Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
AAPS J ; 24(3): 45, 2022 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-35314909

RESUMO

Inhibitory effects of asunaprevir, daclatasvir, grazoprevir, paritaprevir, simeprevir, and voxilaprevir, direct-acting antiviral (DAA) drugs for the treatment of chronic hepatitis C virus (HCV) infection, were evaluated in vitro against a range of clinically important drug transporters. In vitro inhibition studies were conducted using transporter transfected cells and membrane vesicles. The risk of clinical drug-drug interactions (DDIs) was assessed using simplified static models recommended by regulatory agencies. Furthermore, we refined and developed static models to predict complex DDIs with several statins (pitavastatin, rosuvastatin, atorvastatin, and pravastatin) by mechanistically assessing differential inhibitory effects of perpetrator drugs on multiple transporters, such as organic anion transporting polypeptides (OATP1B), breast cancer resistance protein (BCRP), multidrug resistance protein 2 (MRP2), organic anion transporter 3 (OAT3), and cytochrome P450 CYP3A enzyme, as they are known to contribute to absorption, distribution, metabolism and excretion (ADME) of above statins. These models successfully predicted a total of 46 statin DDIs, including above DAA drugs and their fix-dose combination regimens. Predicted plasma area under curve ratio (AUCR) with and without perpetrator drugs was within ~ 2-fold of observed values. In contrast, simplified static R-value model resulted in increased false negative and false positive predictions when different prediction cut-off values were applied. Our studies suggest that mechanistic static model is a promising and useful tool to provide more accurate prediction of the risk and magnitude of DDIs with statins in early drug development and may help to improve the management of clinical DDIs for HCV drugs to ensure effective and safe HCV therapy. GRAPHICAL ABSTRACT.


Assuntos
Hepatite C Crônica , Inibidores de Hidroximetilglutaril-CoA Redutases , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Antivirais , Interações Medicamentosas , Hepacivirus/metabolismo , Hepatite C Crônica/tratamento farmacológico , Humanos , Proteínas de Membrana Transportadoras/metabolismo , Proteínas de Neoplasias/metabolismo
2.
Viruses ; 13(8)2021 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-34452431

RESUMO

Islatravir (MK-8591) is a nucleoside reverse transcriptase translocation inhibitor in development for the treatment and prevention of HIV-1. The potential for islatravir to interact with commonly co-prescribed medications was studied in vitro. Elimination of islatravir is expected to be balanced between adenosine deaminase-mediated metabolism and renal excretion. Islatravir did not inhibit uridine diphosphate glucuronosyltransferase 1A1 or cytochrome p450 (CYP) enzymes CYP1A2, 2B6, 2C8, 2C9, 2C19, 2D6, or 3A4, nor did it induce CYP1A2, 2B6, or 3A4. Islatravir did not inhibit hepatic transporters organic anion transporting polypeptide (OATP) 1B1, OATP1B3, organic cation transporter (OCT) 1, bile salt export pump (BSEP), multidrug resistance-associated protein (MRP) 2, MRP3, or MRP4. Islatravir was neither a substrate nor a significant inhibitor of renal transporters organic anion transporter (OAT) 1, OAT3, OCT2, multidrug and toxin extrusion protein (MATE) 1, or MATE2K. Islatravir did not significantly inhibit P-glycoprotein and breast cancer resistance protein (BCRP); however, it was a substrate of BCRP, which is not expected to be of clinical significance. These findings suggest islatravir is unlikely to be the victim or perpetrator of drug-drug interactions with commonly co-prescribed medications, including statins, diuretics, anti-diabetic drugs, proton pump inhibitors, anticoagulants, benzodiazepines, and selective serotonin reuptake inhibitors.


Assuntos
Desoxiadenosinas/metabolismo , Interações Medicamentosas , Preparações Farmacêuticas/metabolismo , Inibidores da Transcriptase Reversa/metabolismo , Animais , Transporte Biológico , Sistema Enzimático do Citocromo P-450/metabolismo , Desoxiadenosinas/sangue , Cães , Infecções por HIV/tratamento farmacológico , Humanos , Técnicas In Vitro , Células Madin Darby de Rim Canino , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Transportadores de Ânions Orgânicos/metabolismo , Coelhos
3.
J Antimicrob Chemother ; 74(7): 1894-1903, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30891606

RESUMO

OBJECTIVES: To identify the transporters involved in renal elimination of relebactam, and to assess the potential of relebactam as a perpetrator or victim of drug-drug interactions (DDIs) for major drug transporters. METHODS: A series of bidirectional transport, uptake and inhibition studies were conducted in vitro using transfected cell lines and membrane vesicles. The inhibitory effects of relebactam on major drug transporters, as well as the inhibitory effects of commonly used antibiotics/antifungals on organic anion transporter (OAT) 3-mediated uptake of relebactam, were assessed. RESULTS: Relebactam was shown to be a substrate of OAT3, OAT4, and multidrug and toxin extrusion (MATE) proteins MATE1 and MATE2K. Relebactam did not show profound inhibition across a panel of transporters, including organic anion-transporting polypeptides 1B1 and 1B3, OAT1, OAT3, organic cation transporter 2, MATE1, MATE2K, breast cancer resistance protein, multidrug resistance protein 1 and the bile salt export pump. Among the antibiotics/antifungals assessed for potential DDIs, probenecid demonstrated the most potent in vitro inhibition of relebactam uptake; however, such in vitro data did not translate into clinically relevant DDIs, suggesting that relebactam can be co-administered with OAT inhibitors, such as probenecid. CONCLUSIONS: Overall, relebactam has low potential to be a victim or perpetrator of DDIs with major drug transporters.


Assuntos
Compostos Azabicíclicos/farmacocinética , Transporte Biológico , Rim/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Inibidores de beta-Lactamases/farmacocinética , Animais , Linhagem Celular , Vesículas Extracelulares , Humanos , Modelos Biológicos
4.
Artigo em Inglês | MEDLINE | ID: mdl-30745395

RESUMO

Doravirine is a novel nonnucleoside reverse transcriptase inhibitor for the treatment of human immunodeficiency virus type 1 infection. In vitro studies were conducted to assess the potential for drug interactions with doravirine via major drug-metabolizing enzymes and transporters. Kinetic studies confirmed that cytochrome P450 3A (CYP3A) plays a major role in the metabolism of doravirine, with ∼20-fold-higher catalytic efficiency for CYP3A4 versus CYP3A5. Doravirine was not a substrate of breast cancer resistance protein (BCRP) and likely not a substrate of organic anion transporting polypeptide 1B1 (OATP1B1) or OATP1B3. Doravirine was not a reversible inhibitor of major CYP enzymes (CYP1A2, -2B6, -2C8, -2C9, -2C19, -2D6, and -3A4) or of UGT1A1, nor was it a time-dependent inhibitor of CYP3A4. No induction of CYP1A2 or -2B6 was observed in cultured human hepatocytes; small increases in CYP3A4 mRNA (≤20%) were reported at doravirine concentrations of ≥10 µM but with no corresponding increase in enzyme activity. In vitro transport studies indicated a low potential for interactions with substrates of BCRP, P-glycoprotein, OATP1B1 and OATP1B3, the bile salt extrusion pump (BSEP), organic anion transporter 1 (OAT1) and OAT3, organic cation transporter 2 (OCT2), and multidrug and toxin extrusion 1 (MATE1) and MATE2K proteins. In summary, these in vitro findings indicate that CYP3A4 and CYP3A5 mediate the metabolism of doravirine, although with different catalytic efficiencies. Clinical trials reported elsewhere confirm that doravirine is subject to drug-drug interactions (DDIs) via CYP3A inhibitors and inducers, but they support the notion that DDIs (either direction) are unlikely via other major drug-metabolizing enzymes and transporters.


Assuntos
Interações Medicamentosas/fisiologia , Piridonas/metabolismo , Triazóis/metabolismo , Animais , Transporte Biológico/fisiologia , Linhagem Celular , Citocromo P-450 CYP3A/metabolismo , Cães , Células HEK293 , Hepatócitos/metabolismo , Humanos , Cinética , Células Madin Darby de Rim Canino , Proteínas de Membrana Transportadoras/metabolismo
5.
Blood ; 120(16): 3288-97, 2012 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-22936659

RESUMO

Sézary syndrome (SS) is an aggressive cutaneous T-cell lymphoma (CTCL) of unknown etiology in which malignant cells circulate in the peripheral blood. To identify viral elements, gene fusions, and gene expression patterns associated with this lymphoma, flow cytometry was used to obtain matched pure populations of malignant Sézary cells (SCs) versus nonmalignant CD4(+) T cells from 3 patients for whole transcriptome, paired-end sequencing with an average depth of 112 million reads per sample. Pathway analysis of differentially expressed genes identified mis-regulation of PI3K/Akt, TGFß, and NF-κB pathways as well as T-cell receptor signaling. Bioinformatic analysis did not detect either nonhuman transcripts to support a viral etiology of SS or recurrently expressed gene fusions, but it did identify 21 SC-associated annotated long noncoding RNAs (lncRNAs). Transcriptome assembly by multiple algorithms identified 13 differentially expressed unannotated transcripts termed Sézary cell-associated transcripts (SeCATs) that include 12 predicted lncRNAs and a novel transcript with coding potential. High-throughput sequencing targeting the 3' end of polyadenylated transcripts in archived tumors from 24 additional patients with tumor-stage CTCL confirmed the differential expression of SC-associated lncRNAs and SeCATs in CTCL. Our findings characterize the SS transcriptome and support recent reports that implicate lncRNA dysregulation in human malignancies.


Assuntos
Biomarcadores Tumorais/genética , Perfilação da Expressão Gênica , Micose Fungoide/genética , RNA Longo não Codificante/genética , Síndrome de Sézary/genética , Neoplasias Cutâneas/genética , Citometria de Fluxo , Humanos , Micose Fungoide/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Prognóstico , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Síndrome de Sézary/patologia , Neoplasias Cutâneas/patologia , Células Tumorais Cultivadas
6.
Cell ; 136(1): 62-74, 2009 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-19135889

RESUMO

Members of the sirtuin (SIRT) family of NAD-dependent deacetylases promote longevity in multiple organisms. Deficiency of mammalian SIRT6 leads to shortened life span and an aging-like phenotype in mice, but the underlying molecular mechanisms are unclear. Here we show that SIRT6 functions at chromatin to attenuate NF-kappaB signaling. SIRT6 interacts with the NF-kappaB RELA subunit and deacetylates histone H3 lysine 9 (H3K9) at NF-kappaB target gene promoters. In SIRT6-deficient cells, hyperacetylation of H3K9 at these target promoters is associated with increased RELA promoter occupancy and enhanced NF-kappaB-dependent modulation of gene expression, apoptosis, and cellular senescence. Computational genomics analyses revealed increased activity of NF-kappaB-driven gene expression programs in multiple Sirt6-deficient tissues in vivo. Moreover, haploinsufficiency of RelA rescues the early lethality and degenerative syndrome of Sirt6-deficient mice. We propose that SIRT6 attenuates NF-kappaB signaling via H3K9 deacetylation at chromatin, and hyperactive NF-kappaB signaling may contribute to premature and normal aging.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , NF-kappa B/metabolismo , Sirtuínas/metabolismo , Fator de Transcrição RelA/metabolismo , Acetilação , Animais , Linhagem Celular , Cromatina/metabolismo , Cruzamentos Genéticos , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Humanos , Longevidade/genética , Camundongos , NF-kappa B/genética , Regiões Promotoras Genéticas , Sirtuínas/genética , Fator de Transcrição RelA/genética
7.
Cancer Res ; 68(2): 369-78, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18199530

RESUMO

A major goal of cancer research is to match specific therapies to molecular targets in cancer. Genome-scale expression profiling has identified new subtypes of cancer based on consistent patterns of variation in gene expression, leading to improved prognostic predictions. However, how these new genetic subtypes of cancers should be treated is unknown. Here, we show that a gene module map can guide the prospective identification of targeted therapies for genetic subtypes of cancer. By visualizing genome-scale gene expression in cancer as combinations of activated and deactivated functional modules, gene module maps can reveal specific functional pathways associated with each subtype that might be susceptible to targeted therapies. We show that in human breast cancers, activation of a poor-prognosis "wound signature" is strongly associated with induction of both a mitochondria gene module and a proteasome gene module. We found that 3-bromopyruvic acid, which inhibits glycolysis, selectively killed breast cells expressing the mitochondria and wound signatures. In addition, inhibition of proteasome activity by bortezomib, a drug approved for human use in multiple myeloma, abrogated wound signature expression and selectively killed breast cells expressing the wound signature. Thus, gene module maps may enable rapid translation of complex genomic signatures in human disease to targeted therapeutic strategies.


Assuntos
Mapeamento Cromossômico/métodos , Redes Reguladoras de Genes/fisiologia , Marcação de Genes , Terapia Genética , Neoplasias/genética , Neoplasias/terapia , Algoritmos , Antineoplásicos/uso terapêutico , Ácidos Borônicos/uso terapêutico , Bortezomib , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Processamento Eletrônico de Dados , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genes Mitocondriais , Humanos , Invasividade Neoplásica , Neoplasias/classificação , Neoplasias/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Prognóstico , Complexo de Endopeptidases do Proteassoma/genética , Pirazinas/uso terapêutico , Células Tumorais Cultivadas , Ferimentos e Lesões/genética
8.
Cancer Res ; 68(2): 506-15, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18199546

RESUMO

CSN5 has been implicated as a candidate oncogene in human breast cancers by genetic linkage with activation of the poor-prognosis, wound response gene expression signature. CSN5 is a subunit of the eight-protein COP9 signalosome, a signaling complex with multiple biochemical activities; the mechanism of CSN5 action in cancer development remains poorly understood. Here, we show that CSN5 isopeptidase activity is essential for breast epithelial transformation and progression. Amplification of CSN5 is required for transformation of primary human breast epithelial cells by defined oncogenes. The transforming effects of CSN5 require CSN subunits for assembly of the full COP9 signalosome and the isopeptidase activity of CSN5, which potentiates the transcriptional activity of MYC. Transgenic inhibition of CSN5 isopeptidase activity blocks breast cancer progression evoked by MYC and RAS in vivo. These results highlight CSN5 isopeptidase activity in breast cancer progression, suggesting it as a therapeutic target in aggressive human breast cancers.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carbono-Nitrogênio Liases/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Complexos Multiproteicos/metabolismo , Peptídeo Hidrolases/metabolismo , Peptídeo Hidrolases/fisiologia , Animais , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Complexo do Signalossomo COP9 , Carbono-Nitrogênio Liases/fisiologia , Transformação Celular Neoplásica/genética , Progressão da Doença , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Nus , Modelos Biológicos , Transplante de Neoplasias , Peptídeo Hidrolases/genética , Retroviridae/genética , Transdução Genética , Células Tumorais Cultivadas
9.
Nat Genet ; 38(4): 421-30, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16518402

RESUMO

Gene expression signatures encompassing dozens to hundreds of genes have been associated with many important parameters of cancer, but mechanisms of their control are largely unknown. Here we present a method based on genetic linkage that can prospectively identify functional regulators driving large-scale transcriptional signatures in cancer. Using this method we show that the wound response signature, a poor-prognosis expression pattern of 512 genes in breast cancer, is induced by coordinate amplifications of MYC and CSN5 (also known as JAB1 or COPS5). This information enabled experimental recapitulation, functional assessment and mechanistic elucidation of the wound signature in breast epithelial cells.


Assuntos
Neoplasias da Mama/genética , Transcrição Gênica/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Mapeamento Cromossômico , Imunofluorescência , Genes myc , Ligação Genética , Humanos , Repetições de Microssatélites
10.
Biochem J ; 397(1): 101-8, 2006 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-16563127

RESUMO

OM (oncostatin M) activates the human LDLR [LDL (low-density lipoprotein) receptor] gene transcription in HepG2 cells through the SIRE (sterol-independent regulatory element) of LDLR promoter. The SIRE sequence consists of a C/EBP (CCAAT/enhancer-binding protein)-binding site and a CRE (cAMP-response element). Our previous studies [Zhang, Ahlborn, Li, Kraemer and Liu (2002) J. Lipid Res. 43, 1477-1485; Zhang, Lin, Abidi, Thiel and Liu (2003) J. Biol. Chem. 278, 44246-44254] have demonstrated that OM transiently induces EGR-1 (early growth response gene product 1) expression and EGR-1 activates LDLR transcription primarily through a protein-protein interaction with C/EBPbeta, which serves as a co-activator of EGR-1. In the present study, we examined the direct role of C/EBPbeta as a transactivator in OM-regulated LDLR gene transcription independent of EGR-1. We show that OM induces C/EBPbeta expression with kinetics slower than EGR-1 induction. A significant increase in C/EBPbeta protein level is detected by 2 h of OM treatment and remains elevated for 24 h. Chromatin immunoprecipitation assays demonstrate that the amount of C/EBPbeta bound to the LDLR SIRE sequence is increased 2.8-fold of control by 2 h of OM treatment, reached the highest level of 8-fold by 4 h, and slowly declined thereafter. To further examine the requirement of C/EBPbeta in OM-stimulated LDLR expression, we developed a His-tagged dominant-negative mutant of C/EBPbeta (His-C/EBPbeta-P4; where P4 is plasmid 4 in our mutation series), consisting of the DNA-binding and leucine zipper domains of C/EBPbeta (amino acids 246-345). Expression of His-C/EBPbeta-P4 in HepG2 cells significantly diminishes the OM-induced increase in LDLR promoter activity and the elevation of endogenous LDLR mRNA expression. Taken together, these new findings identify C/EBPbeta as an OM-induced transactivator in LDLR gene transcription and provide a better understanding of the molecular mechanism underlying the sterol-independent regulation of LDLR expression.


Assuntos
Antineoplásicos/farmacologia , Proteína beta Intensificadora de Ligação a CCAAT/biossíntese , Proteína beta Intensificadora de Ligação a CCAAT/genética , Citocinas/farmacologia , Proteína 1 de Resposta de Crescimento Precoce/fisiologia , Receptores de LDL/biossíntese , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Carcinoma Hepatocelular/genética , Humanos , Cinética , Neoplasias Hepáticas/genética , Mutagênese Sítio-Dirigida , Oncostatina M , Regiões Promotoras Genéticas , Receptores de LDL/efeitos dos fármacos , Receptores de LDL/genética , Elementos Reguladores de Transcrição , Esteróis , Transcrição Gênica/efeitos dos fármacos , Ativação Transcricional , Células Tumorais Cultivadas
11.
Nat Med ; 10(12): 1344-51, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15531889

RESUMO

We identify berberine (BBR), a compound isolated from a Chinese herb, as a new cholesterol-lowering drug. Oral administration of BBR in 32 hypercholesterolemic patients for 3 months reduced serum cholesterol by 29%, triglycerides by 35% and LDL-cholesterol by 25%. Treatment of hyperlipidemic hamsters with BBR reduced serum cholesterol by 40% and LDL-cholesterol by 42%, with a 3.5-fold increase in hepatic LDLR mRNA and a 2.6-fold increase in hepatic LDLR protein. Using human hepatoma cells, we show that BBR upregulates LDLR expression independent of sterol regulatory element binding proteins, but dependent on ERK activation. BBR elevates LDLR expression through a post-transcriptional mechanism that stabilizes the mRNA. Using a heterologous system with luciferase as a reporter, we further identify the 5' proximal section of the LDLR mRNA 3' untranslated region responsible for the regulatory effect of BBR. These findings show BBR as a new hypolipidemic drug with a mechanism of action different from that of statin drugs.


Assuntos
Anticolesterolemiantes/uso terapêutico , Berberina/uso terapêutico , Regulação da Expressão Gênica/efeitos dos fármacos , Hipercolesterolemia/tratamento farmacológico , Receptores de LDL/metabolismo , Animais , Anticolesterolemiantes/farmacologia , Berberina/química , Berberina/farmacologia , Northern Blotting , China , Colesterol/sangue , LDL-Colesterol/sangue , Cricetinae , Primers do DNA , Citometria de Fluxo , Humanos , Fígado/metabolismo , Plasmídeos/genética , Receptores de LDL/genética , Sequências Reguladoras de Ácido Nucleico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Triglicerídeos/sangue , Células Tumorais Cultivadas
12.
Oncogene ; 23(58): 9336-47, 2004 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-15531928

RESUMO

Our previous studies conducted in MCF7-ptsp53 cells have demonstrated that overexpression of the wild-type (wt) p53 at permissive temperature 32 degrees C leads to growth arrest at the G2/M phase of the cell cycle. To identify novel p53-regulated genes that are responsible for the p53-induced G2/M arrest, we conducted cDNA microarray analyses. The array results indicated that the mRNA level of protein regulator of cytokinesis (PRC1) was significantly decreased when the p53 transactivation activity was turned on, suggesting that PRC1 transcription could be downregulated by p53. In this study, we have extensively examined the functional role of p53 in the regulation of PRC1, a cell cycle protein that plays important roles during cytokinesis. We demonstrate that increased expression of the wt p53 either by exogenous transfection or chemical induction results in reduced mRNA and protein expression of PRC1 in HCT116 p53(+/+), HCT116 p53(-/-), MCF-7, T47D, and HeLa cells. Importantly, we show that the decreased PRC1 expression is accompanied by the appearance of binucleated cells, indicating the process of cell division after mitosis being inhibited. By isolation and characterization of a 3 kb genomic fragment containing the 5'-flanking region and part of exon 1 of PRC1 gene, we demonstrate that p53 directly suppresses PRC1 gene transcription. We further locate the p53-responsive sequence to the proximal promoter region -214 to -163, relative to the transcriptional start site. The in vivo interaction of p53 with PRC1 gene promoter is further demonstrated by chromatin immunoprecipitation assay. Taken together, these new findings suggest that p53 may have important roles in the regulation of cytokinesis through controlling the transcription of PRC1.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Sequência de Bases , Proteínas de Ciclo Celular/genética , DNA , Regulação para Baixo , Doxorrubicina/farmacologia , Fluoruracila/farmacologia , Células HeLa , Humanos , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , Ligação Proteica , RNA Mensageiro/genética , Proteína Supressora de Tumor p53/fisiologia
13.
Breast Cancer Res Treat ; 87(2): 123-34, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15377837

RESUMO

Cytokine oncostatin M (OM) strongly inhibits the growth of MCF-7 breast cancer cells through the STAT3 signaling pathway. We have performed cDNA microarray analyses to identify novel OM-regulated genes in MCF-7 cells that are downstream effectors of the STAT3 signaling cascade. We show that expression of the calcium-binding protein S100A9 is strongly induced by OM in MCF-7 cells and this induction is markedly reduced in MCF-7-dnStat3 cells that express a dominant negative mutant of STAT3. We further show the induction of S100A9 by OM in other breast cancer cell lines whose proliferations are inhibited by OM whereas S100A9 is not significantly induced in SKBR-3 or HepG2 cells that do not respond to OM with a growth repression. In addition, inhibition of S100A9 expression with siRNA decreased cell response to OM-induced growth repression. By analyzing a series of S100A9 promoter reporter constructs, we have defined two discrete regions in the S100A9 promoter responsible for OM-induced transcriptional activation. Together these studies identify S100A9 as a novel OM-regulated gene through the STAT3-signaling cascade and suggest its involvement in the growth regulation of breast cancer cells.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/patologia , Calgranulina B/biossíntese , Proteínas de Ligação a DNA/farmacologia , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Peptídeos/farmacologia , Transativadores/farmacologia , Animais , Feminino , Análise de Sequência com Séries de Oligonucleotídeos , Oncostatina M , Fator de Transcrição STAT3 , Transdução de Sinais , Células Tumorais Cultivadas
14.
J Biol Chem ; 278(45): 44246-54, 2003 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-12947119

RESUMO

The sterol-independent regulatory element (SIRE) of the LDL receptor (LDLR) promoter mediates oncostatin M (OM)-induced transcription of the LDLR gene through a cholesterol-independent pathway. Our prior studies have detected specific associations of the zinc finger transcription factor Egr1 with the SIRE sequence in OM-stimulated HepG2 cells. Because the SIRE motif is composed of a c/EBP binding site and a cAMP response element, both of which are quite divergent from the classical GC-rich Egr1 recognition sequences, we hypothesized that Egr1 may regulate LDLR transcription through interacting with members of the c/EBP and CREB families. Here, we show that treating HepG2 cells with OM specifically leads to prominent increases of the levels of c/EBPbeta and Egr1 bound to the LDLR promoter in vivo. In vitro, the binding of Egr1 to the SIRE sequence is weak, but is strikingly enhanced in the presence of HepG2 nuclear extract. Mammalian two-hybrid assays demonstrate that the N-terminal transactivation domain of Egr1 specifically interacts with c/EBPbeta but not with c/EBPalpha or CREB. The OM treatment further enhances this interaction, resulting in a large increase in the Egr1 transactivating activity. The direct protein to protein contact between Egr1 and c/EBPbeta is also demonstrated by co-immunoprecipitation experiments. Furthermore, we show that a mutation of the phosphorylation motif of c/EBPbeta diminished the OM-stimulated interaction of Egr1 and c/EBPbeta. Taken together, we provide strong evidence that Egr1 regulates LDLR transcription via a novel mechanism of protein-protein interaction with c/EBPbeta.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Proteínas Imediatamente Precoces , Receptores de LDL/genética , Fatores de Transcrição/metabolismo , Sítios de Ligação , Proteína beta Intensificadora de Ligação a CCAAT/genética , Carcinoma Hepatocelular , Núcleo Celular/química , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Interações Medicamentosas , Proteína 1 de Resposta de Crescimento Precoce , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Técnicas de Imunoadsorção , Neoplasias Hepáticas , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Oncostatina M , Peptídeos/farmacologia , Fosforilação , Regiões Promotoras Genéticas/genética , Proteínas Recombinantes de Fusão , Elementos de Resposta/fisiologia , Fatores de Transcrição/genética , Transcrição Gênica/efeitos dos fármacos , Ativação Transcricional , Transfecção , Células Tumorais Cultivadas , Técnicas do Sistema de Duplo-Híbrido
15.
Arterioscler Thromb Vasc Biol ; 23(1): 90-6, 2003 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12524230

RESUMO

OBJECTIVE: A recent study identified a new class of compounds designated as the sterol-regulatory element binding protein (SREBP) cleavage-activating protein (SCAP) ligands that putatively bind to SCAP, leading to increased LDL receptor (LDLR) expression. In this study, we examined the effects of SCAP ligand GW707 in comparison with lovastatin and cytokine oncostatin M (OM) on the regulation of LDLR expression in cultured HepG2 cells. METHODS AND RESULTS: Our studies uncovered several new features that distinguish SCAP ligand from lovastatin, a classic 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor, and from OM, which utilize an SREBP-independent regulatory pathway. We show that the induction of LDLR mRNA expression by GW707 is not affected by intracellular cholesterol but is completely abolished by blocking de novo protein synthesis. Moreover, the effects of GW707 but not lovastatin on LDLR promoter activity, mRNA expression, and uptake of 1,1'-dioctadecyl-3,3,3',3'-tetramethyl-indocarbocyanin perchlorate-LDL are markedly enhanced by OM. We further demonstrate that the amounts of the mature form of SREBP-2 translocated to the nucleus under GW707 treatment are increased by costimulating cells with OM. CONCLUSIONS: Our studies provide the first evidence that higher levels of LDLR expression and function can be achieved through simultaneous stimulation of the SREBP-dependent and SREBP-independent pathways, suggesting a strategy to develop an adjunct therapeutic intervention utilizing both pathways.


Assuntos
Citocinas/farmacologia , Proteínas de Membrana/metabolismo , Peptídeos/farmacologia , Receptores de LDL/biossíntese , Esteroides/farmacologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Proteínas Estimuladoras de Ligação a CCAAT/fisiologia , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Citocinas/metabolismo , Proteínas de Ligação a DNA/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Cinética , Ligantes , Lovastatina/metabolismo , Lovastatina/farmacologia , Proteínas de Membrana/farmacologia , Oncostatina M , Biossíntese Peptídica/efeitos dos fármacos , Peptídeos/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de LDL/genética , Esteroides/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1 , Proteína de Ligação a Elemento Regulador de Esterol 2 , Fatores de Transcrição/fisiologia , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA