Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(16)2023 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-37629051

RESUMO

Obesity is a growing public health problem associated with increased risk of type 2 diabetes, cardiovascular disease, nonalcoholic fatty liver disease (NAFLD) and cancer. Here, we identify microRNA-22 (miR-22) as an essential rheostat involved in the control of lipid and energy homeostasis as well as the onset and maintenance of obesity. We demonstrate through knockout and transgenic mouse models that miR-22 loss-of-function protects against obesity and hepatic steatosis, while its overexpression promotes both phenotypes even when mice are fed a regular chow diet. Mechanistically, we show that miR-22 controls multiple pathways related to lipid biogenesis and differentiation. Importantly, genetic ablation of miR-22 favors metabolic rewiring towards higher energy expenditure and browning of white adipose tissue, suggesting that modulation of miR-22 could represent a viable therapeutic strategy for treatment of obesity and other metabolic disorders.


Assuntos
Diabetes Mellitus Tipo 2 , MicroRNAs , Hepatopatia Gordurosa não Alcoólica , Animais , Camundongos , Homeostase , Camundongos Transgênicos , Hepatopatia Gordurosa não Alcoólica/genética , Obesidade/genética , MicroRNAs/genética , Lipídeos
2.
Sci Rep ; 12(1): 18506, 2022 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-36323770

RESUMO

SARS coronavirus 2 (SARS-CoV-2) has caused an ongoing global pandemic with significant mortality and morbidity. At this time, the only FDA-approved therapeutic for COVID-19 is remdesivir, a broad-spectrum antiviral nucleoside analog. Efficacy is only moderate, and improved treatment strategies are urgently needed. To accomplish this goal, we devised a strategy to identify compounds that act synergistically with remdesivir in preventing SARS-CoV-2 replication. We conducted combinatorial high-throughput screening in the presence of submaximal remdesivir concentrations, using a human lung epithelial cell line infected with a clinical isolate of SARS-CoV-2. This identified 20 approved drugs that act synergistically with remdesivir, many with favorable pharmacokinetic and safety profiles. Strongest effects were observed with established antivirals, Hepatitis C virus nonstructural protein 5A (HCV NS5A) inhibitors velpatasvir and elbasvir. Combination with their partner drugs sofosbuvir and grazoprevir further increased efficacy, increasing remdesivir's apparent potency > 25-fold. We report that HCV NS5A inhibitors act on the SARS-CoV-2 exonuclease proofreader, providing a possible explanation for the synergy observed with nucleoside analog remdesivir. FDA-approved Hepatitis C therapeutics Epclusa® (velpatasvir/sofosbuvir) and Zepatier® (elbasvir/grazoprevir) could be further optimized to achieve potency and pharmacokinetic properties that support clinical evaluation in combination with remdesivir.


Assuntos
Tratamento Farmacológico da COVID-19 , Hepatite C , Humanos , SARS-CoV-2 , Antivirais/uso terapêutico , Sofosbuvir/farmacologia , Nucleosídeos/farmacologia , Monofosfato de Adenosina , Alanina , Hepacivirus , Hepatite C/tratamento farmacológico , Pulmão
3.
Cancer Discov ; 11(3): 678-695, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33203734

RESUMO

Circulating tumor cells (CTC) are shed by cancer into the bloodstream, where a viable subset overcomes oxidative stress to initiate metastasis. We show that single CTCs from patients with melanoma coordinately upregulate lipogenesis and iron homeostasis pathways. These are correlated with both intrinsic and acquired resistance to BRAF inhibitors across clonal cultures of BRAF-mutant CTCs. The lipogenesis regulator SREBP2 directly induces transcription of the iron carrier Transferrin (TF), reducing intracellular iron pools, reactive oxygen species, and lipid peroxidation, thereby conferring resistance to inducers of ferroptosis. Knockdown of endogenous TF impairs tumor formation by melanoma CTCs, and their tumorigenic defects are partially rescued by the lipophilic antioxidants ferrostatin-1 and vitamin E. In a prospective melanoma cohort, presence of CTCs with high lipogenic and iron metabolic RNA signatures is correlated with adverse clinical outcome, irrespective of treatment regimen. Thus, SREBP2-driven iron homeostatic pathways contribute to cancer progression, drug resistance, and metastasis. SIGNIFICANCE: Through single-cell analysis of primary and cultured melanoma CTCs, we have uncovered intrinsic cancer cell heterogeneity within lipogenic and iron homeostatic pathways that modulates resistance to BRAF inhibitors and to ferroptosis inducers. Activation of these pathways within CTCs is correlated with adverse clinical outcome, pointing to therapeutic opportunities.This article is highlighted in the In This Issue feature, p. 521.


Assuntos
Ferroptose/genética , Lipogênese/genética , Melanoma/genética , Melanoma/metabolismo , Células Neoplásicas Circulantes/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 2/genética , Transferrina/metabolismo , Biomarcadores Tumorais , Células Cultivadas , Suscetibilidade a Doenças , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Melanoma/patologia , Mutação , Células Neoplásicas Circulantes/patologia , Transdução de Sinais , Análise de Célula Única , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo
4.
Sci Rep ; 9(1): 10369, 2019 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-31316083

RESUMO

de novo fatty acid biosynthesis (DNFA) is a hallmark adaptation of many cancers that supports survival, proliferation, and metastasis. Here we elucidate previously unexplored aspects of transcription regulation and clinical relevance of DNFA in cancers. We show that elevated expression of DNFA genes is characteristic of many tumor types and correlates with poor prognosis, especially in melanomas. Elevated DNFA gene expression depends on the SREBP1 transcription factor in multiple melanoma cell lines. SREBP1 predominantly binds to the transcription start sites of DNFA genes, regulating their expression by recruiting RNA polymerase II to promoters for productive transcription elongation. We find that SREBP1-regulated DNFA represents a survival trait in melanoma cells, regardless of proliferative state and oncogenic mutation status. Indeed, malignant melanoma cells exhibit elevated DNFA gene expression after the BRAF/MEK signaling pathway is blocked (e.g. by BRAF inhibitors), and DNFA expression remains higher in melanoma cells resistant to vemurafenib treatment than in untreated cells. Accordingly, DNFA pathway inhibition, whether by direct targeting of SREBP1 with antisense oligonucleotides, or through combinatorial effects of multiple DNFA enzyme inhibitors, exerts potent cytotoxic effects on both BRAFi-sensitive and -resistant melanoma cells. Altogether, these results implicate SREBP1 and DNFA enzymes as enticing therapeutic targets in melanomas.


Assuntos
Ácidos Graxos/biossíntese , Regulação Neoplásica da Expressão Gênica , Melanoma/metabolismo , Proteínas de Neoplasias/fisiologia , Neoplasias Cutâneas/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Indazóis/farmacologia , Estimativa de Kaplan-Meier , Melanoma/genética , Melanoma/mortalidade , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias/genética , Piperazinas/farmacologia , Prognóstico , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Interferência de RNA , RNA Polimerase II/metabolismo , RNA Interferente Pequeno/genética , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/mortalidade , Transcrição Gênica , Vemurafenib/farmacologia
5.
Cell ; 177(3): 722-736.e22, 2019 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-30955890

RESUMO

Insulin receptor (IR) signaling is central to normal metabolic control and dysregulated in prevalent chronic diseases. IR binds insulin at the cell surface and transduces rapid signaling via cytoplasmic kinases. However, mechanisms mediating long-term effects of insulin remain unclear. Here, we show that IR associates with RNA polymerase II in the nucleus, with striking enrichment at promoters genome-wide. The target genes were highly enriched for insulin-related functions including lipid metabolism and protein synthesis and diseases including diabetes, neurodegeneration, and cancer. IR chromatin binding was increased by insulin and impaired in an insulin-resistant disease model. Promoter binding by IR was mediated by coregulator host cell factor-1 (HCF-1) and transcription factors, revealing an HCF-1-dependent pathway for gene regulation by insulin. These results show that IR interacts with transcriptional machinery at promoters and identify a pathway regulating genes linked to insulin's effects in physiology and disease.


Assuntos
Regulação da Expressão Gênica , Estudo de Associação Genômica Ampla , Receptor de Insulina/metabolismo , Animais , Linhagem Celular Tumoral , Cromatina/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Fator C1 de Célula Hospedeira/antagonistas & inibidores , Fator C1 de Célula Hospedeira/genética , Fator C1 de Célula Hospedeira/metabolismo , Humanos , Insulina/metabolismo , Insulina/farmacologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Ligação Proteica , Subunidades Proteicas/metabolismo , Interferência de RNA , RNA Polimerase II/metabolismo , RNA Interferente Pequeno/metabolismo , Receptor de Insulina/química , Transdução de Sinais/efeitos dos fármacos
6.
PLoS One ; 14(4): e0215022, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30970006

RESUMO

While investigating the role played by de novo lipid (DNL) biosynthesis in cancer cells, we sought a medium condition that would support cell proliferation without providing any serum lipids. Here we report that a defined serum free cell culture medium condition containing insulin, transferrin and selenium (ITS) supports controlled study of transcriptional regulation of de novo fatty acid (DNFA) production and de novo cholesterol synthesis (DNCS) in melanoma cell lines. This lipid-free ITS medium is able to support continuous proliferation of several melanoma cell lines that utilize DNL to support their lipid requirements. We show that the ITS medium stimulates gene transcription in support of both DNFA and DNCS, specifically mediated by SREBP1/2 in melanoma cells. We further found that the ITS medium promoted SREBP1 nuclear localization and occupancy on DNFA gene promoters. Our data show clear utility of this serum and lipid-free medium for melanoma cancer cell culture and lipid-related areas of investigation.


Assuntos
Ácidos Graxos/biossíntese , Insulina/farmacologia , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteína de Ligação a Elemento Regulador de Esterol 2/genética , Ativação Transcricional/efeitos dos fármacos , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Colesterol/biossíntese , Meios de Cultura/química , Humanos , Melanoma/metabolismo , Melanoma/patologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Selênio/farmacologia , Proteína de Ligação a Elemento Regulador de Esterol 1/antagonistas & inibidores , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 2/antagonistas & inibidores , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo , Transferrina/farmacologia
7.
J Biol Chem ; 291(37): 19651-60, 2016 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-27471270

RESUMO

MicroRNAs (miRNAs) are short non-coding RNAs that regulate gene expression by promoting degradation and/or repressing translation of specific target mRNAs. Several miRNAs have been identified that regulate the amplitude of the innate immune response by directly targeting Toll-like receptor (TLR) pathway members and/or cytokines. miR-33a and miR-33b (the latter present in primates but absent in rodents and lower species) are located in introns of the sterol regulatory element-binding protein (SREBP)-encoding genes and control cholesterol/lipid homeostasis in concert with their host gene products. These miRNAs regulate macrophage cholesterol by targeting the lipid efflux transporters ATP binding cassette (ABC)A1 and ABCG1. We and others have previously reported that Abca1(-/-) and Abcg1(-/-) macrophages have increased TLR proinflammatory responses due to augmented lipid raft cholesterol. Given this, we hypothesized that miR-33 would augment TLR signaling in macrophages via a raft cholesterol-dependent mechanism. Herein, we report that multiple TLR ligands down-regulate miR-33 in murine macrophages. In the case of lipopolysaccharide, this is a delayed, Toll/interleukin-1 receptor (TIR) domain-containing adapter-inducing interferon-ß-dependent response that also down-regulates Srebf-2, the host gene for miR-33. miR-33 augments macrophage lipid rafts and enhances proinflammatory cytokine induction and NF-κB activation by LPS. This occurs through an ABCA1- and ABCG1-dependent mechanism and is reversible by interventions upon raft cholesterol and by ABC transporter-inducing liver X receptor agonists. Taken together, these findings extend the purview of miR-33, identifying it as an indirect regulator of innate immunity that mediates bidirectional cross-talk between lipid homeostasis and inflammation.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/imunologia , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/imunologia , Imunidade Inata , Macrófagos/imunologia , Microdomínios da Membrana/imunologia , MicroRNAs/imunologia , Transportador 1 de Cassete de Ligação de ATP/genética , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Animais , Microdomínios da Membrana/genética , Camundongos , Camundongos Knockout , MicroRNAs/genética , Células RAW 264.7 , Proteína de Ligação a Elemento Regulador de Esterol 2/genética , Proteína de Ligação a Elemento Regulador de Esterol 2/imunologia
8.
Nat Med ; 21(11): 1280-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26437365

RESUMO

The hepatic low-density lipoprotein receptor (LDLR) pathway is essential for clearing circulating LDL cholesterol (LDL-C). Whereas the transcriptional regulation of LDLR is well characterized, the post-transcriptional mechanisms that govern LDLR expression are just beginning to emerge. Here we develop a high-throughput genome-wide screening assay to systematically identify microRNAs (miRNAs) that regulate LDLR activity in human hepatic cells. From this screen we identified and characterized miR-148a as a negative regulator of LDLR expression and activity and defined a sterol regulatory element-binding protein 1 (SREBP1)-mediated pathway through which miR-148a regulates LDL-C uptake. In mice, inhibition of miR-148a increased hepatic LDLR expression and decreased plasma LDL-C. Moreover, we found that miR-148a regulates hepatic expression of ATP-binding cassette, subfamily A, member 1 (ABCA1) and circulating high-density lipoprotein cholesterol (HDL-C) levels in vivo. These studies uncover a role for miR-148a as a key regulator of hepatic LDL-C clearance through direct modulation of LDLR expression and demonstrate the therapeutic potential of inhibiting miR-148a to ameliorate an elevated LDL-C/HDL-C ratio, a prominent risk factor for cardiovascular disease.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/genética , HDL-Colesterol/metabolismo , LDL-Colesterol/metabolismo , Hepatócitos/metabolismo , Fígado/metabolismo , MicroRNAs/genética , Receptores de LDL/genética , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Animais , Regulação da Expressão Gênica , Células Hep G2 , Ensaios de Triagem em Larga Escala , Humanos , Camundongos , MicroRNAs/metabolismo , Processamento Pós-Transcricional do RNA , Receptores de LDL/metabolismo , Transdução de Sinais , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo
9.
Nat Commun ; 6: 7505, 2015 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-26106036

RESUMO

Although BRCA1 function is essential for maintaining genomic integrity in all cell types, it is unclear why increased risk of cancer in individuals harbouring deleterious mutations in BRCA1 is restricted to only a select few tissues. Here we show that human mammary epithelial cells (HMECs) from BRCA1-mutation carriers (BRCA1(mut/+)) exhibit increased genomic instability and rapid telomere erosion in the absence of tumour-suppressor loss. Furthermore, we uncover a novel form of haploinsufficiency-induced senescence (HIS) specific to epithelial cells, which is triggered by pRb pathway activation rather than p53 induction. HIS and telomere erosion in HMECs correlate with misregulation of SIRT1 leading to increased levels of acetylated pRb as well as acetylated H4K16 both globally and at telomeric regions. These results identify a novel form of cellular senescence and provide a potential molecular basis for the rapid cell- and tissue- specific predisposition of breast cancer development associated with BRCA1 haploinsufficiency.


Assuntos
Senescência Celular/genética , Células Epiteliais/metabolismo , Genes BRCA1 , Instabilidade Genômica/genética , Haploinsuficiência , Glândulas Mamárias Humanas/metabolismo , Encurtamento do Telômero/genética , Dano ao DNA , Células Epiteliais/citologia , Heterozigoto , Humanos , Glândulas Mamárias Humanas/citologia , Mutação , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Sirtuína 1/genética , Sirtuína 1/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
10.
Sci Transl Med ; 5(212): 212ra162, 2013 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-24259050

RESUMO

MicroRNAs (miRNAs) regulate many aspects of human biology. They target mRNAs for translational repression or degradation through base pairing with 3' untranslated regions, primarily via seed sequences (nucleotides 2 to 8 in the mature miRNA sequence). A number of individual miRNAs and miRNA families share seed sequences and targets, but differ in the sequences outside of the seed. miRNAs have been implicated in the etiology of a wide variety of human diseases and therefore represent promising therapeutic targets. However, potential redundancy of different miRNAs sharing the same seed sequence and the challenge of simultaneously targeting miRNAs that differ significantly in nonseed sequences complicate therapeutic targeting approaches. We recently demonstrated effective inhibition of entire miRNA families using seed-targeting 8-mer locked nucleic acid (LNA)-modified antimiRs in short-term experiments in mammalian cells and in mice. However, the long-term efficacy and safety of this approach in higher organisms, such as humans and nonhuman primates, have not been determined. We show that pharmacological inhibition of the miR-33 family, key regulators of cholesterol/lipid homeostasis, by a subcutaneously delivered 8-mer LNA-modified antimiR in obese and insulin-resistant nonhuman primates results in derepression of miR-33 targets, such as ABCA1, increases circulating high-density lipoprotein cholesterol, and is well tolerated over 108 days of treatment. These findings demonstrate the efficacy and safety of an 8-mer LNA-antimiR against an miRNA family in a nonhuman primate metabolic disease model, suggesting that this could be a feasible approach for therapeutic targeting of miRNA families sharing the same seed sequence in human diseases.


Assuntos
Inativação Gênica , MicroRNAs/antagonistas & inibidores , Animais , HDL-Colesterol/sangue , Feminino , Células Hep G2 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Primatas
11.
Mol Cell ; 51(4): 454-68, 2013 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-23911928

RESUMO

DNA damage is linked to multiple human diseases, such as cancer, neurodegeneration, and aging. Little is known about the role of chromatin accessibility in DNA repair. Here, we find that the deacetylase sirtuin 6 (SIRT6) is one of the earliest factors recruited to double-strand breaks (DSBs). SIRT6 recruits the chromatin remodeler SNF2H to DSBs and focally deacetylates histone H3K56. Lack of SIRT6 and SNF2H impairs chromatin remodeling, increasing sensitivity to genotoxic damage and recruitment of downstream factors such as 53BP1 and breast cancer 1 (BRCA1). Remarkably, SIRT6-deficient mice exhibit lower levels of chromatin-associated SNF2H in specific tissues, a phenotype accompanied by DNA damage. We demonstrate that SIRT6 is critical for recruitment of a chromatin remodeler as an early step in the DNA damage response, indicating that proper unfolding of chromatin plays a rate-limiting role. We present a unique crosstalk between a histone modifier and a chromatin remodeler, regulating a coordinated response to prevent DNA damage.


Assuntos
Adenosina Trifosfatases/metabolismo , Montagem e Desmontagem da Cromatina , Cromatina/genética , Proteínas Cromossômicas não Histona/metabolismo , Dano ao DNA/genética , Reparo do DNA/genética , Instabilidade Genômica , Sirtuínas/metabolismo , Sirtuínas/fisiologia , Adenosina Trifosfatases/genética , Animais , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Imunoprecipitação da Cromatina , Proteínas Cromossômicas não Histona/genética , Hipocampo/citologia , Hipocampo/metabolismo , Histonas/metabolismo , Humanos , Imunoprecipitação , Camundongos , Camundongos Knockout , Nucleossomos/metabolismo , Sirtuínas/genética , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
12.
Cell ; 147(4): 840-52, 2011 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-22035958

RESUMO

Sterol regulatory element-binding proteins (SREBPs) activate genes involved in the synthesis and trafficking of cholesterol and other lipids and are critical for maintaining lipid homeostasis. Aberrant SREBP activity, however, can contribute to obesity, fatty liver disease, and insulin resistance, hallmarks of metabolic syndrome. Our studies identify a conserved regulatory circuit in which SREBP-1 controls genes in the one-carbon cycle, which produces the methyl donor S-adenosylmethionine (SAMe). Methylation is critical for the synthesis of phosphatidylcholine (PC), a major membrane component, and we find that blocking SAMe or PC synthesis in C. elegans, mouse liver, and human cells causes elevated SREBP-1-dependent transcription and lipid droplet accumulation. Distinct from negative regulation of SREBP-2 by cholesterol, our data suggest a feedback mechanism whereby maturation of nuclear, transcriptionally active SREBP-1 is controlled by levels of PC. Thus, nutritional or genetic conditions limiting SAMe or PC production may activate SREBP-1, contributing to human metabolic disorders.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular Tumoral , Retículo Endoplasmático/metabolismo , Humanos , Lipogênese , Camundongos , Modelos Animais , Fosfatidilcolinas/biossíntese , Interferência de RNA , S-Adenosilmetionina/biossíntese
13.
Genes Dev ; 25(1): 17-28, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21205864

RESUMO

Dynamic regulation of histone modifications is critical during development, and aberrant activity of chromatin-modifying enzymes has been associated with diseases such as cancer. Histone demethylases have been shown to play a key role in eukaryotic gene transcription; however, little is known about how their activities are coordinated in vivo to regulate specific biological processes. In Drosophila, two enzymes, dLsd1 (Drosophila ortholog of lysine-specific demethylase 1) and Lid (little imaginal discs), demethylate histone H3 at Lys 4 (H3K4), a residue whose methylation is associated with actively transcribed genes. Our studies show that compound mutation of Lid and dLsd1 results in increased H3K4 methylation levels. However, unexpectedly, Lid mutations strongly suppress dLsd1 mutant phenotypes. Investigation of the basis for this antagonism revealed that Lid opposes the functions of dLsd1 and the histone methyltransferase Su(var)3-9 in promoting heterochromatin spreading at heterochromatin-euchromatin boundaries. Moreover, our data reveal a novel role for dLsd1 in Notch signaling in Drosophila, and a complex network of interactions between dLsd1, Lid, and Notch signaling at euchromatic genes. These findings illustrate the complexity of functional interplay between histone demethylases in vivo, providing insights into the epigenetic regulation of heterochromatin/euchromatin boundaries by Lid and dLsd1 and showing their involvement in Notch pathway-specific control of gene expression in euchromatin.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimologia , Histona Desmetilases/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Oxirredutases N-Desmetilantes/metabolismo , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Regulação da Expressão Gênica , Heterocromatina/metabolismo , Histona-Lisina N-Metiltransferase/genética , Histonas/metabolismo , Metilação , Mutação/genética , Oxirredutases N-Desmetilantes/genética , Fenótipo , Receptores Notch/genética , Transdução de Sinais
14.
Science ; 328(5985): 1566-9, 2010 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-20466882

RESUMO

Proper coordination of cholesterol biosynthesis and trafficking is essential to human health. The sterol regulatory element-binding proteins (SREBPs) are key transcription regulators of genes involved in cholesterol biosynthesis and uptake. We show here that microRNAs (miR-33a/b) embedded within introns of the SREBP genes target the adenosine triphosphate-binding cassette transporter A1 (ABCA1), an important regulator of high-density lipoprotein (HDL) synthesis and reverse cholesterol transport, for posttranscriptional repression. Antisense inhibition of miR-33 in mouse and human cell lines causes up-regulation of ABCA1 expression and increased cholesterol efflux, and injection of mice on a western-type diet with locked nucleic acid-antisense oligonucleotides results in elevated plasma HDL. Our findings indicate that miR-33 acts in concert with the SREBP host genes to control cholesterol homeostasis and suggest that miR-33 may represent a therapeutic target for ameliorating cardiometabolic diseases.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , HDL-Colesterol/sangue , Colesterol/metabolismo , MicroRNAs/metabolismo , Proteínas de Ligação a Elemento Regulador de Esterol/genética , Regiões 3' não Traduzidas , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Linhagem Celular , Dieta , Regulação da Expressão Gênica , Homeostase , Humanos , Íntrons , Fígado/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Oligonucleotídeos Antissenso/farmacologia , Interferência de RNA , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 2/genética , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo , Proteínas de Ligação a Elemento Regulador de Esterol/metabolismo , Regulação para Cima
15.
Nature ; 455(7212): 552-6, 2008 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-18794899

RESUMO

The E2F1 transcription factor can promote proliferation or apoptosis when activated, and is a key downstream target of the retinoblastoma tumour suppressor protein (pRB). Here we show that E2F1 is a potent and specific inhibitor of beta-catenin/T-cell factor (TCF)-dependent transcription, and that this function contributes to E2F1-induced apoptosis. E2F1 deregulation suppresses beta-catenin activity in an adenomatous polyposis coli (APC)/glycogen synthase kinase-3 (GSK3)-independent manner, reducing the expression of key beta-catenin targets including c-MYC. This interaction explains why colorectal tumours, which depend on beta-catenin transcription for their abnormal proliferation, keep RB1 intact. Remarkably, E2F1 activity is also repressed by cyclin-dependent kinase-8 (CDK8), a colorectal oncoprotein. Elevated levels of CDK8 protect beta-catenin/TCF-dependent transcription from inhibition by E2F1. Thus, by retaining RB1 and amplifying CDK8, colorectal tumour cells select conditions that collectively suppress E2F1 and enhance the activity of beta-catenin.


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Fator de Transcrição E2F1/antagonistas & inibidores , Fator de Transcrição E2F1/metabolismo , Proteína do Retinoblastoma/metabolismo , Transcrição Gênica , beta Catenina/antagonistas & inibidores , beta Catenina/metabolismo , Proteína da Polipose Adenomatosa do Colo/metabolismo , Apoptose , Linhagem Celular , Quinase 8 Dependente de Ciclina , Regulação da Expressão Gênica , Genes myc/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Proteína do Retinoblastoma/genética , Transdução de Sinais , Fatores de Transcrição TCF/metabolismo , Proteínas Wnt/metabolismo
16.
Nature ; 452(7187): 604-9, 2008 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-18385733

RESUMO

Multidrug resistance (MDR) is a serious complication during treatment of opportunistic fungal infections that frequently afflict immunocompromised individuals, such as transplant recipients and cancer patients undergoing cytotoxic chemotherapy. Improved knowledge of the molecular pathways controlling MDR in pathogenic fungi should facilitate the development of novel therapies to combat these intransigent infections. MDR is often caused by upregulation of drug efflux pumps by members of the fungal zinc-cluster transcription-factor family (for example Pdr1p orthologues). However, the molecular mechanisms are poorly understood. Here we show that Pdr1p family members in Saccharomyces cerevisiae and the human pathogen Candida glabrata directly bind to structurally diverse drugs and xenobiotics, resulting in stimulated expression of drug efflux pumps and induction of MDR. Notably, this is mechanistically similar to regulation of MDR in vertebrates by the PXR nuclear receptor, revealing an unexpected functional analogy of fungal and metazoan regulators of MDR. We have also uncovered a critical and specific role of the Gal11p/MED15 subunit of the Mediator co-activator and its activator-targeted KIX domain in antifungal/xenobiotic-dependent regulation of MDR. This detailed mechanistic understanding of a fungal nuclear receptor-like gene regulatory pathway provides novel therapeutic targets for the treatment of multidrug-resistant fungal infections.


Assuntos
Candida glabrata/metabolismo , Farmacorresistência Fúngica , Proteínas Fúngicas/metabolismo , Regulação Fúngica da Expressão Gênica , Receptores de Esteroides/metabolismo , Saccharomyces cerevisiae/metabolismo , Animais , Antifúngicos/metabolismo , Antifúngicos/farmacologia , Candida glabrata/efeitos dos fármacos , Candida glabrata/genética , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Farmacorresistência Fúngica/genética , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Regulação Fúngica da Expressão Gênica/genética , Genes Fúngicos/genética , Complexo Mediador , Família Multigênica , Receptor de Pregnano X , Estrutura Terciária de Proteína , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Transativadores/química , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica/genética , Xenobióticos/metabolismo
17.
J Biol Chem ; 282(44): 32152-7, 2007 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-17848560

RESUMO

Magicin, a protein that we isolated earlier as an interactor of the neurofibromatosis 2 protein merlin, was independently identified as MED28, a subunit of the mammalian Mediator complex. Mediator complex is an evolutionarily conserved transcriptional cofactor, which plays an essential role in positive and negative gene regulation. Distinct Mediator subunit composition is thought to contribute to gene regulation specificity based on the interaction of specific subunits with subsets of transcription factors. Here we report that down-regulation of Med28 expression in NIH3T3 cells results in a significant induction of several genes associated with smooth muscle cell (SMC) differentiation. Conversely, overexpression of MED28 represses expression of SMC genes, in concordance with our knockdown data. More importantly, multipotent mesenchymal-derived murine precursors can transdifferentiate into SMCs when Med28 is down-regulated. Our data also show that Med28 functions as a negative regulator of SMC differentiation in concert with other Mediator subunits including Med6, Med8, and Med18 within the Mediator head module. Our results provide strong evidence that MED28 may function as a scaffolding protein by maintaining the stability of a submodule within the head module and that components of this submodule act together in a gene regulatory program to suppress SMC differentiation. The results presented here demonstrate for the first time that the mammalian Mediator subunit MED28 functions as a repressor of SMC differentiation, which could have implications for disorders associated with abnormalities in SMC growth and differentiation, including atherosclerosis, asthma, hypertension, and smooth muscle tumors.


Assuntos
Transdiferenciação Celular , Proteínas do Citoesqueleto/metabolismo , Fibroblastos/citologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Miócitos de Músculo Liso/citologia , Animais , Proteínas do Citoesqueleto/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Complexo Mediador , Camundongos , Células NIH 3T3 , Interferência de RNA , Regulação para Cima
18.
Nat Cell Biol ; 9(2): 225-32, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17187060

RESUMO

The retinoblastoma protein (pRB) negatively regulates the progression from G1 to S phase of the cell cycle, in part, by repressing E2F-dependent transcription. pRB also possesses E2F-independent functions that contribute to cell-cycle control--for example, during pRB-mediated cell-cycle arrest pRB associates with Skp2, the F-box protein of the Skp1-Cullin-F-box protein (SCF) E3 ubiquitin ligase complex, and promotes the stability of the cyclin-dependent kinase-inhibitor p27(Kip1) through an unknown mechanism. Degradation of p27(Kip1) is mediated by ubiquitin-dependent targeting of p27(Kip1) by SCF -Skp2 (ref. 4). Here, we report a novel interaction between pRB and the anaphase-promoting complex/cyclosome (APC/C) that controls p27(Kip1) stability by targeting Skp2 for ubiquitin-mediated degradation. Cdh1, an activator of APC/C, not only interacts with pRB but is also required for a pRB-induced cell-cycle arrest. The results reveal an unexpected physical convergence between the pRB tumour-suppressor protein and E3 ligase complexes, and raise the possibility that pRB may direct APC/C to additional targets during pRB-mediated cell-cycle exit.


Assuntos
Anáfase , Proteína do Retinoblastoma/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Ciclossomo-Complexo Promotor de Anáfase , Caderinas/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Fase G1 , Humanos , Fase S , Proteínas Quinases Associadas a Fase S/metabolismo , Ubiquitina/metabolismo
19.
Nature ; 418(6898): 641-6, 2002 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-12167862

RESUMO

The transforming growth factor beta (TGF beta) family of cytokines, including Nodal, Activin and bone morphogenetic protein (BMP), have essential roles in development and tumorigenesis. TGF beta molecules activate the Smad family of signal transducers, which form complexes with specific DNA-binding proteins to regulate gene expression. Two discrete Smad-dependent signalling pathways have been identified: TGF beta, Activin and Nodal signal via the Smad2 (or Smad3)-Smad4 complex, whereas BMP signals via the Smad1-Smad4 complex. How distinct Smad complexes regulate specific gene expression is not fully understood. Here we show that ARC105, a component of the activator-recruited co-factor (ARC) complex or the metazoan Mediator complex, is essential for TGF beta/Activin/Nodal/Smad2/3 signal transduction. Expression of ARC105 stimulates Activin/Nodal/Smad2 signalling in Xenopus laevis embryos, inducing axis duplication and mesendoderm differentiation, and enhances TGF beta response in human cells. Depletion of ARC105 inhibits TGF beta/Activin/Nodal/Smad2/3 signalling and Xenopus axis formation, but not BMP/Smad1 signalling. ARC105 protein binds to Smad2/3-Smad4 in response to TGF beta and is recruited to Activin/Nodal-responsive promoters in chromatin in a Smad2-dependent fashion. Thus ARC105 is a specific and key ARC/Mediator component linking TGF beta/Activin/Nodal/Smad2/3 signalling to transcriptional activation.


Assuntos
Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Proteínas de Xenopus/química , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriologia , Xenopus laevis/metabolismo , Ativinas/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Substâncias Macromoleculares , Mesoderma/citologia , Mesoderma/efeitos dos fármacos , Mesoderma/metabolismo , Fatores de Crescimento Neural , Proteína Nodal , Regiões Promotoras Genéticas/genética , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Smad , Proteína Smad2 , Proteína Smad3 , Proteína Smad4 , Transativadores/metabolismo , Xenopus laevis/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA