Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 13(1): 5453, 2022 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-36114190

RESUMO

Survival of motor neuron (SMN) functions in diverse biological pathways via recognition of symmetric dimethylarginine (Rme2s) on proteins by its Tudor domain, and deficiency of SMN leads to spinal muscular atrophy. Here we report a potent and selective antagonist with a 4-iminopyridine scaffold targeting the Tudor domain of SMN. Our structural and mutagenesis studies indicate that both the aromatic ring and imino groups of compound 1 contribute to its selective binding to SMN. Various on-target engagement assays support that compound 1 specifically recognizes SMN in a cellular context and prevents the interaction of SMN with the R1810me2s of RNA polymerase II subunit POLR2A, resulting in transcription termination and R-loop accumulation mimicking SMN depletion. Thus, in addition to the antisense, RNAi and CRISPR/Cas9 techniques, potent SMN antagonists could be used as an efficient tool to understand the biological functions of SMN.


Assuntos
RNA Polimerase II , Proteínas do Complexo SMN , Humanos , Neurônios Motores/metabolismo , Atrofia Muscular Espinal/metabolismo , RNA Polimerase II/efeitos dos fármacos , RNA Polimerase II/metabolismo , Proteínas do Complexo SMN/antagonistas & inibidores , Proteínas do Complexo SMN/efeitos dos fármacos , Proteínas do Complexo SMN/metabolismo
2.
Lipids Health Dis ; 19(1): 144, 2020 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-32563265

RESUMO

BACKGROUND: Moderate alcohol intake in human increases HDL-cholesterol, and has protective effects against cardiovascular disease (CVD). Although de novo lipid synthesis inhibitors are highly effective in lowering total and LDL-cholesterol they have only modest effects on raising HDL-C. A better understanding of the mechanism of ethanol-mediated HDL-C regulation could suggest new therapeutic approaches for CVD. METHODS: Human hepatoblastoma (HepG2) and colorectal epithelial adenocarcinoma (Caco-2) cells were incubated in the presence of varying concentrations of ethanol in the culture medium, with or without addition of de novo lipid synthesis (DNLS) inhibitors (atorvastatin and/or TOFA). ApoA1 protein was measured by Western blot, and RNA of lipid pathway genes APOA1, APOC3, APOA4, APOB100, HMGCR, LDLR, and SREBF2 by quantitative RT-PCR. Lipoproteins (VLDL, LDL, and HDL) and lipids were also monitored. RESULTS: Ethanol stimulated ApoA1 protein (both cytoplasmic and secreted) and APOA1 RNA levels in HepG2 cells in a dose sensitive way, with ~ 50% upregulation at 100 mM ethanol in the medium. The effect was not observed in intestinal-derived Caco-2 cells. DNLS inhibitors did not block the upregulation of ApoA1 RNA by ethanol; TOFA alone produced a modest increase in ApoA1 RNA. Ethanol had no effect on ABCA1 protein levels. Addition of ethanol to the cell medium led to modest increases in de novo synthesis of total cholesterol, cholesteryl esters and triglycerides, and as expected these increases were blocked when the lipid synthesis inhibitors were added. Ethanol stimulated a small increase in HDL and VLDL but not LDL synthesis. Ethanol in the cell medium also induced modest but measurable increases in the RNA of APOC3, APOA4, APOB, LDLR, and HMGCR genes. Unlike APOA1, induction of RNA from APOC3 and APOA4 was also observed in Caco-2 cells as well as HepG2 cells. CONCLUSION: This study has verified the previously reported upregulation of APOA1 by exposure of HepG2, but not Caco-2 cells, to ethanol in the culture medium. It is shown for the first time that the effect is dependent on RNA polymerase II-mediated transcription, but not on de novo biosynthesis of cholesterol or fatty acids, and therefore is not a generalized metabolic response to ethanol exposure. Some other lipid pathway genes are also modulated by ethanol exposure of cells. The results reported here suggest that the proximal signaling molecule leading to increased APOA1 gene expression in response to ethanol exposure may be free acetate or acetyl-CoA. TAKE HOME: Upregulation of ApoA1 gene expression in hepatoma cells in culture, upon exposure to moderate ethanol concentrations in the medium, occurs at the level of RNA and is not dependent on new cholesterol or fatty acid synthesis. The primary signaling molecule may be free acetate or acetyl-CoA. These results are important for understanding the mechanism by which moderate alcohol consumption leads to upregulation of serum HDL-cholesterol in humans, and suggests new approaches to targeting HDL as a risk factor for cardiovascular disease.


Assuntos
Consumo de Bebidas Alcoólicas/genética , Apolipoproteína A-I/genética , Doenças Cardiovasculares/genética , HDL-Colesterol/genética , Apolipoproteína C-III/genética , Células CACO-2 , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/prevenção & controle , HDL-Colesterol/biossíntese , HDL-Colesterol/efeitos dos fármacos , LDL-Colesterol/genética , Etanol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células Hep G2 , Humanos , Lipídeos/biossíntese , Lipídeos/genética , RNA Polimerase II/efeitos dos fármacos , Proteína de Ligação a Elemento Regulador de Esterol 2/genética
3.
Int J Mol Sci ; 21(9)2020 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-32397434

RESUMO

Inhibitors targeting the general RNA polymerase II (RNAPII) transcription machinery are candidate therapeutics in cancer and other complex diseases. Here, we review the molecular targets and mechanisms of action of these compounds, framing them within the steps of RNAPII transcription. We discuss the effects of transcription inhibitors in vitro and in cellular models (with an emphasis on cancer), as well as their efficacy in preclinical and clinical studies. We also discuss the rationale for inhibiting broadly acting transcriptional regulators or RNAPII itself in complex diseases.


Assuntos
Antineoplásicos/farmacologia , Quinases Ciclina-Dependentes/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Terapia de Alvo Molecular , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias/tratamento farmacológico , RNA Polimerase II/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Catálise/efeitos dos fármacos , Ensaios Clínicos como Assunto , Quinase 8 Dependente de Ciclina/antagonistas & inibidores , Quinase 9 Dependente de Ciclina/antagonistas & inibidores , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Proteínas de Neoplasias/fisiologia , Neoplasias/enzimologia , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , RNA Polimerase II/fisiologia
4.
Nat Commun ; 10(1): 5444, 2019 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-31784510

RESUMO

Elevated glucose consumption is fundamental to cancer, but selectively targeting this pathway is challenging. We develop a high-throughput assay for measuring glucose consumption and use it to screen non-small-cell lung cancer cell lines against bioactive small molecules. We identify Milciclib that blocks glucose consumption in H460 and H1975, but not in HCC827 or A549 cells, by decreasing SLC2A1 (GLUT1) mRNA and protein levels and by inhibiting glucose transport. Milciclib blocks glucose consumption by targeting cyclin-dependent kinase 7 (CDK7) similar to other CDK7 inhibitors including THZ1 and LDC4297. Enhanced PIK3CA signaling leads to CDK7 phosphorylation, which promotes RNA Polymerase II phosphorylation and transcription. Milciclib, THZ1, and LDC4297 lead to a reduction in RNA Polymerase II phosphorylation on the SLC2A1 promoter. These data indicate that our high-throughput assay can identify compounds that regulate glucose consumption and that CDK7 is a key regulator of glucose consumption in cells with an activated PI3K pathway.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Quinases Ciclina-Dependentes/antagonistas & inibidores , Transportador de Glucose Tipo 1/efeitos dos fármacos , Glucose/metabolismo , Neoplasias Pulmonares/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Células A549 , Linhagem Celular Tumoral , Classe I de Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Ensaios de Triagem em Larga Escala , Humanos , Fenilenodiaminas/farmacologia , Fosforilação , Pirazóis/farmacologia , Pirimidinas/farmacologia , Quinazolinas/farmacologia , RNA Polimerase II/efeitos dos fármacos , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , RNA Mensageiro/metabolismo , Transdução de Sinais , Triazinas/farmacologia , Quinase Ativadora de Quinase Dependente de Ciclina
5.
Nat Rev Genet ; 19(6): 385-397, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29556092

RESUMO

Proteotoxic stress, that is, stress caused by protein misfolding and aggregation, triggers the rapid and global reprogramming of transcription at genes and enhancers. Genome-wide assays that track transcriptionally engaged RNA polymerase II (Pol II) at nucleotide resolution have provided key insights into the underlying molecular mechanisms that regulate transcriptional responses to stress. In addition, recent kinetic analyses of transcriptional control under heat stress have shown how cells 'prewire' and rapidly execute genome-wide changes in transcription while concurrently becoming poised for recovery. The regulation of Pol II at genes and enhancers in response to heat stress is coupled to chromatin modification and compartmentalization, as well as to co-transcriptional RNA processing. These mechanistic features seem to apply broadly to other coordinated genome-regulatory responses.


Assuntos
Cromatina/metabolismo , Regulação da Expressão Gênica , Resposta ao Choque Térmico , RNA Polimerase II/metabolismo , Transcrição Gênica , Animais , Cromatina/genética , Elementos Facilitadores Genéticos , Humanos , Regiões Promotoras Genéticas , RNA Polimerase II/efeitos dos fármacos
6.
Clin Immunol ; 159(1): 13-22, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25926428

RESUMO

We showed previously that 17ß estradiol (E2) led to improved survival in nephrotoxic serum induced nephritis (NTN) in male mice. In this study we determined whether E2 regulates vascular cell adhesion molecule (VCAM)-1, an adhesion molecule that is upregulated in kidney during autoimmune nephritis, in mesangial cells (MC). We show that E2 inhibited VCAM-1 up-regulation in kidneys in vivo during NTN, and in MCs upon TNFα stimulation. VCAM-1 up-regulation in MCs was controlled by the transcription factor NFκB. E2 inhibited RNA polymerase II recruitment to the VCAM-1 promoter, but not p65 recruitment. Interestingly E2 inhibited TNFα stimulated interaction between poly (ADP-ribose) polymerase-1 (PARP-1) and p65. As PARP-1 is required for VCAM-1 upregulation in MCs, our data suggest that E2 may inhibit pre-initiation complex formation at VCAM-1 promoter by inhibiting PARP-1 recruitment to p65. We propose that E2 plays an important role in regulating renal inflammation locally.


Assuntos
Estradiol/farmacologia , Estrogênios/farmacologia , Glomerulonefrite , Células Mesangiais/efeitos dos fármacos , RNA Mensageiro/metabolismo , Molécula 1 de Adesão de Célula Vascular/efeitos dos fármacos , Animais , Células Cultivadas , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Rim/efeitos dos fármacos , Rim/metabolismo , Células Mesangiais/metabolismo , Camundongos , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/efeitos dos fármacos , Poli(ADP-Ribose) Polimerases/metabolismo , RNA Polimerase II/efeitos dos fármacos , RNA Polimerase II/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição RelA/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo , Regulação para Cima , Molécula 1 de Adesão de Célula Vascular/genética
7.
Cancer Res ; 72(6): 1529-37, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22282656

RESUMO

The androgen receptor (AR) signaling axis plays a key role in the pathogenesis of prostate cancer. In this study, we found that the protein tyrosine phosphatase PTP1B, a well-established regulator of metabolic signaling, was induced after androgen stimulation of AR-expressing prostate cancer cells. PTP1B induction by androgen occurred at the mRNA and protein levels to increase PTP1B activity. High-resolution chromosome mapping revealed AR recruitment to two response elements within the first intron of the PTP1B encoding gene PTPN1, correlating with an AR-mediated increase in RNA polymerase II recruitment to the PTPN1 transcriptional start site. We found that PTPN1 and AR genes were coamplified in metastatic tumors and that PTPN1 amplification was associated with a subset of high-risk primary tumors. Functionally, PTP1B depletion delayed the growth of androgen-dependent human prostate tumors and impaired androgen-induced cell migration and invasion in vitro. However, PTP1B was also required for optimal cell migration of androgen-independent cells. Collectively, our results established the AR as a transcriptional regulator of PTPN1 transcription and implicated PTP1B in a tumor-promoting role in prostate cancer. Our findings support the preclinical testing of PTP1B inhibitors for prostate cancer treatment.


Assuntos
Progressão da Doença , Neoplasias da Próstata/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 1/genética , Receptores Androgênicos/metabolismo , Androgênios/farmacologia , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Íntrons/efeitos dos fármacos , Masculino , Camundongos , Camundongos SCID , Neoplasias da Próstata/tratamento farmacológico , RNA Polimerase II/efeitos dos fármacos , RNA Polimerase II/metabolismo , Receptores Androgênicos/genética , Elementos de Resposta , Sítio de Iniciação de Transcrição/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Leuk Lymphoma ; 52(10): 1999-2006, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21728742

RESUMO

Flavopiridol is a cyclin-dependent kinase inhibitor that induces cell cycle arrest, apoptosis, and clinical responses in selected patients with acute myeloid leukemia (AML). A better understanding of the molecular pathways targeted by flavopiridol is needed to design optimal combinatorial therapy. Here, we report that in vivo administration of flavopiridol induced expression of the BCL-2 anti-apoptotic gene in leukemic blasts from adult patients with refractory AML. Moreover, flavopiridol repressed the expression of genes encoding oncogenic transcription factors (HMGA1, STAT3, E2F1) and the major subunit of RNA Polymerase II. Our results provide mechanistic insight into the cellular pathways targeted by flavopiridol. Although further studies are needed, our findings also suggest that blocking anti-apoptotic pathways could enhance cytotoxicity with flavopiridol.


Assuntos
Flavonoides/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , Piperidinas/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Fatores de Transcrição/antagonistas & inibidores , Adulto , Antineoplásicos , Proteínas Reguladoras de Apoptose , Crise Blástica/tratamento farmacológico , Feminino , Flavonoides/administração & dosagem , Flavonoides/uso terapêutico , Humanos , Leucemia Mieloide Aguda/patologia , Masculino , Pessoa de Meia-Idade , Oncogenes , Piperidinas/administração & dosagem , Piperidinas/uso terapêutico , Inibidores de Proteínas Quinases , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , RNA Polimerase II/efeitos dos fármacos , Adulto Jovem
9.
Cancer Res ; 70(20): 8055-65, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20817775

RESUMO

RNA splicing is required to remove introns from pre-mRNA, and alternative splicing generates protein diversity. Topoisomerase I (Top1) has been shown to be coupled with splicing by regulating serine/arginine-rich splicing proteins. Prior studies on isolated genes also showed that Top1 poisoning by camptothecin (CPT), which traps Top1 cleavage complexes (Top1cc), can alter RNA splicing. Here, we tested the effect of Top1 inhibition on splicing at the genome-wide level in human colon carcinoma HCT116 and breast carcinoma MCF7 cells. The RNA of HCT116 cells treated with CPT for various times was analyzed with ExonHit Human Splice Array. Unlike other exon array platforms, the ExonHit arrays include junction probes that allow the detection of splice variants with high sensitivity and specificity. We report that CPT treatment preferentially affects the splicing of splicing-related factors, such as RBM8A, and generates transcripts coding for inactive proteins lacking key functional domains. The splicing alterations induced by CPT are not observed with cisplatin or vinblastine and are not simply due to reduced Top1 activity, as Top1 downregulation by short interfering RNA did not alter splicing like CPT treatment. Inhibition of RNA polymerase II (Pol II) hyperphosphorylation by 5,6-dichloro-1-ß-d-ribofuranosylbenzimidazole (DRB) blocked the splicing alteration induced by CPT, which suggests that the rapid Pol II hyperphosphorylation induced by CPT interferes with normal splicing. The preferential effect of CPT on genes encoding splicing factors may explain the abnormal splicing of a large number of genes in response to Top1cc.


Assuntos
DNA Topoisomerases Tipo I/intoxicação , Estudo de Associação Genômica Ampla/métodos , Processamento Alternativo/efeitos dos fármacos , Processamento Alternativo/genética , Antineoplásicos Fitogênicos/farmacologia , Camptotecina/farmacologia , Cisplatino/farmacologia , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , DNA Topoisomerases Tipo I/genética , Proteínas de Ligação a DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Regulação para Baixo/efeitos dos fármacos , Éxons/efeitos dos fármacos , Éxons/genética , Variação Genética , Humanos , Modelos Estatísticos , Fosforilação , RNA Polimerase II/efeitos dos fármacos , RNA Polimerase II/metabolismo , Splicing de RNA/genética , RNA Interferente Pequeno/efeitos dos fármacos , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vimblastina/farmacologia
10.
Mol Cell Biol ; 30(2): 399-412, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19917725

RESUMO

Activation of estrogen receptor alpha (ERalpha) results in both induction and repression of gene transcription; while mechanistic details of estrogen induction are well described, details of repression remain largely unknown. We characterized several ERalpha-repressed targets and examined in detail the mechanism for estrogen repression of Reprimo (RPRM), a cell cycle inhibitor. Estrogen repression of RPRM is rapid and robust and requires a tripartite interaction between ERalpha, histone deacetylase 7 (HDAC7), and FoxA1. HDAC7 is the critical HDAC needed for repression of RPRM; it can bind to ERalpha and represses ERalpha's transcriptional activity--this repression does not require HDAC7's deacetylase activity. We further show that the chromatin pioneer factor FoxA1, well known for its role in estrogen induction of genes, is recruited to the RPRM promoter, is necessary for repression of RPRM, and interacts with HDAC7. Like other FoxA1 recruitment sites, the RPRM promoter is characterized by H3K4me1/me2. Estrogen treatment causes decreases in H3K4me1/me2 and release of RNA polymerase II (Pol II) from the RPRM proximal promoter. Overall, these data implicate a novel role for HDAC7 and FoxA1 in estrogen repression of RPRM, a mechanism which could potentially be generalized to many more estrogen-repressed genes and hence be important in both normal physiology and pathological processes.


Assuntos
Proteínas de Ciclo Celular/genética , Receptor alfa de Estrogênio/metabolismo , Regulação da Expressão Gênica , Glicoproteínas/genética , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Histona Desacetilases/metabolismo , Proteínas de Ciclo Celular/antagonistas & inibidores , Linhagem Celular Tumoral , Metilação de DNA/efeitos dos fármacos , Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio/efeitos dos fármacos , Estrogênios/farmacologia , Fulvestranto , Glicoproteínas/antagonistas & inibidores , Fator 3-alfa Nuclear de Hepatócito/efeitos dos fármacos , Histona Desacetilases/efeitos dos fármacos , Histonas/efeitos dos fármacos , Histonas/metabolismo , Humanos , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/fisiologia , RNA Polimerase II/efeitos dos fármacos , RNA Polimerase II/metabolismo , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/fisiologia
11.
Cancer Res ; 68(13): 5257-66, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18593926

RESUMO

Triptolide, an active component of the medicinal herb lei gong teng, is a potent anticancer and anti-inflammatory therapeutic. It potently inhibits nuclear factor-kappaB transcriptional activation after DNA binding, although a precise mechanism is as yet unknown. Here, we report that triptolide also induces distinct nuclear substructural changes in HeLa cells. These changes in the nucleolus and nuclear speckles are reversible and dependent on both time and concentration. Furthermore, nuclear changes occurred within hours of triptolide treatment and were calcium and caspase independent. Rounding of nuclear speckles, an indication of transcriptional arrest, was evident and was associated with a decrease in RNA polymerase II (RNA Pol II) COOH-terminal domain Ser(2) phosphorylation. Additionally, the nucleolus disassembled and RNA Pol I activity declined after RNA Pol II inhibition. We therefore conclude that triptolide causes global transcriptional arrest as evidenced by inactivity of RNA Pol I and II and the subsequent alteration in nuclear substructure.


Assuntos
Núcleo Celular/efeitos dos fármacos , Núcleo Celular/ultraestrutura , Diterpenos/farmacologia , Fenantrenos/farmacologia , Transcrição Gênica/efeitos dos fármacos , Antineoplásicos Alquilantes/farmacologia , Cálcio/farmacologia , Caspases/metabolismo , Caspases/fisiologia , Núcleo Celular/metabolismo , Células Clonais , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/fisiologia , Compostos de Epóxi/farmacologia , Células HeLa , Humanos , Proteínas Nucleares/metabolismo , Nucleofosmina , Fosfoproteínas/metabolismo , Proteínas Pol1 do Complexo de Iniciação de Transcrição/metabolismo , RNA Polimerase II/efeitos dos fármacos , RNA Polimerase II/metabolismo , Proteínas de Ligação a RNA/metabolismo , Fatores de Tempo , Distribuição Tecidual , Nucleolina
12.
Pediatr Blood Cancer ; 50(4): 772-8, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18000861

RESUMO

BACKGROUND: New agents are needed for treatment of children with relapsed acute lymphoblastic leukemia (ALL). Based on altered expression of cell cycle regulatory proteins, including frequent p16 (INK4A) and p15 (INK4B) deletions, flavopiridol (FP; Alvocidib) is an attractive agent for relapsed ALL. PROCEDURE: We evaluated the efficacy of FP in ALL cell lines using cell proliferation assays, determined the effects of FP treatment on cell growth and viability in cell lines and patient samples, examined cell cycle kinetics, and evaluated the effect of FP on endogenous cyclin-dependent kinase (CDK) activity, Mcl-1 expression, and RNA polymerase II expression and phosphorylation. RESULTS: ALL cell lines are sensitive to FP. At lower concentrations, FP induces transient G(1)-S cell cycle arrest and modest levels of apoptosis in cell lines. In contrast, a sustained G(1)-S and G(2)-M arrest and substantial apoptosis are observed following exposure to higher FP concentrations. After treatment with FP, ALL cell lines have decreased expression of retinoblastoma protein phosphorylated at serines 795 and 807/811, indicating reduced CDK activity. We also show that ALL cell lines are sensitive to clinically achievable concentrations of FP in medium supplemented with human serum and that FP reduces the expression of Mcl-1 and phosphorylated forms of the C-terminal domain of RNA polymerase II. FP also increases cell death by approximately twofold over baseline in primary ALL blasts. CONCLUSIONS: These data provide a biological rationale for testing FP in relapsed ALL.


Assuntos
Antineoplásicos/farmacologia , Flavonoides/farmacologia , Piperidinas/farmacologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Apoptose/efeitos dos fármacos , Western Blotting , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Proteína de Sequência 1 de Leucemia de Células Mieloides , Proteínas de Neoplasias/efeitos dos fármacos , Recidiva Local de Neoplasia/tratamento farmacológico , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , RNA Polimerase II/efeitos dos fármacos , Proteína do Retinoblastoma/efeitos dos fármacos
13.
Mol Microbiol ; 63(4): 1237-47, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17257309

RESUMO

The Plasmodium falciparum var gene family codes for a major virulence factor in this most lethal of human malaria parasites. A single var protein variant type is expressed on each infected red blood cell, with antigenic variation allowing progeny parasites to escape host immune detection. The control of mutually exclusive var gene expression in the parasite relies on in situ epigenetic changes. Whether control of expression occurs at transcription initiation or post transcription, however, remains to be established. Recent evidence supports existence of a unique var transcription site at the nuclear periphery containing the dominantly expressed var gene, although silent var genes can colocalize to the same region. We demonstrate here that exclusive var gene expression is controlled at the level of transcription initiation during ring stages and that var genes are transcribed by RNA polymerase II. This represents another example where P. falciparum differs from the paradigm for antigenic variation, Trypanosoma brucei.


Assuntos
Plasmodium falciparum/genética , Proteínas de Protozoários/genética , RNA Polimerase II/metabolismo , Amanitinas/farmacologia , Animais , Regulação da Expressão Gênica , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/crescimento & desenvolvimento , Proteínas de Protozoários/metabolismo , RNA Polimerase II/efeitos dos fármacos , Transcrição Gênica
14.
Genes Cells ; 11(4): 373-81, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16611241

RESUMO

TFII-I was initially identified as the general transcription factor that binds to initiator (Inr) elements in vitro. Subsequent studies have shown that TFII-I activates transcription of various genes either through Inr elements or through other upstream elements in vivo. Since, however, most studies so far on TFII-I have been limited to over-expression and reporter gene assays, we reevaluated the role of TFII-I in vivo by using stable knockdown with siRNA and by examining the expression of endogenous genes. Contrary to the widely accepted view, here we show that TFII-I is not important for cell viability in general but rather inhibits the growth of MCF-7 human breast cancer cells. MCF-7 cells are known to proliferate in an estrogen-dependent manner. Through analysis of TFII-I's cell-type specific growth inhibitory effect, we show evidence that TFII-I down-regulates a subset of estrogen-responsive genes, only those containing Inr elements, by recruiting estrogen receptor (ER) alpha and corepressors to these promoters. Thus, this study has revealed an unexpected new role of TFII-I as a negative regulator of transcription and cell proliferation.


Assuntos
Receptor alfa de Estrogênio/fisiologia , Estrogênios/genética , Estrogênios/metabolismo , Regulação da Expressão Gênica/fisiologia , Elementos de Resposta/fisiologia , Fatores de Transcrição TFII/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Regulação para Baixo , Receptor alfa de Estrogênio/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Presenilina-2 , RNA Polimerase II/efeitos dos fármacos , RNA Polimerase II/metabolismo , RNA Interferente Pequeno/farmacologia , Elementos de Resposta/efeitos dos fármacos , Relação Estrutura-Atividade , Fatores de Transcrição TFII/antagonistas & inibidores , Fatores de Transcrição TFII/farmacologia , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética
15.
Mol Cancer Ther ; 5(1): 138-48, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16432172

RESUMO

Up-regulated signal transducers and activators of transcription (STAT)-mediated signaling is believed to contribute to the pathogenesis of a variety of solid and hematologic cancers. Consequently, inhibition of STAT-mediated signaling has recently been proposed as a potential new therapeutic approach to the treatment of cancers. Having shown previously that the pan-cyclin-dependent kinase inhibitor flavopiridol binds to DNA and seems to kill cancer cells via that process in some circumstances, we evaluated the hypothesis that flavopiridol might consequently disrupt STAT3/DNA interactions, attenuate STAT3-directed transcription, and down-regulate STAT3 downstream polypeptides, including the antiapoptotic polypeptide Mcl-1. SDS-PAGE/immunoblotting and reverse transcription-PCR were used to assess RNA and polypeptide levels, respectively. DNA cellulose affinity chromatography and a nuclear elution assay were used to evaluate the ability of flavopiridol to disrupt STAT3/DNA interactions. A STAT3 luciferase reporter assay was used to examine the ability of flavopiridol to attenuate STAT3-directed transcription. Colony-forming assays were used to assess cytotoxic synergy between flavopiridol and AG490. Flavopiridol was found to (a) disrupt STAT3/DNA interactions (DNA cellulose affinity chromatography and nuclear elution assay), (b) attenuate STAT3-directed transcription (STAT3 luciferase reporter assay), and (c) down-regulate the STAT3 downstream antiapoptotic polypeptide Mcl-1 at the transcriptional level (reverse transcription-PCR and SDS-PAGE/immunoblotting). Furthermore, flavopiridol, but not the microtubule inhibitor paclitaxel, could be combined with the STAT3 pathway inhibitor AG490 to achieve cytotoxic synergy in A549 human non-small cell lung cancer cells. Collectively, these data suggest that flavopiridol can attenuate STAT3-directed transcription in a targeted fashion and may therefore be exploitable clinically in the development of chemotherapy regimens combining flavopiridol and other inhibitors of STAT3 signaling pathways.


Assuntos
Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Piperidinas/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Fator de Transcrição STAT3/efeitos dos fármacos , Tirfostinas/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , DNA/metabolismo , Regulação para Baixo , Ensaios de Seleção de Medicamentos Antitumorais , Flavonoides/administração & dosagem , Humanos , Janus Quinase 1 , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Proteína de Sequência 1 de Leucemia de Células Mieloides , Proteínas de Neoplasias/efeitos dos fármacos , Proteínas de Neoplasias/genética , Fosfoproteínas/efeitos dos fármacos , Fosfoproteínas/metabolismo , Piperidinas/administração & dosagem , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Polimerase II/efeitos dos fármacos , RNA Polimerase II/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transcrição Gênica , Células Tumorais Cultivadas
16.
RNA ; 12(2): 256-62, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16373483

RESUMO

To determine mechanistically how siRNAs mediate transcriptional gene silencing (TGS) in human cells, we have measured histone methylation at targeted promoters, the dependency on active transcription, and whether or not both strands of the siRNA are required for siRNA-mediated TGS. We report here that siRNA treatment increases both H3K9 and H3K27 methylation of the targeted EF1A promoter and that this increase is dependent on nuclear specific delivery of the siRNA. We also find that TGS can be directed by the antisense strand alone, and requires active transcription by RNA polymerase II in human cells as evidenced by sensitivity to alpha-amanatin. The observation of antisense strand-specific siRNA-mediated TGS of EF1A was substantiated by targeting the U3 region of the HIV-1 LTR/promoter. Furthermore, we show that the antisense strand of siRNA EF52 associates with the transiently expressed Flag-tagged DNMT3A, the targeted EF1A promoter, and trimethylated H3K27. The observations reported here implicate a functional link between siRNA-mediated targeting of genomic regions (promoters), RNA Pol II function, histone methylation, and DNMT3A and support a paradigm in which the antisense strands of siRNAs alone can direct sequence-specific transcriptional gene silencing in human cells.


Assuntos
Histonas/metabolismo , Interferência de RNA , RNA Antissenso/metabolismo , RNA Interferente Pequeno/metabolismo , Amanitinas/farmacologia , Sequência de Bases , Células Cultivadas , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Metiltransferase 3A , Repetição Terminal Longa de HIV , Histonas/genética , Humanos , Metilação , Dados de Sequência Molecular , Proteínas Nucleares/genética , Inibidores da Síntese de Ácido Nucleico/farmacologia , Regiões Promotoras Genéticas , Interferência de RNA/efeitos dos fármacos , RNA Polimerase II/efeitos dos fármacos , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , RNA Antissenso/genética , RNA Interferente Pequeno/genética , Fatores de Transcrição/genética
17.
Cell ; 121(6): 913-23, 2005 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-15960978

RESUMO

In order to study mechanisms and regulation of RNA polymerase II (RNAPII) ubiquitylation and degradation, highly purified factors were used to reconstitute RNAPII ubiquitylation in vitro. We show that arrested RNAPII elongation complexes are the preferred substrates for ubiquitylation. Accordingly, not only DNA-damage-dependent but also DNA-damage-independent transcriptional arrest results in RNAPII ubiquitylation in vivo. Def1, known to be required for damage-induced degradation of RNAPII, stimulates ubiquitylation of RNAPII only in an elongation complex. Ubiquitylation of RNAPII is dependent on its C-terminal repeat domain (CTD). Moreover, CTD phosphorylation at serine 5, a hallmark of the initiating polymerase, but not at serine 2, a hallmark of the elongating polymerase, completely inhibits ubiquitylation. In agreement with this, ubiquitylated RNAPII is hypophosphorylated at serine 5 in vivo, and mutation of the serine 5 phosphatase SSU72 inhibits RNAPII degradation. These results identify several mechanisms that confine ubiquitylation of RNAPII to the forms of the enzyme that arrest during elongation.


Assuntos
RNA Polimerase II/metabolismo , Transcrição Gênica , Ubiquitina/metabolismo , Sistema Livre de Células , Proteínas Cromossômicas não Histona/metabolismo , Proteínas Cromossômicas não Histona/farmacologia , Fosfoproteínas Fosfatases/química , Fosfoproteínas Fosfatases/farmacologia , Fosforilação , RNA Polimerase II/efeitos dos fármacos , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/farmacologia , Serina/química , Ubiquitina/efeitos dos fármacos
18.
Plant Physiol ; 134(4): 1598-613, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15047896

RESUMO

To investigate the role of stored and neosynthesized mRNAs in seed germination, we examined the effect of alpha-amanitin, a transcriptional inhibitor targeting RNA polymerase II, on the germination of nondormant Arabidopsis seeds. We used transparent testa mutants, of which seed coat is highly permeable, to better ascertain that the drug can reach the embryo during seed imbibition. Even with the most permeable mutant (tt2-1), germination (radicle protrusion) occurred in the absence of transcription, while subsequent seedling growth was blocked. In contrast, germination was abolished in the presence of the translational inhibitor cycloheximide. Taken together, the results highlight the role of stored proteins and mRNAs for germination in Arabidopsis and show that in this species the potential for germination is largely programmed during the seed maturation process. The alpha-amanitin-resistant germination exhibited characteristic features. First, this germination was strongly slowed down, indicating that de novo transcription normally allows the synthesis of factor(s) activating the germination rate. Second, the sensitivity of germination to gibberellic acid was reduced 15-fold, confirming the role of this phytohormone in germination. Third, de novo synthesis of enzymes involved in reserve mobilization and resumption of metabolic activity was repressed, thus accounting for the failure in seedling establishment. Fourth, germinating seeds can recapitulate at least part of the seed maturation program, being capable of using mRNAs stored during development. Thus, commitment to germination and plant growth requires transcription of genes allowing the imbibed seed to discriminate between mRNAs to be utilized in germination and those to be destroyed.


Assuntos
Amanitinas/farmacologia , Arabidopsis/genética , Germinação/genética , Proteoma/análise , RNA Mensageiro/genética , Sementes/genética , Arabidopsis/efeitos dos fármacos , Arabidopsis/crescimento & desenvolvimento , Cicloeximida/farmacologia , Germinação/efeitos dos fármacos , Germinação/fisiologia , Giberelinas/farmacologia , Mutação , Inibidores da Síntese de Ácido Nucleico/farmacologia , Reguladores de Crescimento de Plantas/farmacologia , Inibidores da Síntese de Proteínas/farmacologia , Proteoma/genética , RNA Polimerase II/efeitos dos fármacos , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , RNA Mensageiro/metabolismo , Sementes/efeitos dos fármacos , Sementes/metabolismo , Uridina Monofosfato/metabolismo
19.
Mol Cell Biol ; 23(17): 6129-38, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12917335

RESUMO

Transcriptional activation of the heat shock genes during the heat shock response in Drosophila has been intimately linked to phosphorylation of histone H3 at serine 10, whereas repression of non-heat-shock genes correlates with dephosphorylation of histone H3. It is then possible that specific kinase and/or phosphatase activities may regulate histone phosphorylation and therefore transcription activation and repression, respectively. We find that treatment of cells with strong phosphatase inhibitors interferes with the genome-wide dephosphorylation of histone H3 normally observed at non-heat-shock genes during heat shock. Mutants in protein phosphatase type 2A (PP2A) also display reduced genome-wide H3 dephosphorylation, and sites of H3 phosphorylation that do not contain heat shock genes remain transcriptionally active during heat shock in PP2A mutants. Finally, the SET protein, a potent and highly selective inhibitor of PP2A activity that inhibits PP2A-mediated dephosphorylation of Ser10-phosphorylated H3, is detected at transcriptionally active regions of polytene chromosomes. These results suggest that activation and repression of gene expression during heat shock might be regulated by changes in PP2A activity controlled by the SET protein.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Histonas/genética , Histonas/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Transcrição Gênica , Animais , Proteínas Cromossômicas não Histona , Drosophila melanogaster , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica , Resposta ao Choque Térmico/efeitos dos fármacos , Resposta ao Choque Térmico/genética , Larva , Mutação , Ácido Okadáico/farmacologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosfoproteínas Fosfatases/genética , Fosforilação , Proteína Fosfatase 2 , Proteínas/genética , Proteínas/metabolismo , RNA Polimerase II/efeitos dos fármacos , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Glândulas Salivares/efeitos dos fármacos , Glândulas Salivares/fisiologia , Serina/metabolismo , Fatores de Transcrição
20.
J Cell Biol ; 159(5): 777-82, 2002 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-12473686

RESUMO

RNA polymerase II transcribes most eukaryotic genes. Its catalytic subunit was tagged with green fluorescent protein and expressed in Chinese hamster cells bearing a mutation in the same subunit; it complemented the defect and so was functional. Photobleaching revealed two kinetic fractions of polymerase in living nuclei: approximately 75% moved rapidly, but approximately 25% was transiently immobile (association t1/2 approximately 20 min) and transcriptionally active, as incubation with 5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole eliminated it. No immobile but inactive fraction was detected, providing little support for the existence of a stable holoenzyme, or the slow stepwise assembly of a preinitiation complex on promoters or the nuclear substructure. Actinomycin D decreased the rapidly moving fraction, suggesting that engaged polymerases stall at intercalated molecules while others initiate. When wild-type cells containing only the endogenous enzyme were incubated with [3H]uridine, nascent transcripts became saturated with tritium with similar kinetics (t1/2 approximately 14 min). These data are consistent with a polymerase being mobile for one half to five sixths of a transcription cycle, and rapid assembly into the preinitiation complex. Then, most expressed transcription units would spend significant times unassociated with engaged polymerases.


Assuntos
RNA Polimerase II/metabolismo , Transcrição Gênica , Animais , Linhagem Celular , Núcleo Celular/enzimologia , Núcleo Celular/metabolismo , Células Cultivadas , Cricetinae , Dactinomicina/farmacologia , Diclororribofuranosilbenzimidazol/farmacologia , Regulação Enzimológica da Expressão Gênica , Proteínas de Fluorescência Verde , Cinética , Proteínas Luminescentes/metabolismo , Modelos Genéticos , Mutagênese/fisiologia , Mutação , Inibidores da Síntese de Ácido Nucleico/farmacologia , Periodicidade , Fotodegradação , RNA Polimerase II/efeitos dos fármacos , Temperatura , Fatores de Tempo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA