Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Hepatology ; 72(5): 1771-1785, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32060938

RESUMO

BACKGROUND AND AIMS: This study examined whether enhanced susceptibility of steatotic liver to ischemia-reperfusion (I/R) injury is due to impaired recruitment of bone marrow (BM) progenitors of liver sinusoidal endothelial cells (LSECs, also called sinusoidal endothelial cell progenitor cells [sprocs]) with diminished repair of injured LSECs and whether restoring signaling to recruit BM sprocs reduces I/R injury. APPROACH AND RESULTS: Hepatic vessels were clamped for 1 hour in rats fed a high-fat, high-fructose (HFHF) diet for 5, 10, or 15 weeks. Matrix metalloproteinase 9 (MMP-9) antisense oligonucleotides (ASO) or an MMP inhibitor were used to induce liver-selective MMP-9 inhibition. HFHF rats had mild, moderate, and severe steatosis, respectively, at 5, 10, and 15 weeks. I/R injury was enhanced in HFHF rats; this was accompanied by complete absence of hepatic vascular endothelial growth factor (VEGF)-stromal cell-derived factor 1 (sdf1) signaling, leading to lack of BM sproc recruitment. Liver-selective MMP-9 inhibition to protect against proteolytic cleavage of hepatic VEGF using either MMP-9 ASO or intraportal MMP inhibitor in 5-week and 10-week HFHF rats enhanced hepatic VEGF-sdf1 signaling, increased BM sproc recruitment, and reduced alanine aminotransferase (ALT) by 92% and 77% at 5 weeks and by 80% and 64% at 10 weeks of the HFHF diet, respectively. After I/R injury in 15-week HFHF rats, the MMP inhibitor reduced active MMP-9 expression by 97%, ameliorated histologic evidence of injury, and reduced ALT by 58%, which is comparable to control rats sustaining I/R injury. Rescue therapy with intraportal MMP inhibitor, given after ischemia, in the 5-week HFHF rat reduced ALT by 71% and reduced necrosis. CONCLUSIONS: Lack of signaling to recruit BM sprocs that repair injured LSECs renders steatotic liver more susceptible to I/R injury. Liver-selective MMP-9 inhibition enhances VEGF-sdf1 signaling and recruitment of BM sprocs, which markedly protects against I/R injury, even in severely steatotic rats.


Assuntos
Células Progenitoras Endoteliais/efeitos dos fármacos , Fígado Gorduroso/etiologia , Metaloproteinase 9 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz/farmacologia , Traumatismo por Reperfusão/prevenção & controle , Animais , Transplante de Medula Óssea , Dieta Hiperlipídica , Açúcares da Dieta/efeitos adversos , Modelos Animais de Doenças , Suscetibilidade a Doenças/terapia , Células Progenitoras Endoteliais/patologia , Fígado Gorduroso/diagnóstico , Fígado Gorduroso/tratamento farmacológico , Frutose/efeitos adversos , Humanos , Fígado/irrigação sanguínea , Fígado/diagnóstico por imagem , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Inibidores de Metaloproteinases de Matriz/uso terapêutico , Microvasos/citologia , Microvasos/efeitos dos fármacos , Microvasos/patologia , Ratos , Traumatismo por Reperfusão/etiologia
2.
J Biol Chem ; 294(50): 19255-19268, 2019 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-31645432

RESUMO

MAF1 homolog, negative regulator of RNA polymerase III (MAF1) is a key repressor of RNA polymerase (pol) III-dependent transcription and functions as a tumor suppressor. Its expression is frequently down-regulated in primary human hepatocellular carcinomas (HCCs). However, this reduction in MAF1 protein levels does not correlate with its transcript levels, indicating that MAF1 is regulated post-transcriptionally. Here, we demonstrate that MAF1 is a labile protein whose levels are regulated through the ubiquitin-dependent proteasome pathway. We found that MAF1 ubiquitination is enhanced upon mTOR complex 1 (TORC1)-mediated phosphorylation at Ser-75. Moreover, we observed that the E3 ubiquitin ligase cullin 2 (CUL2) critically regulates MAF1 ubiquitination and controls its stability and subsequent RNA pol III-dependent transcription. Analysis of the phenotypic consequences of modulating either CUL2 or MAF1 protein expression revealed changes in actin cytoskeleton reorganization and altered sensitivity to doxorubicin-induced apoptosis. Repression of RNA pol III-dependent transcription by chemical inhibition or knockdown of BRF1 RNA pol III transcription initiation factor subunit (BRF1) enhanced HCC cell sensitivity to doxorubicin, suggesting that MAF1 regulates doxorubicin resistance in HCC by controlling RNA pol III-dependent transcription. Together, our results identify the ubiquitin proteasome pathway and CUL2 as important regulators of MAF1 levels. They suggest that decreases in MAF1 protein underlie chemoresistance in HCC and perhaps other cancers and point to an important role for MAF1 and RNA pol III-mediated transcription in chemosensitivity and apoptosis.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Doxorrubicina/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Repressoras/antagonistas & inibidores , Ubiquitina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Knockout , Fosforilação/efeitos dos fármacos , Proteínas Repressoras/deficiência , Proteínas Repressoras/metabolismo
3.
Cell Rep ; 24(7): 1852-1864, 2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-30110641

RESUMO

RNA polymerase (pol) III transcribes a variety of small untranslated RNAs involved in transcription, RNA processing, and translation. RNA pol III and its components are altered in various human developmental disorders, yet their roles in cell fate determination and development are poorly understood. Here we demonstrate that Maf1, a transcriptional repressor, promotes induction of mouse embryonic stem cells (mESCs) into mesoderm. Reduced Maf1 expression in mESCs and preadipocytes impairs adipogenesis, while ectopic Maf1 expression in Maf1-deficient cells enhances differentiation. RNA pol III repression by chemical inhibition or knockdown of Brf1 promotes adipogenesis. Altered RNA pol III-dependent transcription produces select changes in mRNAs with a significant enrichment of adipogenic gene signatures. Furthermore, RNA pol III-mediated transcription positively regulates long non-coding RNA H19 and Wnt6 expression, established adipogenesis inhibitors. Together, these studies reveal an important and unexpected function for RNA pol III-mediated transcription and Maf1 in mesoderm induction and adipocyte differentiation.


Assuntos
Adipócitos/metabolismo , Adipogenia/genética , RNA Polimerase III/genética , Proteínas Repressoras/genética , Transcrição Gênica , Adipócitos/citologia , Animais , Fator 1 de Resposta a Butirato , Diferenciação Celular , Corpos Embrioides/citologia , Corpos Embrioides/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Mesoderma/citologia , Mesoderma/crescimento & desenvolvimento , Mesoderma/metabolismo , Camundongos , Camundongos Nus , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Cultura Primária de Células , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , RNA Polimerase III/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
4.
Oncotarget ; 8(30): 48832-48845, 2017 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-28415573

RESUMO

The TATA-binding protein (TBP) plays a central role in eukaryotic gene transcription. Given its key function in transcription initiation, TBP was initially thought to be an invariant protein. However, studies showed that TBP expression is upregulated by oncogenic signaling pathways. Furthermore, depending on the cell type, small increases in cellular TBP amounts can induce changes in cellular growth properties towards a transformed phenotype. Here we sought to identify the specific TBP-regulated gene targets that drive its ability to induce tumorigenesis. Using microarray analysis, our results reveal that increases in cellular TBP concentrations produce selective alterations in gene expression that include an enrichment for genes involved in angiogenesis. Accordingly, we find that TBP levels modulate VEGFA expression, the master regulator of angiogenesis. Increases in cellular TBP amounts induce VEGFA expression and secretion to enhance cell migration and tumor vascularization. TBP mediates changes in VEGFA transcription requiring its recruitment at a hypoxia-insensitive proximal TSS, revealing a mechanism for VEGF regulation under non-stress conditions. The results are clinically relevant as TBP expression is significantly increased in both colon adenocarcinomas as well as adenomas relative to normal tissue. Furthermore, TBP expression is positively correlated with VEGFA expression. Collectively, these studies support the idea that increases in TBP expression contribute to enhanced VEGFA transcription early in colorectal cancer development to drive tumorigenesis.


Assuntos
Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteína de Ligação a TATA-Box/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Sítios de Ligação , Movimento Celular/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Neoplasias do Colo/patologia , Expressão Gênica , Perfilação da Expressão Gênica , Genes Reporter , Humanos , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , RNA Polimerase II/metabolismo , Proteína de Ligação a TATA-Box/genética , Sítio de Iniciação de Transcrição , Transcriptoma , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
Trends Endocrinol Metab ; 27(10): 742-750, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27296319

RESUMO

PTEN is a critical tumor suppressor whose dysregulation leads to metabolic disease and cancer. How these diseases are linked at a molecular level is poorly understood. Maf1 is a novel PTEN target that connects PTEN's ability to repress intracellular lipid accumulation with its tumor suppressor function. Maf1 represses the expression of rRNAs and tRNAs to restrain biosynthetic capacity and oncogenic transformation. Recent studies demonstrate that Maf1 also controls intracellular lipid accumulation. In animal models, dysregulation of RNA polymerase I- and III-dependent transcription, and subsequent upregulation of rRNAs and tRNAs, leads to altered lipid metabolism and storage. Together these results identify unexpected connections between RNA and lipid metabolism that may help explain the strong epidemiological association between obesity and cancer.


Assuntos
Metabolismo dos Lipídeos/fisiologia , PTEN Fosfo-Hidrolase/metabolismo , RNA/metabolismo , Proteínas Repressoras/metabolismo , Animais , Humanos , Metabolismo dos Lipídeos/genética , PTEN Fosfo-Hidrolase/genética , RNA/genética , Proteínas Repressoras/genética
6.
J Dent Educ ; 80(1): 73-82, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26729687

RESUMO

The aims of this study were to document the extent of nutritional content in U.S. dental hygiene program curricula; identify program directors' opinions, perceptions, and barriers to expanding nutritional content; and evaluate if a proposed nutrition curriculum model would be beneficial. This mixed methods study involved quantitative and qualitative aspects. An invitation letter was sent to all 335 directors of entry-level U.S. dental hygiene programs. In response, 55 directors submitted nutrition course syllabi from their programs (16.4% of the total) for the quantitative analysis. In addition, 14 nutrition instructors and ten program directors were interviewed regarding their perceptions and opinions of nutrition education for dental hygiene students. All aspects of the content analysis results revealed that nutrition content in entry-level dental hygiene programs is diverse. Some programs did not include nutrition content, while others provided oral and systemic nutrition intervention subject matter. Some programs offered multiple clinical nutrition applications and patient contact opportunities while most required none. The interview results disclosed a variety of opinions and perceptions of dental hygienists' role in nutrition. Several interviewees viewed dental hygienists' role in nutrition to be an integral part of patient care, while others indicated no role or providing caries prevention counseling only. Although dental hygienists are expected to provide nutrition assessments and interventions, no standards or standardized competencies exist for nutrition in dental hygiene education. A standardized nutrition model could be beneficial for entry-level programs to ensure dental hygienists possess basic knowledge to perform nutrition assessments and intervention to address Healthy People 2020's intervention initiatives.


Assuntos
Currículo , Higienistas Dentários/educação , Ciências da Nutrição/educação , Pessoal Administrativo , Atitude do Pessoal de Saúde , Aconselhamento , Cárie Dentária/prevenção & controle , Dietética/educação , Docentes , Promoção da Saúde , Humanos , Avaliação Nutricional , Relações Profissional-Paciente , Estados Unidos
7.
PLoS Genet ; 10(12): e1004789, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25502566

RESUMO

Maf1 was initially identified as a transcriptional repressor of RNA pol III-transcribed genes, yet little is known about its other potential target genes or its biological function. Here, we show that Maf1 is a key downstream target of PTEN that drives both its tumor suppressor and metabolic functions. Maf1 expression is diminished with loss of PTEN in both mouse models and human cancers. Consistent with its role as a tumor suppressor, Maf1 reduces anchorage-independent growth and tumor formation in mice. PTEN-mediated changes in Maf1 expression are mediated by PTEN acting on PI3K/AKT/FoxO1 signaling, revealing a new pathway that regulates RNA pol III-dependent genes. This regulatory event is biologically relevant as diet-induced PI3K activation reduces Maf1 expression in mouse liver. We further identify lipogenic enzymes as a new class of Maf1-regulated genes whereby Maf1 occupancy at the FASN promoter opposes SREBP1c-mediated transcription activation. Consistent with these findings, Maf1 inhibits intracellular lipid accumulation and increasing Maf1 expression in mouse liver abrogates diet-mediated induction of lipogenic enzymes and triglycerides. Together, these results establish a new biological role for Maf1 as a downstream effector of PTEN/PI3K signaling and reveal that Maf1 is a key element by which this pathway co-regulates lipid metabolism and oncogenesis.


Assuntos
Carcinogênese , Metabolismo dos Lipídeos/genética , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Repressoras/metabolismo , Animais , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Células Hep G2 , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/complicações , Neoplasias/genética , Obesidade/complicações , Obesidade/genética , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/genética , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Transdução de Sinais
8.
Aging Cell ; 12(6): 1000-11, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23826727

RESUMO

Tissue regeneration diminishes with age, concurrent with declining hormone levels including growth factors such as insulin-like growth factor-1 (IGF-1). We investigated the molecular basis for such decline in pancreatic ß-cells where loss of proliferation occurs early in age and is proposed to contribute to the pathogenesis of diabetes. We studied the regeneration capacity of ß-cells in mouse model where PI3K/AKT pathway downstream of insulin/IGF-1 signaling is upregulated by genetic deletion of Pten (phosphatase and tensin homologue deleted on chromosome 10) specifically in insulin-producing cells. In this model, PTEN loss prevents the decline in proliferation capacity in aged ß-cells and restores the ability of aged ß-cells to respond to injury-induced regeneration. Using several animal and cell models where we can manipulate PTEN expression, we found that PTEN blocks cell cycle re-entry through a novel pathway leading to an increase in p16(ink4a), a cell cycle inhibitor characterized for its role in cellular senescence/aging. A downregulation in p16(ink4a) occurs when PTEN is lost as a result of cyclin D1 induction and the activation of E2F transcription factors. The activation of E2F transcriptional factors leads to methylation of p16(ink4a) promoter, an event that is mediated by the upregulation of polycomb protein, Ezh2. These analyses establish a novel PTEN/cyclin D1/E2F/Ezh2/p16(ink4a) signaling network responsible for the aging process and provide specific evidence for a molecular paradigm that explain how decline in growth factor signals such as IGF-1 (through PTEN/PI3K signaling) may control regeneration and the lack thereof in aging cells.


Assuntos
Ciclo Celular , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , PTEN Fosfo-Hidrolase/metabolismo , Envelhecimento/patologia , Animais , Proliferação de Células , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Metilação de DNA/genética , Regulação para Baixo/genética , Proteína Potenciadora do Homólogo 2 de Zeste , Deleção de Genes , Humanos , Camundongos , PTEN Fosfo-Hidrolase/deficiência , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Transdução de Sinais , Regulação para Cima/genética
9.
J Biol Chem ; 288(26): 19288-95, 2013 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-23673667

RESUMO

RNA polymerase (pol) III transcribes genes that determine biosynthetic capacity. Induction of these genes is required for oncogenic transformation. The transcriptional repressor, Maf1, plays a central role in the repression of these and other genes that promote oncogenesis. Our studies identify an important new role for SUMOylation in repressing RNA pol III-dependent transcription. We show that a key mechanism by which this occurs is through small ubiquitin-like modifier (SUMO) modification of Maf1 by both SUMO1 and SUMO2. Mutation of each lysine residue revealed that Lys-35 is the major SUMOylation site on Maf1 and that the deSUMOylase, SENP1, is responsible for controlling Maf1K35 SUMOylation. SUMOylation of Maf1 is unaffected by rapamycin inhibition of mammalian target of rapamycin (mTOR) and mTOR-dependent Maf1 phosphorylation. By preventing SUMOylation at Lys-35, Maf1 is impaired in its ability to both repress transcription and suppress colony growth. Although SUMOylation does not alter Maf1 subcellular localization, Maf1K35R is defective in its ability to associate with RNA pol III. This impairs Maf1 recruitment to tRNA gene promoters and its ability to facilitate the dissociation of RNA pol III from these promoters. These studies identify a novel role for SUMOylation in controlling Maf1 and RNA pol III-mediated transcription. Given the emerging roles of SENP1, Maf1, and RNA pol III transcription in oncogenesis, our studies support the idea that deSUMOylation of Maf1 and induction of its gene targets play a critical role in cancer development.


Assuntos
Endopeptidases/metabolismo , Regulação Neoplásica da Expressão Gênica , RNA Polimerase III/metabolismo , Proteínas Repressoras/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Células COS , Linhagem Celular Tumoral , Proliferação de Células , Chlorocebus aethiops , Cisteína Endopeptidases , Células HEK293 , Humanos , Lisina/química , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Neoplasias/metabolismo , Processamento de Proteína Pós-Traducional , Fatores de Transcrição/metabolismo , Transcrição Gênica
10.
Mol Cell Biol ; 32(6): 1124-38, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22252323

RESUMO

VPS4B, an AAA ATPase (ATPase associated with various cellular activities), participates in vesicular trafficking and autophagosome maturation in mammalian cells. In solid tumors, hypoxia is a common feature and an indicator of poor treatment outcome. Our studies demonstrate that exogenous or endogenous (assessed with anchorage-independent three-dimensional multicellular spheroid culture) hypoxia induces VPS4B downregulation by the ubiquitin-proteasome system. Inhibition of VPS4B function by short hairpin VPS4B (sh-VPS4B) or expression of dominant negative VPS4B(E235Q) promotes anchorage-independent breast cancer cell growth and resistance to gefitinib, U0126, and genotoxicity. Biochemically, hyperactivation of epidermal growth factor receptor (EGFR), a receptor tyrosine kinase essential for cell proliferation and survival, accompanied by increased EGFR accumulation and altered intracellular compartmentalization, is observed in cells with compromised VPS4B. Furthermore, enhanced FOS/JUN induction and AP-1 promoter activation are noted in EGF-treated cells with VPS4B knockdown. However, VPS4B depletion does not affect EGFRvIII stability or its associated signaling. An inverse correlation between VPS4B expression and EGFR abundance is observed in breast tumors, and high-grade or recurrent breast carcinomas exhibit lower VPS4B expression. Together, our findings highlight a potentially critical role of VPS4B downregulation or chronic-hypoxia-induced VPS4B degradation in promoting tumor progression, unveiling a nongenomic mechanism for EGFR overproduction in human breast cancer.


Assuntos
Adenosina Trifosfatases/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Receptores ErbB/metabolismo , Transdução de Sinais , ATPases Associadas a Diversas Atividades Celulares , Adenosina Trifosfatases/genética , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Hipóxia Celular , Linhagem Celular , Linhagem Celular Tumoral , Regulação para Baixo , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Fosforilação , RNA Mensageiro/genética , Ratos , Esferoides Celulares
11.
West J Nurs Res ; 34(3): 340-76, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21403057

RESUMO

This study examined whether parent-youth dyads participating in the Strengthening Families Program 10-14 (SFP 10-14) would demonstrate greater postprogram family cohesion, communication, involvement, and supervision and if youth would report less alcohol, tobacco, and other drugs involvement in contrast to a comparison group. From 16 randomly selected schools, we recruited 167 parent-youth dyads: 86 from intervention and 81 from comparison schools. The intention-to-treat analysis found one significant change in family environment. Considering dose, it was found that among dyads receiving a full dose, all the outcomes were in the expected direction and effect sizes were moderate. Among dyads receiving a partial dose, 10 of 18 outcomes were in the direction opposite that expected. Youth participation in alcohol, tobacco, and other drugs was very low and did not differ postprogram. Although the expected outcomes were not realized, findings descriptive of dosage effects make a valuable contribution to the field. Study of factors that distinguish intervention completers from noncompleters is recommended.


Assuntos
Família , Relações Pais-Filho , Adolescente , Adulto , Feminino , Humanos , Masculino , Transtornos Relacionados ao Uso de Substâncias/prevenção & controle
12.
J Biol Chem ; 286(4): 2393-401, 2011 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-21106530

RESUMO

Chronic alcohol consumption is associated with steatohepatitis and cirrhosis, enhancing the risk for hepatocellular carcinoma. RNA polymerase (pol) III transcribes a variety of small, untranslated RNAs, including tRNAs and 5S rRNAs, which determine the biosynthetic capacity of cells. Increased RNA pol III-dependent transcription, observed in transformed cells and human tumors, is required for oncogenic transformation. Given that alcohol consumption increases risk for liver cancer, we examined whether alcohol regulates this class of genes. Ethanol induces RNA pol III-dependent transcription in both HepG2 cells and primary mouse hepatocytes in a manner that requires ethanol metabolism and the activation of JNK1. This regulatory event is mediated, at least in part, through the ability of ethanol to induce expression of the TFIIIB components, Brf1, and the TATA-binding protein (TBP). Induction of TBP, Brf1, and RNA pol III-dependent gene expression is driven by enhanced c-Jun expression. Ethanol promotes a marked increase in the direct recruitment of c-Jun to TBP, Brf1, and tRNA gene promoters. Chronic alcohol administration in mice leads to enhanced expression of TBP, Brf1, tRNA, and 5S rRNA gene transcription in the liver. These alcohol-dependent increases are more pronounced in transgenic animals that express the HCV NS5A protein that display increased incidence of liver tumors. Together, these results identify a new class of genes that are regulated by alcohol through the co-regulation of TFIIIB components and define a central role for c-Jun in this process.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Nucleares/biossíntese , Proteínas Proto-Oncogênicas c-jun/metabolismo , RNA Polimerase III/metabolismo , Proteínas de Ligação a RNA/biossíntese , Fatores Associados à Proteína de Ligação a TATA/biossíntese , Proteína de Ligação a TATA-Box/metabolismo , Animais , Fator 1 de Resposta a Butirato , Carcinoma Hepatocelular/induzido quimicamente , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Depressores do Sistema Nervoso Central/efeitos adversos , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Etanol/efeitos adversos , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Regulação da Expressão Gênica/genética , Células Hep G2 , Humanos , Cirrose Hepática Alcoólica/genética , Cirrose Hepática Alcoólica/metabolismo , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas c-jun/genética , RNA Polimerase III/genética , RNA Ribossômico 5S/biossíntese , RNA Ribossômico 5S/genética , RNA de Transferência/biossíntese , RNA de Transferência/genética , Proteínas de Ligação a RNA/genética , Elementos de Resposta/genética , Fatores Associados à Proteína de Ligação a TATA/genética , Proteína de Ligação a TATA-Box/genética , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética , Proteínas não Estruturais Virais/biossíntese , Proteínas não Estruturais Virais/genética
13.
Proc Natl Acad Sci U S A ; 106(31): 12682-7, 2009 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-19620725

RESUMO

RNA polymerase (pol) III-dependent transcription is subject to stringent regulation by tumor suppressors and oncogenic proteins and enhanced RNA pol III transcription is essential for cellular transformation and tumorigenesis. Since the c-Jun N-terminal kinases (JNKs) display both oncogenic and tumor suppressor properties, the roles of these proteins in regulating RNA pol III transcription were examined. In both mouse and human cells, loss or reduction in JNK1 expression represses RNA pol III transcription. In contrast, loss or reduction in JNK2 expression induces transcription. The JNKs coordinately regulate expression of all 3 TFIIIB subunits. While JNK1 positively regulates TBP expression, the RNA pol III-specific factors, Brf1 and Bdp1, JNK2 negatively regulates their expression. Brf1 is coregulated with TBP through the JNK target, Elk-1. Reducing Elk-1 expression decreases Brf1 expression. Decreasing JNK1 expression reduces Elk-1 occupancy at the Brf1 promoter, while decreasing JNK2 expression enhances recruitment of Elk-1 to the Brf1 promoter. In contrast, regulation of Bdp1 occurs through JNK-mediated alterations in TBP expression. Altered TBP expression mimics the effect of reduced JNK1 or JNK2 levels on Bdp1 expression. Decreasing JNK1 expression reduces the occupancy of TBP at the Bdp1 promoter, while decreasing JNK2 expression enhances recruitment of TBP to the Bdp1 promoter. Together, these results provide a molecular mechanism for regulating RNA pol III transcription through the coordinate control of TFIIIB subunit expression and elucidate opposing functions for the JNKs in regulating a large class of genes that dictate the biosynthetic capacity of cells.


Assuntos
Proteína Quinase 8 Ativada por Mitógeno/fisiologia , Proteína Quinase 9 Ativada por Mitógeno/fisiologia , RNA Polimerase III/fisiologia , Fator de Transcrição TFIIIB/genética , Transcrição Gênica , Regulação da Expressão Gênica , Humanos , Regiões Promotoras Genéticas , Subunidades Proteicas , Proteínas Tirosina Fosfatases não Receptoras/genética , Fatores Associados à Proteína de Ligação a TATA/genética , Proteína de Ligação a TATA-Box/genética , Proteína de Ligação a TATA-Box/metabolismo , Proteínas Elk-1 do Domínio ets/metabolismo
14.
Mol Cell Biol ; 28(20): 6483-95, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18710943

RESUMO

The epidermal growth factor receptor (EGFR) family regulates essential biological processes. Various epithelial tumors are linked to EGFR overexpression or expression of variant forms, such as the EGFR1 variant, EGFRvIII. Perturbations in expression of the transcription initiation factor, TATA-binding protein (TBP), alter cellular growth properties. Here we demonstrate that EGFR1 and EGFRvIII, but not HER2, induce TBP expression at a transcriptional level through distinct mechanisms. EGFR1 enhances the phosphorylation and function of Elk-1, recruiting it to the TBP promoter. In contrast, EGFRvIII robustly induces c-jun expression, stimulating recruitment of c-fos/c-jun to an overlapping AP-1 site. Enhancing c-jun expression alone induces TBP promoter activity through the AP-1 site. To determine the underlying mechanism for differences in Elk-1 function and c-jun expression by these receptors, we inhibited the internalization of EGFR1. Persistent EGFR1 cell surface occupancy mimics EGFRvIII-mediated effects on Elk-1 and c-jun and switches the requirement of Elk-1 to AP-1 for TBP promoter induction. Together, these studies define a new molecular mechanism for the regulation of TBP expression. In addition, we identify distinct molecular targets of EGFR1 and EGFRvIII and demonstrate the importance of receptor internalization in distinguishing their specific functions.


Assuntos
Receptores ErbB/metabolismo , Transdução de Sinais , Proteína de Ligação a TATA-Box/metabolismo , Animais , Linhagem Celular , Clatrina/metabolismo , Endocitose , Ativação Enzimática , Humanos , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mimetismo Molecular , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Receptor ErbB-2/metabolismo , Proteína de Ligação a TATA-Box/genética , Fator de Transcrição AP-1/metabolismo , Transcrição Gênica , Proteínas Elk-1 do Domínio ets/metabolismo
15.
J Biol Chem ; 283(28): 19184-91, 2008 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-18456653

RESUMO

RNA polymerase (pol) III transcription, responsible for the synthesis of various stable RNAs, including 5 S rRNAs and tRNAs, is regulated by oncogenic proteins and tumor suppressors. Although it is well established that RNA pol III-dependent transcription is deregulated in transformed cells and malignant tumors, it has not been determined whether this represents a cause or consequence of these processes. We show that Rat1a fibroblasts undergoing oncogenic transformation by the TATA-binding protein or c-Myc display enhanced RNA pol III transcription. Decreased expression of the RNA pol III-specific transcription factor Brf1 prevented this increase in RNA pol III transcription. Although the overall proliferation rates of these cells remained unchanged, the ability of cells to grow in an anchorage-independent manner and form tumors in mice was markedly reduced. Although overexpression of Brf1 modestly stimulated RNA pol III transcription, expression of a phosphomimic, Brf1-T145D, more significantly induced transcription. However, these increases in transcription were not sufficient to promote cellular transformation. Together, these results demonstrate that enhanced RNA pol III transcription is essential for anchorage-independent growth and tumorigenesis and that these events can be uncoupled from effects on anchorage-dependent proliferation.


Assuntos
Transformação Celular Neoplásica/metabolismo , Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Polimerase III/metabolismo , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Substituição de Aminoácidos , Animais , Adesão Celular/genética , Linhagem Celular , Proliferação de Células , Transformação Celular Neoplásica/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Camundongos , Camundongos Nus , Mutação de Sentido Incorreto , Proteínas Proto-Oncogênicas c-myc/genética , RNA Polimerase III/genética , RNA Ribossômico 5S/biossíntese , RNA Ribossômico 5S/genética , RNA de Transferência/biossíntese , RNA de Transferência/genética , Ratos , Fatores Associados à Proteína de Ligação a TATA/genética , Proteína de Ligação a TATA-Box/genética , Proteína de Ligação a TATA-Box/metabolismo , Transcrição Gênica/genética
16.
Mol Cell Biol ; 28(12): 4204-14, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18391023

RESUMO

PTEN, a tumor suppressor whose function is frequently lost in human cancers, possesses a lipid phosphatase activity that represses phosphatidylinositol 3-kinase (PI3K) signaling, controlling cell growth, proliferation, and survival. The potential for PTEN to regulate the synthesis of RNA polymerase (Pol) III transcription products, including tRNAs and 5S rRNAs, was evaluated. The expression of PTEN in PTEN-deficient cells repressed RNA Pol III transcription, whereas decreased PTEN expression enhanced transcription. Transcription repression by PTEN was uncoupled from PTEN-mediated effects on the cell cycle and was independent of p53. PTEN acts through its lipid phosphatase activity, inhibiting the PI3K/Akt/mTOR/S6K pathway to decrease transcription. PTEN, through the inactivation of mTOR, targets the TFIIIB complex, disrupting the association between TATA-binding protein and Brf1. Kinetic analysis revealed that PTEN initially induces a decrease in the serine phosphorylation of Brf1, leading to a selective reduction in the occupancy of all TFIIIB subunits on tRNA(Leu) genes, whereas prolonged PTEN expression results in the enhanced serine phosphorylation of Bdp1. Together, these results demonstrate a new class of genes regulated by PTEN through its ability to repress the activation of PI3K/Akt/mTOR/S6K signaling.


Assuntos
Regulação Neoplásica da Expressão Gênica , PTEN Fosfo-Hidrolase/metabolismo , RNA Polimerase III/metabolismo , Fator de Transcrição TFIIIB/química , Ciclo Celular , Linhagem Celular Tumoral , Citoplasma/metabolismo , Humanos , Modelos Biológicos , Fosfatidato Fosfatase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Transdução de Sinais , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo
18.
Mol Cell ; 26(3): 367-79, 2007 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-17499043

RESUMO

Most eukaryotic transcriptional regulators act in an RNA polymerase (Pol)-selective manner. Here we show that the human Maf1 protein negatively regulates transcription by all three nuclear Pols. Changes in Maf1 expression affect Pol I- and Pol III-dependent transcription in human glioblastoma lines. These effects are mediated, in part, through the ability of Maf1 to repress transcription of the TATA binding protein, TBP. Maf1 targets an Elk-1-binding site in the TBP promoter, and its occupancy of this region is reciprocal with that of Elk-1. Similarly, Maf1 occupancy of Pol III genes is inversely correlated with that of the initiation factor TFIIIB and Pol III. The phenotypic consequences of reducing Maf1 expression include changes in cell morphology and the accumulation of actin stress fibers, whereas Maf1 overexpression suppresses anchorage-independent growth. Together with the ability of Maf1 to reduce biosynthetic capacity, these findings support the idea that Maf1 regulates the transformation state of cells.


Assuntos
Núcleo Celular/metabolismo , RNA Polimerases Dirigidas por DNA/metabolismo , Regulação da Expressão Gênica , Proteínas Repressoras/metabolismo , Proteína de Ligação a TATA-Box/metabolismo , Fator de Transcrição TFIIIB/metabolismo , Transcrição Gênica/fisiologia , Sítios de Ligação , Processos de Crescimento Celular , Núcleo Celular/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , RNA Polimerases Dirigidas por DNA/genética , Humanos , Regiões Promotoras Genéticas/genética , Ligação Proteica , Interferência de RNA , RNA Polimerase I/genética , RNA Polimerase I/metabolismo , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , RNA Polimerase III/genética , RNA Polimerase III/metabolismo , Proteínas Repressoras/genética , Fatores Associados à Proteína de Ligação a TATA/genética , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Proteína de Ligação a TATA-Box/genética , Fator de Transcrição TFIIIB/genética , Células Tumorais Cultivadas
19.
Mol Cell Biol ; 27(1): 54-64, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17074809

RESUMO

Emerging evidence supports the idea that the c-Jun N-terminal kinases (JNKs) possess overlapping but distinct functions. The potential roles of the ubiquitously expressed JNK1 and JNK2 in regulating expression of the central transcription initiation factor, TATA-binding protein (TBP), were examined. Relative to wild-type fibroblasts, TBP was decreased in Jnk1(-/-) cells and increased in Jnk2(-/-) cells. Similarly, reduction of JNK1 in human hepatoma cells decreased TBP expression, whereas reduction of JNK2 enhanced it. JNK-mediated regulation of TBP expression occurs at the transcriptional level through their ability to target Elk-1, which directly regulates the TBP promoter in response to epidermal growth factor stimulation. JNK1 increases, whereas JNK2 decreases, the phosphorylation state of Elk-1, which differentially affects Elk-1 occupancy at a defined site within the TBP promoter. These JNK-mediated alterations in TBP expression, alone, serve to regulate c-Jun expression and fibroblast proliferation rates. These studies uncovered several new molecular events that distinguish the functions of JNK1 and JNK2 that are critical for their regulation of cellular proliferation.


Assuntos
Regulação da Expressão Gênica , Proteína Quinase 8 Ativada por Mitógeno/fisiologia , Proteína Quinase 9 Ativada por Mitógeno/fisiologia , Proteínas Proto-Oncogênicas c-jun/biossíntese , Proteína de Ligação a TATA-Box/biossíntese , Proteína de Ligação a TATA-Box/fisiologia , Proteínas Elk-1 do Domínio ets/fisiologia , Animais , Apoptose , Proliferação de Células , Fibroblastos/metabolismo , Humanos , Camundongos , Regiões Promotoras Genéticas , Proteína Proto-Oncogênica c-ets-1/metabolismo , Transcrição Gênica
20.
Mol Cell Biol ; 25(16): 6899-911, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16055704

RESUMO

PTEN is a tumor suppressor whose function is frequently lost in human cancer. It possesses a lipid phosphatase activity that represses the activation of PI3 kinase/Akt signaling, leading to decreased cell growth, proliferation, and survival. The potential for PTEN to regulate transcription of the large rRNAs by RNA polymerase I (RNA Pol I) was investigated. As increased synthesis of rRNAs is a hallmark of neoplastic transformation, the ability of PTEN to control the transcription of rRNAs might be crucial for its tumor suppressor function. The expression of PTEN in PTEN-deficient cells represses RNA Pol I transcription, while decreasing PTEN expression enhances transcription. PTEN-mediated repression requires its lipid phosphatase activity and is independent of the p53 status of the cell. This event can be uncoupled from PTEN's ability to regulate the cell cycle. RNA Pol I is regulated through PI3 kinase/Akt/mammalian target of rapamycin/S6 kinase, and the expression of constitutively activated S6 kinase is able to abrogate transcription repression by PTEN. No change in the expression of the RNA Pol I transcription components, upstream binding factor or SL1, was observed upon PTEN expression. However, chromatin immunoprecipitation assays demonstrate that PTEN differentially reduces the occupancy of the SL1 subunits on the rRNA gene promoter. Furthermore, PTEN induces dissociation of the SL1 subunits. Together, these results demonstrate that PTEN represses RNA Pol I transcription through a novel mechanism that involves disruption of the SL1 complex.


Assuntos
Monoéster Fosfórico Hidrolases/fisiologia , Proteínas Pol1 do Complexo de Iniciação de Transcrição/fisiologia , RNA Polimerase I/metabolismo , Transcrição Gênica , Proteínas Supressoras de Tumor/fisiologia , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Ciclina D1/metabolismo , DNA/química , Relação Dose-Resposta a Droga , Humanos , Immunoblotting , Imunoprecipitação , PTEN Fosfo-Hidrolase , Fosfatidilinositol 3-Quinases/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Proteínas Pol1 do Complexo de Iniciação de Transcrição/metabolismo , Regiões Promotoras Genéticas , RNA Ribossômico/química , RNA Interferente Pequeno/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais , Endonucleases Específicas para DNA e RNA de Cadeia Simples/metabolismo , Fatores de Tempo , Transfecção , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA