Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Theranostics ; 9(24): 7268-7281, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31695767

RESUMO

Rationale: An imbalance between protein synthesis and degradation is one of the mechanisms of cardiac hypertrophy. Increased transcription in cardiomyocytes can lead to excessive protein synthesis and cardiac hypertrophy. Maf1 is an RNA polymerase III (RNA pol III) inhibitor that plays a pivotal role in regulating transcription. However, whether Maf1 regulates of cardiac hypertrophy remains unclear. Methods: Cardiac hypertrophy was induced in vivo by thoracic aortic banding (AB) surgery. Both the in vivo and in vitro gain- and loss-of-function experiments by Maf1 knockout (KO) mice and adenoviral transfection were used to verify the role of Maf1 in cardiac hypertrophy. RNA pol III and ERK1/2 inhibitor were utilized to identify the effects of RNA pol III and ERK1/2. The possible interaction between Maf1 and ERK1/2 was clarified by immunoprecipitation (IP) analysis. Results: Four weeks after surgery, Maf1 KO mice exhibited significantly exacerbated AB-induced cardiac hypertrophy characterized by increased heart size, cardiomyocyte surface area, and atrial natriuretic peptide (ANP) expression and by exacerbated pulmonary edema. Also, the deficiency of Maf1 causes more severe cardiac dilation and dysfunction than wild type (WT) mice after pressure overload. In contrast, compared with adenoviral-GFP injected mice, mice injected with adenoviral-Maf1 showed significantly ameliorated AB-induced cardiac hypertrophy. In vitro study has demonstrated that Maf1 could significantly block phenylephrine (PE)-induced cardiomyocyte hypertrophy by inhibiting RNA pol III transcription. However, application of an RNA pol III inhibitor markedly improved Maf1 knockdown-promoted cardiac hypertrophy. Moreover, ERK1/2 was identified as a regulator of RNA pol III, and ERK1/2 inhibition by U0126 significantly repressed Maf1 knockdown-promoted cardiac hypertrophy accompanied by suppressed RNA pol III transcription. Additionally, IP analysis demonstrated that Maf1 could directly bind ERK1/2, suggesting Maf1 could interact with ERK1/2 and then inhibit RNA pol III transcription so as to attenuate the development of cardiac hypertrophy. Conclusions: Maf1 ameliorates PE- and AB-induced cardiac hypertrophy by inhibiting RNA pol III transcription via ERK1/2 signaling suppression.


Assuntos
Cardiomegalia/metabolismo , RNA Polimerase III/metabolismo , Proteínas Repressoras/metabolismo , Animais , Cardiomegalia/etiologia , Cardiomegalia/genética , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenilefrina/efeitos adversos , RNA Polimerase III/antagonistas & inibidores , RNA Polimerase III/genética , Ratos , Ratos Sprague-Dawley , Proteínas Repressoras/genética
2.
Mol Cell Biol ; 40(1)2019 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-31658995

RESUMO

Mycophenolic acid (MPA) is the active metabolite of mycophenolate mofetil, a drug that is widely used for immunosuppression in organ transplantation and autoimmune diseases, as well as anticancer chemotherapy. It inhibits IMP dehydrogenase, a rate-limiting enzyme in de novo synthesis of guanidine nucleotides. MPA treatment interferes with transcription elongation, resulting in a drastic reduction of pre-rRNA and pre-tRNA synthesis, the disruption of the nucleolus, and consequently cell cycle arrest. Here, we investigated the mechanism whereby MPA inhibits RNA polymerase III (Pol III) activity, in both yeast and mammalian cells. We show that MPA rapidly inhibits Pol III by depleting GTP. Although MPA treatment can activate p53, this is not required for Pol III transcriptional inhibition. The Pol III repressor MAF1 is also not responsible for inhibiting Pol III in response to MPA treatment. We show that upon MPA treatment, the levels of selected Pol III subunits decrease, but this is secondary to transcriptional inhibition. Chromatin immunoprecipitation (ChIP) experiments show that Pol III does not fully dissociate from tRNA genes in yeast treated with MPA, even though there is a sharp decrease in the levels of newly transcribed tRNAs. We propose that in yeast, GTP depletion may lead to Pol III stalling.


Assuntos
Inibidores Enzimáticos/farmacologia , Imunossupressores/farmacologia , Ácido Micofenólico/farmacologia , RNA Polimerase III/antagonistas & inibidores , RNA de Transferência/genética , Transcrição Gênica/efeitos dos fármacos , Animais , Regulação Fúngica da Expressão Gênica/efeitos dos fármacos , Camundongos , Células RAW 264.7 , RNA Polimerase III/metabolismo , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/antagonistas & inibidores , Proteínas de Saccharomyces cerevisiae/metabolismo
3.
Nucleic Acids Res ; 47(8): 3937-3956, 2019 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-30820548

RESUMO

RNA polymerase (pol) III occurs in two forms, containing either the POLR3G subunit or the related paralogue POLR3GL. Whereas POLR3GL is ubiquitous, POLR3G is enriched in undifferentiated cells. Depletion of POLR3G selectively triggers proliferative arrest and differentiation of prostate cancer cells, responses not elicited when POLR3GL is depleted. A small molecule pol III inhibitor can cause POLR3G depletion, induce similar differentiation and suppress proliferation and viability of cancer cells. This response involves control of the fate-determining factor NANOG by small RNAs derived from Alu short interspersed nuclear elements. Tumour initiating activity in vivo can be reduced by transient exposure to the pol III inhibitor. Untransformed prostate cells appear less sensitive than cancer cells to pol III depletion or inhibition, raising the possibility of a therapeutic window.


Assuntos
Antineoplásicos/farmacologia , Regulação Neoplásica da Expressão Gênica , Células-Tronco Neoplásicas/efeitos dos fármacos , Neoplasias da Próstata/tratamento farmacológico , RNA Polimerase III/genética , Bibliotecas de Moléculas Pequenas/farmacologia , Idoso , Elementos Alu/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Chaperona BiP do Retículo Endoplasmático , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Proteína Homeobox Nanog/genética , Proteína Homeobox Nanog/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Prostatectomia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Neoplasias da Próstata/cirurgia , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Polimerase III/antagonistas & inibidores , RNA Polimerase III/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
4.
J Virol ; 86(8): 4527-37, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22345436

RESUMO

We have used the RAW 264.7 murine macrophage-like cell line as a platform to characterize the recognition and early signaling response to recombinant adenoviral vectors (rAdV). Infection of RAW 264.7 cells triggers an early response (2 to 6 h postinfection) that includes phosphorylation of the interferon (IFN) response factor 3 (IRF3) transcription factor, upregulation of IRF3 primary response genes (interferon-stimulated gene 56 [ISG56], beta IFN [IFN-ß]), and subsequent type I IFN secondary signaling (STAT1/2 phosphorylation). Using short hairpin RNA (shRNA) lentiviral vectors, we show an essential role for Tank binding kinase 1 (TBK1) in this pathway. Data also support a role for STING (MITA) as an adaptor functioning in response to rAdV infection. Using UV/psoralen (Ps)-inactivated virus to block viral transcription, Ps-inactivated virus stimulated primary (IRF3) and secondary (STAT1/2) activation events to the same degree as untreated virus. IRF3 phosphorylation was not blocked in RAW 264.7 cells pretreated with the RNA polymerase III inhibitor ML60218. However, they were compromised in the type I IFN-dependent secondary response (phosphorylation of STAT1/STAT2). At 24 h postinfection, ML60218-treated cells were compromised in the overall antiviral response. Therefore, initial sensing of rAdV or viral DNA (vDNA) does not depend on viral template transcription, but ML60218 treatment influences cellular cascades required for an antiviral response to rAdV. Using overexpression or knockdown assays, we examined how four DNA sensors influence the antiviral response. Knockdown of DNA Activator of Interferon (DAI) and p204, the murine ortholog to IFI16, had minimal influence on IRF3 phosphorylation. However, knockdown of absent in melanoma 2 (AIM2) and the helicase DDX41 resulted in diminished levels of (pser388)IRF3 following rAdV infection. Based on these data, multiple DNA sensors contribute to an antiviral DNA recognition response, leading to TBK1-dependent IRF3 phosphorylation in RAW 264.7 cells.


Assuntos
Adenoviridae/fisiologia , Fator Regulador 3 de Interferon/metabolismo , Macrófagos/imunologia , Macrófagos/virologia , Adenoviridae/genética , Adenoviridae/metabolismo , Adenovírus Humanos/genética , Adenovírus Humanos/metabolismo , Animais , Linhagem Celular , RNA Helicases DEAD-box , DNA Helicases/metabolismo , Proteínas de Ligação a DNA , Inibidores Enzimáticos/farmacologia , Inativação Gênica , Genoma Viral , Glicoproteínas/metabolismo , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Modelos Biológicos , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , RNA Polimerase III/antagonistas & inibidores , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica
5.
Invest New Drugs ; 30(1): 316-26, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20839029

RESUMO

BACKGROUND: TAS-106 is a novel nucleoside analog that inhibits RNA polymerases I, II and II and has demonstrated robust antitumor activity in a wide range of models of human cancer in preclinical studies. This study was performed to principally evaluate the feasibility of administering TAS-106 as a bolus intravenous (IV) infusion every 3 weeks. PATIENTS AND METHODS: Patients with advanced solid malignancies were treated with escalating doses of TAS-106 as a single bolus IV infusion every 3 weeks. Plasma and urine sampling were performed during the first course to characterize the pharmacokinetic profile of TAS-106 and assess pharmacodynamic relationships. RESULTS: Thirty patients were treated with 66 courses of TAS-106 at eight dose levels ranging from 0.67-9.46 mg/m(2). A cumulative sensory peripheral neuropathy was the principal dose-limiting toxicity (DLT) of TAS-106 at the 6.31 mg/m(2) dose level, which was determined to be the maximum tolerated dose (MTD). Other mild-moderate drug-related toxicities include asthenia, anorexia, nausea, vomiting, myelosuppression, and dermatologic effects. Major objective antitumor responses were not observed. The pharmacokinetics of TAS-106 were dose-proportional. The terminal elimination half-life (t(1/2)) averaged 11.3 ± 3.3 h. Approximately 71% of TAS-106 was excreted in the urine as unchanged drug. Pharmacodynamic relationships were observed between neuropathy and: C(5min;) AUC(0-inf;) and dermatologic toxicity. CONCLUSIONS: The recommended phase II dose of TAS-106 is 4.21 mg/m(2). However, due to a cumulative drug-related peripheral sensory neuropathy that proved to be dose-limiting, further evaluation of this bolus every 21 day infusion schedule will not be pursued and instead, an alternate dosing schedule of TAS-106 administered as a continuous 24-hour infusion will be explored to decrease C(max) in efforts to minimize peripheral neuropathy and maximize antitumor activity.


Assuntos
Antineoplásicos/administração & dosagem , Citidina/análogos & derivados , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/farmacocinética , Neoplasias/tratamento farmacológico , RNA Polimerase III/antagonistas & inibidores , RNA Polimerase II/antagonistas & inibidores , RNA Polimerase I/antagonistas & inibidores , Adulto , Idoso , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Área Sob a Curva , Biotransformação , Citidina/administração & dosagem , Citidina/efeitos adversos , Citidina/farmacocinética , Esquema de Medicação , Inibidores Enzimáticos/efeitos adversos , Estudos de Viabilidade , Feminino , Meia-Vida , Humanos , Infusões Intravenosas , Estimativa de Kaplan-Meier , Masculino , Dose Máxima Tolerável , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/patologia , RNA Polimerase I/metabolismo , RNA Polimerase II/metabolismo , RNA Polimerase III/metabolismo , Texas , Resultado do Tratamento
6.
J Mol Biol ; 387(3): 523-31, 2009 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-19361418

RESUMO

RNA polymerase III (RNA pol III) transcribes structural RNAs involved in RNA processing (U6 snRNA) and translation (tRNA), thereby regulating the growth rate of cells. Proper initiation by RNA pol III requires the transcription factor TFIIIB. Gene-external U6 snRNA transcription requires TFIIIB consisting of Bdp1, TBP, and Brf2. Transcription from the gene-internal tRNA promoter requires TFIIIB composed of Bdp1, TBP, and Brf1. TFIIIB is a target of tumor suppressors, including PTEN, ARF, p53, and RB, and RB-related pocket proteins. Breast cancer susceptibility gene 1 (BRCA1) tumor suppressor plays a role in DNA repair, cell cycle regulation, apoptosis, genome integrity, and ubiquitination. BRCA1 has a conserved amino-terminal RING domain, an activation domain 1 (AD1), and an acidic carboxyl-terminal domain (BRCA1 C-terminal region). In Saccharomyces cerevisiae, TFIIB interacts with the BRCA1 C-terminal region domain of Fcp1p, an RNA polymerase II phosphatase. The TFIIIB subunits Brf1 and Brf2 are structurally similar to TFIIB. Hence, we hypothesize that RNA pol III may be regulated by BRCA1 via the TFIIB family members Brf1 and Brf2. Here we report that: (1) BRCA1 inhibits both VAI (tRNA) and U6 snRNA RNA pol III transcription; (2) the AD1 of BRCA1 is responsible for inhibition of U6 snRNA transcription, whereas the RING domain and AD1 of BRCA1 are required for VAI transcription inhibition; and (3) overexpression of Brf1 and Brf2 alleviates inhibition of U6 snRNA and VAI transcription by BRCA1. Taken together, these data suggest that BRCA1 is a general repressor of RNA pol III transcription.


Assuntos
Proteína BRCA1/metabolismo , RNA Polimerase III/antagonistas & inibidores , RNA Polimerase III/metabolismo , Fator de Transcrição TFIIIB/metabolismo , Transcrição Gênica , Idoso , Proteína BRCA1/genética , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular , Feminino , Humanos , RNA Polimerase III/genética , RNA Nuclear Pequeno/genética , RNA Nuclear Pequeno/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Fator de Transcrição TFIIIB/genética
7.
Nucleic Acids Res ; 35(9): 3046-52, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17439968

RESUMO

The tumour suppressor protein ARF provides a defence mechanism against hyperproliferative stresses that can result from the aberrant activation of oncogenes. Accordingly, ARF is silenced or deleted in many human cancers. Activation of ARF can arrest growth and cell cycle progression, or trigger apoptosis. A principle mediator of these effects is p53, which ARF stabilizes by binding and inhibiting MDM2. However, ARF has additional targets and remains able to block growth in the absence of p53, albeit less efficiently. For example, ARF can suppress rRNA production in a p53-independent manner. We have found that the synthesis of tRNA by RNA polymerase III is also inhibited in response to ARF. However, in contrast to its effects on rRNA synthesis, ARF is unable to inhibit tRNA gene transcription when p53 is ablated. These results add to the growing list of cellular changes that can be triggered by ARF induction.


Assuntos
RNA Polimerase III/antagonistas & inibidores , Proteínas Repressoras/metabolismo , Proteína Supressora de Tumor p14ARF/metabolismo , Linhagem Celular , Fator de Transcrição E2F1/metabolismo , Humanos , Transcrição Gênica , Proteína Supressora de Tumor p53/fisiologia
8.
Proc Natl Acad Sci U S A ; 103(41): 15044-9, 2006 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-17005718

RESUMO

Maf1 is an essential and specific mediator of transcriptional repression in the RNA polymerase (pol) III system. Maf1-dependent repression occurs in response to a wide range of conditions, suggesting that the protein itself is targeted by the major nutritional and stress-signaling pathways. We show that Maf1 is a substrate for cAMP-dependent PKA in vitro and is differentially phosphorylated on PKA sites in vivo under normal versus repressing conditions. PKA activity negatively regulates Maf1 function because strains with unregulated high PKA activity block repression of pol III transcription in vivo, and strains lacking all PKA activity are hyperrepressible. Nuclear accumulation of Maf1 is required for transcriptional repression and is regulated by two nuclear localization sequences in the protein. An analysis of PKA phosphosite mutants shows that the localization of Maf1 is affected via the N-terminal nuclear localization sequence. In particular, mutations that prevent phosphorylation at PKA consensus sites promote nuclear accumulation of Maf1 without inducing repression. These results indicate that negative regulation of Maf1 by PKA is achieved by inhibiting its nuclear import and suggest that a PKA-independent activation step is required for nuclear Maf1 to function in the repression of pol III transcription. Finally, we report a previously undescribed phenotype for Maf1 in tRNA gene-mediated silencing of nearby RNA pol II transcription.


Assuntos
Núcleo Celular/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/química , RNA Polimerase III/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica/fisiologia , Transporte Ativo do Núcleo Celular/genética , Núcleo Celular/genética , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Repressão Enzimática/genética , Sinais de Localização Nuclear/química , Sinais de Localização Nuclear/genética , RNA Polimerase III/antagonistas & inibidores , RNA Polimerase III/biossíntese , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Fatores de Transcrição/genética
9.
Nucleic Acids Res ; 34(1): 286-94, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16407335

RESUMO

RNA polymerase (pol) III transcription decreases when primary cultures of rat neonatal cardiomyocytes are exposed to low oxygen tension. Previous studies in fibroblasts have shown that the pol III-specific transcription factor IIIB (TFIIIB) is bound and regulated by the proto-oncogene product c-Myc, the mitogen-activated protein kinase ERK and the retinoblastoma tumour suppressor protein, RB. The principal function of TFIIIB is to recruit pol III to its cognate gene template, an activity that is known to be inhibited by RB and stimulated by ERK. We demonstrate by chromatin immunoprecipitation (ChIP) that c-Myc also stimulates pol III recruitment by TFIIIB. However, hypoxic conditions cause TFIIIB dissociation from c-Myc and ERK, at the same time as increasing its interaction with RB. Consistent with this, ChIP assays indicate that the occupancy of tRNA genes by pol III is significantly reduced, whereas promoter binding by TFIIIB is undiminished. The data suggest that hypoxia can inhibit pol III transcription by altering the interactions between TFIIIB and its regulators and thus compromising its ability to recruit the polymerase. These effects are independent of cell cycle changes.


Assuntos
Regulação da Expressão Gênica , Miócitos Cardíacos/metabolismo , RNA Polimerase III/antagonistas & inibidores , RNA de Transferência/genética , Transcrição Gênica , Animais , Hipóxia Celular , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Polimerase III/metabolismo , Ratos , Ratos Sprague-Dawley , Proteína do Retinoblastoma/metabolismo
10.
Mol Cell Biol ; 25(8): 3247-60, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15798209

RESUMO

Human U1 and U6 snRNA genes are transcribed by RNA polymerases II and III, respectively. While the p53 tumor suppressor protein is a general repressor of RNA polymerase III transcription, whether p53 regulates snRNA gene transcription by RNA polymerase II is uncertain. The data presented herein indicate that p53 is an effective repressor of snRNA gene transcription by both polymerases. Both U1 and U6 transcription in vitro is repressed by recombinant p53, and endogenous p53 occupancy at these promoters is stimulated by UV light. In response to UV light, U1 and U6 transcription is strongly repressed. Human U1 genes, but not U6 genes, contain a high-affinity p53 response element located within the core promoter region. Nonetheless, this element is not required for p53 repression and mutant p53 molecules that do not bind DNA can maintain repression, suggesting a reliance on protein interactions for p53 promoter recruitment. Recruitment may be mediated by the general transcription factors TATA-box binding protein and snRNA-activating protein complex, which interact well with p53 and function for both RNA polymerase II and III transcription.


Assuntos
RNA Polimerase III/antagonistas & inibidores , RNA Polimerase II/antagonistas & inibidores , RNA Nuclear Pequeno/metabolismo , Proteínas Repressoras/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Regulação para Baixo , Humanos , Regiões Promotoras Genéticas/genética , RNA Polimerase II/fisiologia , RNA Polimerase III/fisiologia , RNA Nuclear Pequeno/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Elementos de Resposta/genética , Proteína de Ligação a TATA-Box/fisiologia , Transcrição Gênica/efeitos da radiação , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/fisiologia , Raios Ultravioleta
11.
Eur J Cancer ; 40(1): 21-7, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14687785

RESUMO

This review provides a summary of the European Association for Cancer Research Award Lecture, presented at the ECCO12 meeting in Copenhagen in September 2003. It describes what we have learnt about the mechanisms responsible for deregulating RNA polymerase III transcription in transformed cells. A network has been discovered of unanticipated links to key tumour suppressors and oncogenes. Novel functions have been revealed for RB, p53 and c-Myc, that may help explain their profound biological effects.


Assuntos
Neoplasias/genética , RNA Polimerase III/genética , Transcrição Gênica , Divisão Celular , Tamanho Celular , Genes Supressores de Tumor , Humanos , Neoplasias/enzimologia , Oncogenes , RNA Polimerase III/antagonistas & inibidores , Proteína do Retinoblastoma/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo
12.
Cancer Lett ; 123(2): 199-205, 1998 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-9489489

RESUMO

The c-myc promoter has a unique characteristic showing both RNA polymerase II (pol II) and RNA polymerase III (pol III) activities. Previous studies demonstrated that activating PKC results in upregulation of c-myc expression from its pol II promoter. However, how PKC activation affects expression from the pol III promoter of the c-myc gene is not well understood. This study examines the effect of PKC on the pol III transcription from the c-myc gene by using an in vitro system. We report the inhibition of the c-myc pol III transcript by activating PKC. Further, either a phosphocellulose fraction of HeLa whole cell extract (WCE) enriched for transcription factor TF IIIB, or recombinant TATA-box binding protein could restore the inhibited c-myc pol III transcription under conditions that activate PKC. A role has been proposed for the c-myc pol III transcript in the regulation of c-myc gene expression. Therefore, this report discusses the significance of the downregulation of c-myc expression from its pol III promoter and the possible interplay between the pol II and pol III promoters of this gene.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Genes myc/fisiologia , Proteína Quinase C/fisiologia , RNA Polimerase III/antagonistas & inibidores , Fracionamento Celular , Ativação Enzimática , Células HeLa , Humanos , Regiões Promotoras Genéticas , RNA Polimerase III/fisiologia , TATA Box/fisiologia , Acetato de Tetradecanoilforbol/farmacologia
13.
J Virol ; 71(3): 2577-80, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9032404

RESUMO

The HTLV-1 promoter directs RNA polymerase II transcription of viral genomic RNA in vivo. However, it has been reported that in vitro, a unique RNA polymerase, with characteristics of RNA polymerases II and III, is capable of HTLV-1 transcription (G. Piras, F. Kashanchi, M. F. Radonovich, J. F. Duvall, and J. N. Brady, J. Virol. 68:6170-6179, 1994). To further characterize the polymerase involved in HTLV-1 transcription in vitro, runoff transcription assays were performed with a variety of extracts and RNA polymerase inhibitors. Under all in vitro reaction conditions tested, RNA polymerase II appeared to be the only polymerase capable of correct transcriptional initiation from the HTLV-1 promoter. Synthesis of the specific HTLV-1 RNA transcript showed sensitivities to the RNA polymerase inhibitors tagetitoxin and alpha-amanitin that are consistent with RNA polymerase II transcription. Together, these data indicate that in vitro, as in vivo, the HTLV-1 promoter directs transcription by RNA polymerase II.


Assuntos
Amanitinas/farmacologia , Vírus Linfotrópico T Tipo 1 Humano/genética , RNA Polimerase II/antagonistas & inibidores , RNA Viral/biossíntese , Transcrição Gênica , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/farmacologia , Ácidos Dicarboxílicos/farmacologia , Produtos do Gene tax/farmacologia , Células HeLa , Humanos , Compostos Organofosforados/farmacologia , RNA Polimerase III/antagonistas & inibidores
15.
Nature ; 382(6586): 88-90, 1996 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-8657311

RESUMO

Transcription by RNA polymerase (pol) III is under cell-cycle control, being higher in S and G2 than in G0 and early G1 phases. Many transformed cell types have elevated pol III activity, presumably to sustain sufficient protein synthesis for unrestrained growth. The retinoblastoma tumour-suppressor protein (Rb) restricts cellular proliferation, and is often found mutated in transformed cells. Here we demonstrate that Rb can repress the level of transcription from pol III templates both in vitro and vivo. Analysis of Rb-deficient SAOS2 cells and primary fibroblasts from Rb-/- mice demonstrates elevated levels of pol III activity in the absence of functional Rb protein. Rb-induced repression of pol III activity is alleviated by mutations in the Rb pocket domain that occur naturally in tumours, and by viral transforming proteins that bind and inactivate Rb. These results implicate repression of pol III transcription as a mechanism for Rb-induced growth arrest, and suggest that restraining protein biosynthesis may be important in the prevention of tumour development.


Assuntos
Regulação da Expressão Gênica/fisiologia , RNA Polimerase III/metabolismo , Proteínas Repressoras/metabolismo , Proteína do Retinoblastoma/fisiologia , Proteínas E1A de Adenovirus/genética , Animais , Sequência de Bases , Células Cultivadas , Primers do DNA , Fibroblastos , Camundongos , Dados de Sequência Molecular , Mutação , RNA Polimerase III/antagonistas & inibidores , RNA Viral/genética , Proteína do Retinoblastoma/genética , Moldes Genéticos , Transcrição Gênica , Células Tumorais Cultivadas
16.
Carcinogenesis ; 16(5): 1017-21, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7767959

RESUMO

We have previously shown that stilbene estrogen (diethyl-stilbestrol, DES) covalently binds to nonhistone nuclear proteins both in vivo and in vitro. In this study, we demonstrate the differential effects of DES exposure on in organelle transcriptional activity in nuclei isolated from kidney (target organ of cancer) and liver (non target organ) of hamsters. Kidney RNA polymerase (RNA pol) I and III activities were significantly inhibited by 50% at days 8 and 15 of DES exposure compared to that of controls. Liver RNA pol I and III activities were only modestly inhibited (17 and 22%, respectively) by 2 and 8 days of DES exposure, respectively. However, longer exposure of DES to animals did not produce any significant effects on RNA pol I activity. The activity of RNA pol II was affected by DES exposure in both liver and kidney. DES treatment for two days resulted in an increase in RNA pol II activity in kidney. The enhanced enzyme activity was decreased to 50% of that of the control at 15 days of DES treatment. Unlike RNA pol I and III, RNA pol II activity in the liver was inhibited in a time-dependent fashion in response to DES exposure. To understand the mechanism of transcriptional inhibition by DES, we analyzed the effect of DES exposure on the expression of hepatic RNA pol II at both mRNA and protein levels and also phosphorylation of hepatic RNA pol II. The total amount of transcripts or protein contents of hepatic RNA pol II was not altered in response to DES exposure to hamster for 15 days. Total phosphorylation of hepatic RNA pol II was also not affected by 15 days DES exposure. However tyrosine phosphorylation of hepatic RNA pol II was lowered by 2.8-fold compared to that of control enzyme in response to DES exposure for 15 days. An inhibitory effect of DES on the total RNA polymerase activity in both kidney and liver nuclei in the presence of endogenous template was observed in vitro. No inhibitory effect of DES was observed in vitro on transcriptional activity in the presence of exogenously added DNA template. Based on these data it appears that the in vivo inhibition of transcription by DES may be due to alterations in chromatin template or the level of transcription regulating proteins and not due to decreased availability of the chromatin template and/or RNA polymerase. Whether DES related inhibition of transcriptional activity plays a role in the development of kidney cancer is not clear.


Assuntos
Dietilestilbestrol/farmacologia , Rim/enzimologia , Fígado/enzimologia , RNA Polimerase III/metabolismo , RNA Polimerase II/metabolismo , RNA Polimerase I/metabolismo , Transcrição Gênica/efeitos dos fármacos , Animais , Northern Blotting , Cricetinae , Rim/efeitos dos fármacos , Fígado/efeitos dos fármacos , Masculino , Mesocricetus , Especificidade de Órgãos , Fosforilação , RNA Polimerase I/antagonistas & inibidores , RNA Polimerase I/isolamento & purificação , RNA Polimerase II/antagonistas & inibidores , RNA Polimerase II/isolamento & purificação , RNA Polimerase III/antagonistas & inibidores , RNA Polimerase III/isolamento & purificação , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Valores de Referência , Fatores de Tempo
18.
J Virol ; 67(7): 4195-204, 1993 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8510221

RESUMO

Adenovirus E1A encodes two major proteins of 289 and 243 amino acids (289R and 243R), which both have transcription regulatory properties. E1A-289R is a transactivator whereas E1A-243R primarily functions as a repressor of transcription. Here we show that E1A repression is not restricted to RNA polymerase II genes but also includes the adenovirus virus-associated (VA) RNA genes. These genes are transcribed by RNA polymerase III and have previously been suggested to be the target of an E1A-289R-mediated transactivation. Surprisingly, we found that during transient transfection both E1A proteins repressed VA RNA transcription. E1A repression of VA RNA transcription required both conserved regions 1 and 2 and therefore differed from the E1A-mediated inhibition of simian virus 40 enhancer activity which primarily required conserved region 1. The repression was counteracted by the E1B-19K protein, which also, in the absence of E1A, enhanced the accumulation of VA RNA. Importantly, we show that efficient VA RNA transcription requires expression of both E1A and the E1B-19K protein during virus infection.


Assuntos
Proteínas E1A de Adenovirus/farmacologia , Regulação Viral da Expressão Gênica , RNA Polimerase III/antagonistas & inibidores , Transcrição Gênica/efeitos dos fármacos , Animais , Chlorocebus aethiops , Análise Mutacional de DNA , Células HeLa , Humanos , RNA Polimerase II/antagonistas & inibidores , RNA Mensageiro/genética , RNA Viral/genética , Proteínas Repressoras/genética , Relação Estrutura-Atividade , Ativação Transcricional
19.
J Clin Invest ; 91(6): 2665-72, 1993 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8390487

RESUMO

In this study, autoantibodies to RNA polymerase II from sera of patients with systemic sclerosis have been identified and characterized. These antibodies immunoprecipitated polypeptides of 220 kD (IIA) and 145 kD (IIC), the two largest subunits of RNA polymerase II, and bound both subunits in immunoblots. These polypeptides were immunoprecipitated by the anti-RNA polymerase II monoclonal antibody 8WG16, which recognizes the carboxyl-terminal domain of the 220-kD subunit, and their identity to the proteins bound by human sera was confirmed in immunodepletion studies. Sera with anti-RNA polymerase II antibodies also immunoprecipitated proteins that were consistent with components of RNA polymerases I and III. In vitro transcription experiments showed that the human antibodies were an effective inhibitor of RNA polymerase II activity. In indirect immunofluorescence studies, anti-RNA polymerase II autoantibodies stained the nucleoplasm, as expected from the known location of RNA polymerase II, and colocalized with the anti-RNA polymerase II monoclonal antibody. The human sera also stained the nucleolus, the location of RNA polymerase I. From a clinical perspective, these antibodies were found in 13 of 278 patients with systemic sclerosis, including 10 with diffuse and three with limited cutaneous disease, but were not detected in sera from patients with other connective tissue diseases and from normal controls. We conclude that anti-RNA polymerase II antibodies are specific to patients with systemic sclerosis, and that they are apparently associated with antibodies to RNA polymerases I and III. These autoantibodies may be useful diagnostically and as a probe for further studies of the biological function of RNA polymerases.


Assuntos
Autoanticorpos/isolamento & purificação , RNA Polimerase II/imunologia , Escleroderma Sistêmico/imunologia , Adenoviridae/genética , Sequência de Aminoácidos , Autoanticorpos/farmacologia , Citomegalovirus/genética , Imunofluorescência , Humanos , Dados de Sequência Molecular , Testes de Precipitina , Regiões Promotoras Genéticas/genética , RNA Polimerase I/antagonistas & inibidores , RNA Polimerase I/imunologia , RNA Polimerase II/antagonistas & inibidores , RNA Polimerase III/antagonistas & inibidores , RNA Polimerase III/imunologia , Escleroderma Sistêmico/enzimologia , Transcrição Gênica/efeitos dos fármacos
20.
J Biochem Toxicol ; 7(1): 13-7, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-1375294

RESUMO

Treatment of isolated rat liver nuclei with 7 beta, 8 alpha-dihydroxy-9 alpha, 10 alpha-epoxy-7,8,9,10- tetrahydrobenzo[a]pyrene, the ultimate carcinogenic metabolite of benzo[a]pyrene, resulted in inhibition of transcription as measured by radioactive precursor incorporation into RNA. The mechanism of inhibition as analyzed by use of different types of inhibitors suggested that the carcinogen acted on both the major components of transcription machinery, that is, the template chromatin and the enzyme RNA polymerases. This action correlates well with the observations made after administration of benzo[a]pyrene to rats.


Assuntos
7,8-Di-Hidro-7,8-Di-Hidroxibenzo(a)pireno 9,10-óxido/toxicidade , Fígado/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Animais , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Dactinomicina/farmacologia , Dimetil Sulfóxido/farmacologia , Técnicas In Vitro , Fígado/metabolismo , RNA/biossíntese , RNA Polimerase I/antagonistas & inibidores , RNA Polimerase II/antagonistas & inibidores , RNA Polimerase III/antagonistas & inibidores , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA