human c-myc promoter. We found that both proteins bind to the same sites of c-myc promoter, upstream to both the P1 and P2 initiation sites. Cotransfection experiments, in mammalian and insect cells, indicated that Sp1 trans-activate c-myc promoter, whereas Sp3 did not. In addition, enforced expression of Sp3 repressed Sp1-mediated activation of c-myc. Finally, we found that Sp1 and E2F-1/DP-1 cooperatively trans-activate c-myc promoter. In contrast enforced expression of Sp3 fails to repress E2F-1/DP-1-mediated activation. PMID: 7753559 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 125: Bone. 1995 May;16(5):587-93. Characterization of the 5'-flanking region of the human tartrate-resistant acid phosphatase (TRAP) gene. Reddy SV, Kuzhandaivelu N, Acosta LG, Roodman GD. Department of Medicine/Hematology, Audie Murphy Veterans Administration Hospital, University of Texas Health Science Center, San Antonio 78284, USA. Tartrate-resistant acid phosphatase (TRAP) is expressed at high levels in osteoclasts and may play an important role in the bone resorptive process. However, factors regulating human TRAP gene expression have not been clearly defined. Therefore, we isolated a genomic clone (CL-9) for TRAP containing a 14-kb insert. A restriction map was generated for this insert, and a 2.6-kb ApaI fragment containing the 5'-flanking region was subcloned. Sequence analysis of this fragment revealed the presence of candidate transcription factor-binding sequences for H-APF-1, SP1, GATA2, and the c-Myc proto-oncogene. PCR analysis of RNA isolated from human osteoclastomas and pagetic bone revealed a 276-bp intron at -1 bp to -276 bp relative to the ATG and a transcript originating from this intron. Rapid amplification of the 5' end of the human TRAP mRNA by PCR indicated the presence of a 93-bp untranslated region 5' from the intron. Promoter activity was detected in the DNA fragment from +1 bp to -1903 bp relative to the ATG initiation codon, which drove the transient expression of a luciferase reporter gene when transfected into HRE H9 rabbit endometrial cells. Comparison of the human TRAP 5'-flanking region with mouse TRAP and uteroferrin revealed 41% and 47% homology, respectively. This suggests that regulation of human TRAP gene expression may differ from that for the murine TRAP gene. PMID: 7654474 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 126: Proc Natl Acad Sci U S A. 1995 Apr 25;92(9):3953-7. Functional interactions between the retinoblastoma (Rb) protein and Sp-family members: superactivation by Rb requires amino acids necessary for growth suppression. Udvadia AJ, Templeton DJ, Horowitz JM. Department of Molecular Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA. The transient expression of the retinoblastoma protein (Rb) regulates the transcription of a variety of growth-control genes, including c-fos, c-myc, and the gene for transforming growth factor beta 1 via discrete promoter sequences termed retinoblastoma control elements (RCE). Previous analyses have shown that Sp1 is one of three RCE-binding proteins identified in nuclear extracts and that Rb functionally interacts with Sp1 in vivo, resulting in the "superactivation" of Sp1-mediated transcription. By immunochemical and biochemical criteria, we report that an Sp1-related transcription factor, Sp3, is a second RCE-binding protein. Furthermore, in transient cotransfection assays, we report that Rb "superactivates" Sp3-mediated RCE-dependent transcription in vivo and that levels of superactivation are dependent on the trans-activator (Sp1 or Sp3) studied. Using expression vectors carrying mutated Rb cDNAs, we have identified two portions of Rb required for superactivation: (i) a portion of the Rb "pocket" (amino acids 614-839) previously determined to be required for physical interactions between Rb and transcription factors such as E2F-1 and (ii) a novel amino-terminal region (amino acids 140-202). Since both of these regions of Rb are targets of mutation in human tumors, our data suggest that superactivation of Sp1/Sp3 may play a role in Rb-mediated growth suppression and/or the induction of differentiation. PMID: 7732011 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 127: J Biol Chem. 1995 Apr 21;270(16):9494-9. Cellular nucleic acid binding protein regulates the CT element of the human c-myc protooncogene. Michelotti EF, Tomonaga T, Krutzsch H, Levens D. Department of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA. The CT element of the c-myc gene is required for promoter P1 usage and can drive expression of a heterologous promoter. Both double strand (Sp1) and single strand (hnRNP K) CT-binding proteins have been implicated as mediators of CT action. Although significant levels of CT activity persisted following Sp1 immunodepletion, EGTA totally abolished transactivation, thus implicating another metal requiring factor in CT element activity. As hnRNP K binds to one strand of the CT element, but has no metal requirement, the opposite (purine-rich strand) was examined as a target for a metal-dependent protein. A zinc-requiring purine strand binding activity was identified as cellular nucleic acid binding protein (CNBP), a protein previously implicated in the regulation of sterol responsive genes. Two forms of CNBP differed in their relative binding to the CT- or sterol-response elements. CNBP was shown to be a bona fide regulator of the CT element by cotransfection of a CNBP expression vector that stimulated expression of a CT-driven but not an AP1-dependent reporter. These data suggest that hnRNP K and CNBP bind to opposite strands and co-regulate the CT element. PMID: 7721877 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 128: Genomics. 1995 Apr 10;26(3):571-9. Retraction in: Hajra A, Collins FS. Genomics. 1996 Nov 15;38(1):107. Structure of the leukemia-associated human CBFB gene. Hajra A, Collins FS. Laboratory of Gene Transfer, National Center for Human Genome Research, National Institutes of Health, Bethesda, Maryland 20892, USA. We have determined the structure of the human CBFB gene, which encodes the beta subunit of the heterodimeric transcription factor core binding factor (CBF). This gene becomes fused to the MYH11 gene encoding smooth muscle myosin heavy chain by an inversion of chromosome 16 that occurs in the M4Eo subtype of acute myeloid leukemia. The CBFB gene contains 6 exons and spans 50 kb. The gene is highly conserved in animal species as distant as Drosophila, and the exon boundaries are in locations identical to those of the murine Cbfb homologue. The CBFB promoter region has typical features of a housekeeping gene, including high G+C content, high frequency of CpG dinucleotides, and lack of canonical TATA and CCAAT boxes. This gene has a single transcriptional start site, 345 nucleotides upstream of the beginning of the coding region. The human and mouse CBFB promoters show conservation of several transcriptional regulatory sequence motifs, including binding sites for Sp1, Ets family members, and Myc, but do not contain any CBF binding sites. The 5' end of the human CBFB gene also contains a highly polymorphic, transcribed CGG repeat that is not present in the murine homologue. Publication Types: Retracted Publication PMID: 7607682 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 129: Genes Dev. 1995 Mar 1;9(5):559-72. Promoter-proximal pausing of RNA polymerase II defines a general rate-limiting step after transcription initiation. Krumm A, Hickey LB, Groudine M. Fred Hutchinson Cancer Center, Seattle, Washington 98104, USA. We have shown previously that the majority of RNA polymerase II complexes initiated at the c-myc gene are paused in the promoter-proximal region, similar to observations in the Drosophila hsp70 gene. Our analyses define the TATA box or initiator sequences in the c-myc gene as necessary components for the establishment of paused RNA polymerase II. Deletion of upstream sequences or even the TATA box does not influence significantly the degree of transcriptional initiation or pausing. Deletion of both the TATA box and sequences at the transcription initiation site, however, abolishes transcriptional pausing of transcription complexes but still allows synthesis of full-length RNA. Further analyses with synthetic promoter constructs reveal that the simple combination of upstream activator with TATA consensus sequences or initiator sequences act synergistically to recruit high levels of RNA polymerase II complexes. Only a minor fraction of these complexes escapes into regions further downstream. Several different trans-activation domains fused to GAL4-DNA-binding domains, including strong activators such as VP16, do not eliminate promoter-proximal pausing of RNA polymerase. Thus, we conclude that pausing of RNA polymerase II is a common phenomenon in eukaryotic transcription and does not require complex promoter structures. Further analyses reveal that enhancers have a modest influence on transcription initiation and on release of transcription complexes out of the pause site but may function primarily to increase the elongation competence of transcription complexes. PMID: 7698646 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 130: J Hepatol. 1995;22(1 Suppl):34-7. Transactivation of cellular gene expression by hepatitis B viral proteins: a possible molecular mechanism of hepatocarcinogenesis. Caselmann WH. Department of Medicine II, University of Munich, Germany. Epidemiologic data indicate the crucial role of chronic hepatitis B virus (HBV) infection in hepatocellular carcinoma (HCC) development. On the molecular level, HBV sequences are frequently integrated in hepatocellular DNA. However, in contrast to the woodchuck model, in which specific HBV-DNA integration is detectable in most cases, insertional (in-) activation of cellular genes seems to be a rare event in man. The recent discovery of transactivating functions exerted by HBx and truncated HBs(urface) proteins supports the notion that transactivation of cellular gene expression could be relevant to hepatocarcinogenesis. HBV transactivator sequences are present in 81% (21/26) of HCC tissues or hepatoma-derived cell lines. At least one transactivator protein was functional in all cases investigated so far. The 16.5-kDa HBx transactivator has been shown to stimulate gene expression from various cellular target sequences. In vitro, HBx displays oncogenic potential. A second type of transactivator is encoded in the preS/S region of HBV. In contrast to HBx, HBs transactivators require carboxyterminal truncation to gain their transactivating function. Unlike full-length M(iddle)HBs, the truncated MHBst is retained in the endoplasmic reticulum and not secreted into the surrounding medium. Cellular gene expression is stimulated by regulatory elements of the human proto-oncogenes c-fos and c-myc, as well as by the hepatic acute-phase interleukin-6 gene. Synthetic binding sites for the transcription factors NF-kappa B, AP-1, AP-2, SRE, and Sp1 render minimal promoters activatable. NF-kappa B-mediated transactivation by MHBst can be suppressed by radical scavenging antioxidants, indirectly suggesting that reactive oxygen intermediates are involved.(ABSTRACT TRUNCATED AT 250 WORDS) Publication Types: Review Review, Tutorial PMID: 7602073 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 131: FEBS Lett. 1994 Oct 17;353(2):180-4. Structure of the gene for human uracil-DNA glycosylase and analysis of the promoter function. Haug T, Skorpen F, Lund H, Krokan HE. UNIGEN Center for Molecular Biology, University of Trondheim, Norway. The gene for human uracil-DNA glycosylase (UNG) contains 4 exons and has an approximate size of 13 kb. The promoter is very GC rich and lacks a TATA box. Nested deletions of the promoter demonstrated that two SP1 elements and a putative c-MYC element proximal to the transcription initiation region were sufficient to support some 27% of the promoter activity, while a clone that in addition contained the elements E2F/SP1/CCAAT increased expression to almost 90% of the full-length construct. A region upstream of these elements appears to exert a negative control function. PMID: 7926048 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 132: 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] --------------------------------------------------------------- 133: Mol Endocrinol. 1994 Sep;8(9):1163-74. Association of USF and c-Myc with a helix-loop-helix-consensus motif in the core promoter of the murine type II beta regulatory subunit gene of cyclic adenosine 3', 5'-monophosphate-dependent protein kinase. Singh IS, Luo Z, Kozlowski MT, Erlichman J. Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461. Previous studies showed that the core promoter of the mouse cAMP-dependent protein kinase regulatory subunit type II beta (RII beta) gene was composed of two functional elements. One element was GC rich and bound the Sp1 transcription factor. The second element contained a helix-loop-helix (HLH)-motif. Each element conferred transcriptional activity when inserted upstream of a reporter gene, chloramphenicol acetyltransferase and transfected into mouse NB2a neuroblastoma cells and Chinese hamster ovary (CHO) cells. The core promoter was further characterized by mutational analysis using electrophoretic mobility shift assays and by transfection into CHO and NB2a cells. Electrophoretic mobility shift assays showed that the HLH-consensus motif, CACGTG, present in the RII beta gene bound nuclear factors present in NB2a and CHO cells. Mutations in the HLH-core motif decreased the binding of these factors and reduced the transcriptional activity of constructs containing the chloramphenicol acetyltransferase reporter when transfected into these cells. The results showed that the central nucleotides as well as the adjacent bases were important for the interaction with the nuclear binding factors. UV cross-linking, Southwestern blot analysis, and interference of the mobility shift patterns by specific antisera directed against USF and c-Myc indicated that both of these transcription factors were forming complexes with the HLH-consensus motif. The results suggest that RII beta transcription may be regulated, in part, by USF and c-Myc in NB2a and CHO cells. PMID: 7838149 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 134: Leukemia. 1994 Jul;8(7):1157-63. Ectopic expression of myc or myn down-regulates immunoglobulin transcription. Sigvardsson M, Johansson K, Larsson LG, Nilsson K, Leanderson T. Immunology Unit, Lund University, Sweden. Co-transfection of expression vectors for c-myc or myn down-regulated the expression from a reporter plasmid containing the chloramphenicol-acetyl-transferase (CAT) gene, under the control of an immunoglobulin kappa promoter and the immunoglobulin heavy-chain enhancer. The effect was dose-dependent and an additive effect was seen when both expression vectors were transfected simultaneously. Deletion mutants lacking the leucine zipper region of c-myc were unable to down-regulate the transcription from the reporter construct. Reporter genes where the immunoglobulin enhancer had been exchanged with a SV40 enhancer or an immunoglobulin minimal enhancer were still down-regulated by a cotransfection with c-myc expression vectors. A minimal promoter containing an octamer motif responded to myc expression while a minimal promoter containing a SP1 motif did not. No direct interactions between myc, myn and Oct proteins could be detected either by band-shift analysis, or by co-translation in vitro followed by precipitation with SP6 kappa promoter bound to magnetic carriers. Furthermore, B cells from mice transgenic for myc showed aberrant expression of transcription factors. PMID: 8035607 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 135: Mol Cell Biol. 1993 Dec;13(12):7612-24. CTCF, a conserved nuclear factor required for optimal transcriptional activity of the chicken c-myc gene, is an 11-Zn-finger protein differentially expressed in multiple forms. Klenova EM, Nicolas RH, Paterson HF, Carne AF, Heath CM, Goodwin GH, Neiman PE, Lobanenkov VV. Institute of Carcinogenesis, Russian Cancer Research Center, Moscow. A novel sequence-specific DNA-binding protein, CTCF, which interacts with the chicken c-myc gene promoter, has been identified and partially characterized (V. V. Lobanenkov, R. H. Nicolas, V. V. Adler, H. Paterson, E. M. Klenova, A. V. Polotskaja, and G. H. Goodwin, Oncogene 5:1743-1753, 1990). In order to test directly whether binding of CTCF to one specific DNA region of the c-myc promoter is important for chicken c-myc transcription, we have determined which nucleotides within this GC-rich region are responsible for recognition of overlapping sites by CTCF and Sp1-like proteins. Using missing-contact analysis of all four nucleotides in both DNA strands and homogeneous CTCF protein purified by sequence-specific chromatography, we have identified three sets of nucleotides which contact either CTCF or two Sp1-like proteins binding within the same DNA region. Specific mutations of 3 of 15 purines required for CTCF binding were designed to eliminate binding of CTCF without altering the binding of other proteins. Electrophoretic mobility shift assay of nuclear extracts showed that the mutant DNA sequence did not bind CTCF but did bind two Sp1-like proteins. When introduced into a 3.3-kbp-long 5'-flanking noncoding c-myc sequence fused to a reporter CAT gene, the same mutation of the CTCF binding site resulted in 10- and 3-fold reductions, respectively, of transcription in two different (erythroid and myeloid) stably transfected chicken cell lines. Isolation and analysis of the CTCF cDNA encoding an 82-kDa form of CTCF protein shows that DNA-binding domain of CTCF is composed of 11 Zn fingers: 10 are of C2H2 class, and 1 is of C2HC class. CTCF was found to be abundant and conserved in cells of vertebrate species. We detected six major nuclear forms of CTCF protein differentially expressed in different chicken cell lines and tissues. We conclude that isoforms of 11-Zn-finger factor CTCF which are present in chicken hematopoietic HD3 and BM2 cells can act as a positive regulator of the chicken c-myc gene transcription. Possible functions of other CTCF forms are discussed. PMID: 8246978 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 136: Nucleic Acids Res. 1993 Nov 11;21(22):5092-100. Specific cleavage of transcription factors by the thiol protease, m-calpain. Watt F, Molloy PL. CSIRO Division of Biomolecular Engineering, Sydney Laboratory, North Ryde, NSW, Australia. The intracellular nonlysosomal calcium-dependent cysteine protease, m-calpain, is shown to specifically cleave the bHLHzip transcription factor USF leaving the binding and dimerisation domains intact. The resultant protein is capable of efficient DNA binding but is no longer able to activate transcription. A surprisingly high proportion of other transcription factors tested, AP1 (c-Fos/c-Jun), Pit-1, Oct-1, CP1a and b, c-Myc, ATF/CREB, AP2 and AP3 but not Sp1, were similarly cleaved by m-calpain to produce specific partial digestion products. These properties make m-calpain a particularly useful protease for proteolytic studies of transcription factors and also raise the possibility that m-calpain may be involved in vivo in regulation of turnover or transcriptional activity of a number of transcription factors. PMID: 8255762 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 137: Trends Biochem Sci. 1993 Nov;18(11):433-7. DNA damage and the DNA-activated protein kinase. Anderson CW. Biology Department, Brookhaven National Laboratory, Upton, NY 11973-5000. DNA-activated protein kinase (DNA-PK) is a nuclear serine/threonine protein kinase that is activated in vitro by DNA fragments. The cellular targets of DNA-PK are nuclear, DNA-binding, regulatory proteins including Sp1, Fos, Jun, Myc, the tumor suppressor protein p53, and RNA polymerase II. These characteristics suggest a role for DNA-PK in coordinating nuclear processes and as a modulator of checkpoint mechanisms activated by DNA damage. Publication Types: Review Review, Tutorial PMID: 8291090 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 138: Eur J Biochem. 1993 Nov 1;217(3):813-22. The promoter region of the human type-I-DNA-topoisomerase gene. Protein-binding sites and sequences involved in transcriptional regulation. Heiland S, Knippers R, Kunze N. Division of Biology, University of Konstanz, Germany. We examined the promoter of the human type-I-DNA topoisomerase gene (hTOP1) for regions protected against DNase I digestion by nuclear proteins from HeLa or from adenovirus-transformed 293 cells. We identified ten protected DNA sequences within 580 bp of DNA upstream of the transcriptional-start sites and one additional site, which is located between the two clusters of transcriptional-start sites. Several of these protein-binding sites have significant similarities to recognition sequences of known transcription factors including factors Sp1, octamer transcription factor, cAMP-responsive-element-binding protein (CREB/ATF), NF-kappa B and members of the Myc-related family of basic/helix-loop-helix/leucine-zipper proteins. Other protein-binding sites show less or no similarities to known consensus sequences. We investigated the physiological significance of these protein-binding sites using a set of deletion and nucleotide-exchange mutants. We conclude that the expression of the hTOP1 gene is regulated by a complex network of negatively and positively acting transcription factors. PMID: 8223637 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 139: Mol Cell Biol. 1993 Sep;13(9):5710-24. Repeated CT elements bound by zinc finger proteins control the absolute and relative activities of the two principal human c-myc promoters. DesJardins E, Hay N. Ben May Institute, University of Chicago, Illinois 60637. Transcription of the human proto-oncogene c-myc is governed by two tandem principal promoters, termed P1 and P2. In general, the downstream promoter, P2, is predominant, which is in contrast to the promoter occlusion phenomenon usually observed in genes containing tandem promoters. A shift in human c-myc promoter usage has been observed in some tumor cells and in certain physiological conditions. However, the mechanisms that regulate promoter usage are not well understood. The present studies identify regulators which are required to promote transcription from both human c-myc promoters, P1 and P2, and have a role in determining their relative activities in vivo. A novel regulatory region located 101 bp upstream of P1 was characterized and contains five tandem repeats of the consensus sequence CCCTCCCC (CT element). The integrity of the region containing all five elements is required to promote transcription from P1 and for maximal activity from P2 in vivo. A single copy of this same element, designated CT-I2, also appears in an inverted orientation 53 bp upstream of the P2 transcription start site. This element has an inhibitory effect on P1 transcription and is required for P2 transcription. The transcription factor Sp1 was identified as the factor that binds specifically to the tandem CT elements upstream of P1 and to the CT-I2 element upstream of P2. In addition, the recently cloned zinc finger protein ZF87, or MAZ, was also able to bind these same elements in vitro. The five tandem CT elements can be functionally replaced by a heterologous enhancer that only in the absence of CT-I2 reverses the promoter usage, similar to what is observed in the translocated c-myc allele of Burkitt's lymphoma cells. PMID: 8355712 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 140: Proc Natl Acad Sci U S A. 1993 Apr 15;90(8):3265-9. Sp-1 binds promoter elements regulated by the RB protein and Sp-1-mediated transcription is stimulated by RB coexpression. Udvadia AJ, Rogers KT, Higgins PD, Murata Y, Martin KH, Humphrey PA, Horowitz JM. Section of Cell Growth, Regulation and Oncogenesis, Duke University Medical Center, Durham, NC 27710. The retinoblastoma (RB) protein is implicated in transcriptional regulation of at least five cellular genes, including c-fos, c-myc, and transforming growth factor beta 1. Cotransfection of RB and truncated promoter constructs has defined a discrete element (retinoblastoma control element; RCE) within the promoters of each of these genes as being necessary for RB-mediated transcription control. Previously, we have shown that RCEs form protein-DNA complexes in vitro with three heretofore unidentified nuclear proteins and mutation of their DNA-binding site within the c-fos RCE results in an abrogation of RCE-dependent transcription in vivo. Here, we demonstrate that one of the nuclear proteins that binds the c-fos, c-myc, and transforming growth factor beta 1 RCEs in vitro is Sp-1 and that Sp-1 stimulates RCE-dependent transcription in vivo. Moreover, we show that Sp-1-mediated transcription is stimulated by the transient coexpression of RB protein. We conclude from these observations that RB may regulate transcription in part by virtue of its ability to functionally interact with Sp-1. PMID: 8475068 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 141: Oncogene. 1993 Mar;8(3):645-54. Regulation of Xenopus c-myc promoter activity in oocytes and embryos. Modak SP, Principaud E, Spohr G. Universite de Geneve, Departement de Biologie Cellulaire, Geneva, Switzerland. We have studied the regulation of transcription of the Xenopus c-myc I gene in oocytes and embryos. Various 5' and internal deletions of a 1310-bp-long c-myc I promoter fragment have been ligated upstream of the chloramphenicol acetyl transferase (CAT) reporter gene and microinjected into oocytes and fertilized eggs. Activity was determined by CAT assay and primer extension. The c-myc promoter drives transcription very efficiently, and a truncated promoter -158/+46 essentially retains full activity. This region contains an overlapping E2F/SP1 site and two tandem Sp1 sites homologous to those found in the c-myc gene of mouse. Internal deletions show that both elements are equally active in oocytes in driving the expression of CAT. A germinal vesicle extract contains a DNA-binding activity specific for an Sp1 consensus sequence but not the E2F site. The data suggest that the high transcription level of the endogenous c-myc gene in Xenopus oocytes is mediated by Sp1 or a related transcription factor. In embryos a different mechanism emerges and the functional role of the Sp1 binding sites appears to be less important. PMID: 8437848 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 142: Mol Endocrinol. 1993 Mar;7(3):387-98. Regulation of transglutaminase type I expression in squamous differentiating rabbit tracheal epithelial cells and human epidermal keratinocytes: effects of retinoic acid and phorbol esters. Saunders NA, Bernacki SH, Vollberg TM, Jetten AM. Cell Biology Section, National Institute of Environmental Health Sciences, National Institutes of Health Research Triangle Park, North Carolina 27709. In the present study we describe the full length cDNA sequence for rabbit transglutaminase type I as well as the sequence for a 2.9-kilobase (kb) promoter fragment of the gene. Transglutaminase type I mRNA expression was inhibited in squamous differentiating epithelia by retinoic acid (RA) in a dose-dependent (EC50 = 1-2 nM) and transcriptional manner. In human epidermal keratinocytes transglutaminase type I mRNA was induced by 12-O-tetradecanoylphorbol-13-acetate (TPA) treatment, and this induction could be inhibited by bryostatin 1. In contrast, TPA treatment inhibited the expression of c-myc mRNA. Bryostatin 1 but not RA could prevent this decrease in c-myc mRNA expression, indicating that transglutaminase type I mRNA expression was associated with differentiation and not growth arrest. An SP1 element was found within 50 base pairs 5' of the transcription initiation site. A TATA-like element (CATAAAC) was found but was not capable of activating transcription. In addition, putative response elements for C-MYC, Ker1/AP2, 2 AP1 sites, a CK-8-mer, and an AP2 site were present in the 2.9-kb fragment. Transfection of RbTE cells with the 2.9-kb fragment ligated to a promoterless luciferase vector resulted in 2.2-fold more luciferase expression in differentiated vs. undifferentiated cells. Furthermore, luciferase activity was induced 7.4-fold in human epidermal keratinocytes induced to differentiate with TPA. TPA-induced luciferase activity was inhibited by both bryostatin 1 and RA. No known RA response elements were identified in the promoter.(ABSTRACT TRUNCATED AT 250 WORDS) PMID: 8097865 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 143: Genes Chromosomes Cancer. 1993 Feb;6(2):124-31. Regulation of transcription by the retinoblastoma protein. Horowitz JM. Section of Cell Growth, Regulation and Oncogenesis, Duke University Medical Center, Durham, NC 27710. The product of the retinoblastoma gene (RB1) is believed to function as a negative regulator of cell growth. Recent experimental results suggest that RB1 may exert its growth-suppressing activity by regulating the transcription of a variety of growth-related genes, including FOS, MYC, and TGFBI. A series of biochemical and molecular analyses suggest that RB1 indirectly affects gene expression via cell-cycle-regulated interactions with transcription factors, such as E2F and SPI. Determination of the mechanisms regulating such protein-protein interactions and the identification of additional targets of RB1 function will provide vital insights into the role of this tumor-suppressor gene in mammalian cell proliferation. Publication Types: Review Review, Tutorial PMID: 7680889 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 144: Mol Cell Biol. 1992 Jun;12(6):2455-63. The retinoblastoma gene product regulates Sp1-mediated transcription. Kim SJ, Onwuta US, Lee YI, Li R, Botchan MR, Robbins PD. Laboratory of Chemoprevention, National Cancer Institute, Bethesda, Maryland 20892. We have demonstrated that the retinoblastoma gene product (Rb) can positively regulate transcription from the fourth promoter of the insulinlike growth factor II gene. Two copies of a motif (the retinoblastoma control element) similar to that found in the human c-fos, transforming growth factor beta 1, and c-myc promoters are responsible for conferring Rb regulation to the fourth promoter of the insulinlike growth factor II gene. We have shown that the transcription factor Sp1 can bind to and stimulate transcription from the retinoblastoma control element motif. Moreover, by using a GAL4-Sp1 fusion protein, we have directly demonstrated that Rb positively regulates Sp1 transcriptional activity in vivo. These results indicate that Rb can function as a positive regulator of transcription and that Sp1 is one potential target, either directly or indirectly, for transcriptional regulation by Rb. PMID: 1588949 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 145: Oncogene. 1991 Nov;6(11):2067-75. Regulatory domains within the P0 promoter of human c-myc. Lang JC, Wilkie NM, Clark AM, Chudleigh A, Talbot S, Whitelaw B, Frame MC. Beatson Institute for Cancer Research, Glasgow, UK. Expression of P0 RNA in some Burkitt lymphoma cell lines varies independently of levels of RNA derived from P1 and P2. These data suggest the possibility that expression of P0 RNA may be capable of independent regulation. In order to investigate this possibility we have isolated putative regulatory domains flanking P0 RNA starts within the human c-myc gene and analysed both their ability to direct expression of control reporter genes and their ability to interact with specific transcription factors. Regulatory regions necessary for expression of P0 RNA have been located within 131 bp 5' of the first major P0 RNA start. DNAase 1 footprint analysis and gel retardation assays demonstrate binding of transcription factors Sp1, NF1 and CBP to this region. NF1 binds specifically to two consensus sequences. The more distal site overlaps with the binding site for CBP, and it is likely that concomitant binding of NF1 and CBP within the distal region of the P0 promoter is not possible. Previous work from our laboratory has described a negative regulatory domain within the 5' flanking region of c-myc. The P0 promoter resides within this domain and therefore may contain a negative regulator of c-myc gene expression. PMID: 1945411 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 146: Oncogene. 1991 Oct;6(10):1843-50. Erratum in: Oncogene. 1992 May;7(5):1047. Characterization of the human N-ras promoter region. Thorn JT, Todd AV, Warrilow D, Watt F, Molloy PL, Iland HJ. Kanematsu Laboratories, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia. Overexpression of ras proto-oncogenes has been implicated in cancer development. We therefore initiated a study of the human N-ras promoter to determine the regions that control N-ras expression and their potential for interaction with DNA-binding proteins. N-ras CAT constructs were stably integrated into K562 cells by electric field-mediated gene transfer in order to determine functional regions within the human N-ras promoter. A significant proportion of promoter activity was found to lie within a 439 bp fragment comprising an untranslated exon (exon 1) with the adjacent 5' sequence and a small CpG island. A 109 bp [corrected] fragment at the 5' end of exon 1 was essential for promoter activity, while a 45 bp [corrected] deletion from within this region decreased promoter activity by two-thirds. Unlike the human H-ras and mouse K-ras promoters, the N-ras promoter did not exhibit bidirectional activity. DNAse footprinting of the 439 bp fragment revealed seven protected regions, many of which contain sequences homologous to known DNA-binding protein sites (MLTF/myc, CREB/ATF, AP-1, AP-2, myb and E4TF1). In contrast, four putative Sp1 sites did not footprint. Using purified MLTF and appropriate competitors in gel shift and DNAase footprinting assays, we demonstrated binding of MLTF to the MLTF consensus sequence within exon 1. PMID: 1923508 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 147: Nucleic Acids Res. 1991 Jun 11;19(11):3081-8. Xenopus laevis c-myc I and II genes: molecular structure and developmental expression. Principaud E, Spohr G. Universite de Geneve, Departement de Biologie Cellulaire, Switzerland. The structure of the two Xenopus laevis c-myc I and c-myc II genes has been investigated by isolating and sequencing genomic and cDNAs clones. In oocytes, c-myc I mRNAs represent 80-90% of the overall amount of c-myc transcripts. The c-myc I expression is controlled primarily by two differentially regulated tandem promoters P1 and P2 which are separated by 50 bases. During oogenesis, maternal c-myc I mRNAs, are transcribed from both promoters whereas zygotic transcripts seem to initiate only from the P2 promoter. Sequence comparison between the promoter regions of c-myc I and II genes reveals the insertion in the c-myc I promoter region, between positions -831 and -389 relative to the P1 start site of a repetitive element. Comparison of X.laevis and mammalian c-myc promoter sequences reveals furthermore the conservation of cis-regulatory elements, including a motif known to be a negative regulator of the human c-myc transcription, a purine rich region, a binding site for the E2-F transcription factor and three SP1 binding sites. Finally, we report characterization of a new c-myc I mRNA which differ at the 5' end. Transcripts are possibly initiated at a putative alternative promoter located further upstream in the genome, and undergoes alternative splicing. PMID: 2057364 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 148: Biochemistry. 1991 Apr 30;30(17):4290-7. Mithramycin blocks transcriptional initiation of the c-myc P1 and P2 promoters. Snyder RC, Ray R, Blume S, Miller DM. Department of Internal Medicine, University of Alabama, Birmingham. The c-myc protooncogene plays an important role in the regulation of cellular proliferation. Mithramycin, a DNA binding antibiotic which binds G-C-rich DNA, inhibits c-myc expression in both differentiating and nondifferentiating cells. The G-C-rich nature of the c-myc promoter suggests that mithramycin may act by directly inhibiting promoter function. The mithramycin binding sites in the c-myc promoter regions were determined by DNAse I footprinting. Particularly prominent mithramycin binding is noted in the regions just 5' of the P1 and P2 promoter TATA boxes. Gel retardation experiments performed in the presence of mithramycin demonstrate that drug binding can prevent the formation of discrete complexes between HeLa cell nuclear proteins and c-myc promoter DNA fragments. Mithramycin also directly blocks the binding of the transcription factor Sp1 to the P1 promoter region. In vitro run-off transcription demonstrates that mithramycin can completely inhibit the in vitro function of both the P1 and P2 promoters. These data suggest that mithramycin inhibits transcription of the c-myc protooncogene by blocking the binding of important regulatory factors, thus preventing formation of the c-myc transcription initiation complex. PMID: 1827033 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 149: Oncogene. 1990 Dec;5(12):1743-53. A novel sequence-specific DNA binding protein which interacts with three regularly spaced direct repeats of the CCCTC-motif in the 5'-flanking sequence of the chicken c-myc gene. Lobanenkov VV, Nicolas RH, Adler VV, Paterson H, Klenova EM, Polotskaja AV, Goodwin GH. Institute of Carcinogenesis, All-Union Cancer Research Center, Moscow, USSR. The chicken c-myc 5'-flanking sequence has previously been shown to bind multiple proteins present in undifferentiated and differentiated red blood cells. In this report the protein binding to one specific region within a hypersensitive site approximately 200 base pairs upstream of the start of transcription has been analysed in detail. Using a combination of a modified agarose gel retardation assay with O-phenanthroline-copper footprinting in situ, missing contact point and methylation interference techniques, two proteins were found to bind to overlapping sequences within 180-230 bp upstream of the start of transcription. One protein resembles the transcription factor Sp1, the other is a protein which binds to three regularly spaced repeats of the core sequence CCCTC. This CCCTC-binding factor was termed CTCF. It requires additional sequences outside the three recognition motifs for tight binding. CTCF was purified to near homogeneity by sequence-specific DNA chromatography. The approximate molecular weight of the CTCF was estimated to be 130,000. Removal of 110 bp sequence binding both CTCF and Sp1-like proteins leads to a 4 to 8-fold increase in transcription of stably transfected c-myc fusion constructs in chicken embryonic fibroblasts, suggesting that the CTCF is likely to be one of multiple nuclear factors involved in the transcriptional regulation of the chicken c-myc gene. PMID: 2284094 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 150: J Biol Chem. 1990 Nov 15;265(32):19961-5. Characterization of the 5'-flanking region of the rat protein kinase C gamma gene. Chen KH, Widen SG, Wilson SH, Huang KP. Endocrinology and Reproduction Research Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892. The 5'-flanking region of protein kinase C (PKC) gamma gene was identified from a rat liver genomic library in a bacteriophage lambda Charon 4A. A 3.6-kilobase (kb) genomic fragment containing the 5'-flanking region, first exon, and first intron was isolated and sequenced. The transcriptional initiation site, identified by S1 mapping and primer extension, was located 243 base pairs upstream from the translational initiation site. Promoter activity of a DNA segment spanning the 5'-flanking region was demonstrated by both in vitro transcription using HeLa cell nuclear extracts and chloramphenicol acetyltransferase assay by transfection of 293 cells with a PKC gamma-CAT fusion construct. Chloramphenicol acetyltransferase assay revealed that a fragment of about 0.16 kb from the transcriptional initiation site was sufficient for promoter activity in these cells, and the construct containing up to 1.6 kb from the cap site was expressed at a similar level. This promoter-active fragment contains several regions similar to defined transcriptional elements in other mammalian promoters, such as those for stimulatory protein 1 (Sp1), activator proteins 1 and 2 (AP1, AP2), c-myc, cAMP regulatory element-binding protein (CREB), and enhancer core (EnhC). Investigation of the genomic structure of PKC gamma gene may lead to the identification of cis-elements controlling tissue-specific and developmental stage-specific expression of PKC gamma. PMID: 2246272 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 151: Oncogene. 1989 May;4(5):549-58. Molecular requirements for transcriptional initiation of the murine c-myc gene. Asselin C, Nepveu A, Marcu KB. Department of Biochemistry, SUNY, Stony Brook 11794-5215. We have identified sequences in the 5' flanking region of the murine c-myc gene's P1 and P2 transcription initiation sites which form specific complexes with nuclear factors of murine and human origin and are also required for normal P1 and P2 usage. Four nuclear factor binding sites were identified within 400 bp 5' of P1 (5'Mf, 5'Mg1, 5'Mg2, and 5'Mg3) and two others within 100 bp 5' of P2 (ME1a1 and ME1a2). The Sp1 transcription factor bound to 5'Mg1 and 5'Mg3 with high affinity and with low affinity to 5'Mg2, ME1a1 and ME1a2 which also bound with high affinity to other factors in crude nuclear extracts. Deletion mutagenesis of sequences 5' of the P1 initiation site revealed that 109 bp encompassing 5'Mg3 and a TATA sequence were sufficient for P1 usage. The ME1a1 binding site 5' of P2 was necessary for maximal P2 activity and the loss of this sequence resulted in enhanced P1 usage. These findings demonstrate that the P1 and P2 initiation sites are independently regulated and that the ME1a1 binding site plays a central role in the normal usage of the c-myc promoters. PMID: 2471130 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 152: Br J Cancer Suppl. 1988 Dec;9:62-6. Transcriptional regulation of the human c-myc gene. Lang JC, Whitelaw B, Talbot S, Wilkie NM. Beatson Institute for Cancer Research, Bearsden, Glasgow, UK. The involvement of c-myc in the genesis of animal neoplasia is now well documented for several systems. In order to define the precise role played by the myc gene in tumorigenesis, a better understanding of the normal regulation of myc expression is necessary. We have begun a study of the cis-acting regulatory sequences within the 5' flanking domain of the human c-myc gene. Regions important for myc promoter function have been identified by linkage to the coding sequences of the bacterial chloramphenicol acetyltransferase (cat) gene. Promoter deletion studies and in vivo competition assays for c-myc/cat recombinant plasmids have allowed the identification of a proximal 'core' promoter region capable of directing high levels of CAT activity. Further upstream a negative regulatory element (NRE2) has been identified which is capable of repressing cat gene expression and which functions by interaction with a transacting factor(s). Preliminary data suggests detection of NRE2 is dependent on both the type and amount of carrier DNA used in transient CAT assays. Initial experiments further indicate the involvement of at least two other distal regulatory domains, a negative regulatory domain (NRE1) and a putative enhancer-type region (E). In vitro footprint analysis has allowed the identification of DNA binding proteins which interact with NRE2 and the 'core' promoter. NRE2 contains binding sites for transcription factors Sp1 and CTF. The 'core' promoter domain appears to be highly complex and possesses several Sp1 binding sites. Publication Types: Review Review, Tutorial PMID: 3076067 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 153: J Virol. 1988 Nov;62(11):4070-7. Multiple protein-binding sites in an intracisternal A particle long terminal repeat. Falzon M, Kuff EL. Laboratory of Biochemistry, National Cancer Institute, Bethesda, Maryland 20892. The long terminal repeats (LTRs) of cloned intracisternal A particles (IAPs) can function as effective promoters in heterologous and homologous cell types (K. K. Lueders, J. W. Fewell, E. L. Kuff, and T. Koch, Mol. Cell. Biol. 4:2128-2135, 1984) and respond to transcriptional factors induced by various nuclear oncogene products (S. Luria and M. Horowitz, J. Virol. 57:998-1003, 1986). Using the first 139 base pairs of the U3 region of a cloned mouse IAP LTR as probe, we demonstrated multiple exonuclease III stop sites which appeared specifically in the presence of nuclear extract protein. Various extracts gave similar footprints, but the amount of nuclear protein required varied up to 10-fold. Cell lines transformed with known nuclear oncogenes, such as adenovirus E1a and E1b (293 cells), simian virus 40 large T antigen (COS7 cells), and c-myc (MOPC-315 cells) had more and/or higher-affinity factors for the IAP LTR than extracts from HeLa, CV1, and NIH 3T3 cells did. DNase I footprinting revealed at least five distinct protein-binding domains within the 139-base-pair region. These domains correspond to segments of highly conserved nucleotide sequence among a number of IAP LTRs. Gel retardation studies with oligonucleotides encompassing the DNase I footprint sites showed that the nuclear factors are present in different proportions and different absolute levels in extracts from different cell types. Moreover, the oligonucleotide probes indicate that individual motifs can be occupied independently of one another. Three of the DNase I footprints include a sequence with homology to the simian virus 40 core enhancer and sequence motifs that closely resemble the binding sites for transcription factors SP1 and AP-1. The other two binding sites are not obviously related to previously recognized motifs. The multiple protein-binding sites in close proximity indicate the complex regulatory mechanism for IAP transcription. PMID: 3139894 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 154: Mol Cell Biol. 1986 Nov;6(11):4093-8. Sequences involved in accurate and efficient transcription of human c-myc genes microinjected into frog oocytes. Nishikura K. By microinjecting a series of deletion mutant constructs into Xenopus laevis oocytes, transcriptional control regions, two promoters, of the human c-myc gene were defined. In the case of the first promoter, sequences between -60 and -37 relative to the transcription start site contained an element essential for promoter activity. In the case of the second promoter, sequences between -66 and -56 relative to the initiation site appeared to be involved in accurate and efficient transcription. In both cases, the region identified as the essential promoter element contained GGGCGG or GGCGGG,GC box-like sequences, suggesting that c-myc gene promoter activity may be controlled by transcription factor Sp1 binding in the microinjected oocytes. PMID: 3025632 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 155: Eur J Biochem. 1986 Aug 15;159(1):181-8. Sequence-specific DNA-binding proteins which interact with (G + C)-rich sequences flanking the chicken c-myc gene. Lobanenkov VV, Nicolas RH, Plumb MA, Wright CA, Goodwin GH. The interaction of nuclear sequence-specific DNA-binding proteins from definitive chicken erythrocytes, thymus and proliferating transformed erythroid precursor (HD3) cells with the 700-base-pair (700-bp) DNA 5'-flanking region of the chicken c-myc gene was investigated by in vitro footprint analysis. The major HD3 protein-binding activity binds to a site (site V) 200 bp upstream from the 'cap' site but, after further fractionation, a second distinct binding activity is detected to a site (site VIII) which contains both the 'CAAT' and 'SP1-binding' consensus sequences. Protein from thymus and erythrocyte cells which express c-myc at lower levels, bind to seven and eight sites respectively. In common with HD3 cell protein, they both bind to site VIII and, although binding to the sequence at site V is also detected, the footprint protection pattern is sufficiently different (site V') to suggest the involvement of different proteins in terminally differentiated and proliferating cells. The DNA-binding activities were partially fractionated by high-performance liquid chromatography gel filtration and include an erythrocyte-specific protein which binds to a c-myc gene poly(dG) homopolymer sequence similar to that found upstream of the chicken beta A-globin gene. PMID: 3743569 [PubMed - indexed for MEDLINE] ---------------------------------------------------------------