Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cancer Immunol Immunother ; 71(7): 1733-1746, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34837101

RESUMEN

Metformin has been found to have inhibitory effects on a variety of tumors. However, its effects on non-small cell lung cancer (NSCLC) remain unclear. We demonstrated that metformin could inhibit the proliferation of A549 and H1299 cells. RNA transcriptome sequencing revealed that PDL1 was significantly downregulated in both cell types following treatment with metformin (P < 0.001). Jaspar analysis and chromatin immunoprecipitation showed that CEBPB could directly bind the promoter region of PDL1. Western blotting showed that protein expression of the isoforms CEBPB-LAP*, CEBPB-LAP, and CEBPB-LIP was significantly upregulated and the LIP/LAP ratio was increased. Gene chip analysis showed that PDL1 was significantly upregulated in A549-CEBPB-LAP cells and significantly downregulated in A549-CEBPB-LIP cells (P < 0.05) compared with CEBPB-NC cells. Dual-luciferase reporter gene assay showed that CEBPB-LAP overexpression could promote transcription of PDL1 and CEBPB-LIP overexpression could inhibit the process. Functional assays showed that the changes in CEBPB isoforms affected the function of NSCLC cells. Western blotting showed that metformin could regulate the function of NSCLC cells via AMPK-CEBPB-PDL1 signaling. Animal experiments showed that tumor growth was significantly inhibited by metformin, and atezolizumab and metformin had a synergistic effect on tumor growth. A total of 1247 patients were retrospectively analyzed, including 166 and 1081 patients in metformin and control groups, respectively. The positive rate of PDL1 was lower than that of the control group (HR = 0.338, 95% CI = 0.235-0.487; P < 0.001). In conclusion, metformin inhibited the proliferation of NSCLC cells and played an anti-tumor role in an AMPK-CEBPB-PDL1 signaling-dependent manner.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Metformina , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Quinasas Activadas por AMP/farmacología , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Metformina/farmacología , Metformina/uso terapéutico , Estudios Retrospectivos , Transducción de Señal
2.
J Hazard Mater ; 465: 132997, 2024 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-38008054

RESUMEN

Presently, the exposure of plasticizers to humans and animals occurs daily, which pose a potential threat to reproductive health. In the present study, a pregnant mouse model exposed to di(2-ethylhexyl) phthalate (DEHP, one of the most common plasticizers) and melatonin was established, and the single-cell transcriptome technology was applied to investigate the effects of melatonin in ovarian cells against DEHP. Results showed that DEHP markedly altered the gene expression pattern of ovarian cells, and severely weakened the histone methylation modification of oocytes. The administration of melatonin recovered the expression of LHX8 and SOHLH1 proteins that essential for primordial follicle formation, and increased the expression of CEBPB, as well as key genes of histone methylation modification (such as Smyd3 and Kdm5a). In addition, the ovarian damage caused by DEHP was also relieved after the overexpression of CEBPB, which suggested melatonin could improve primordial follicle formation progress via enhancing CEBPB expression in mice. Besides, the apoptosis of ovarian cells induced by DEHP also was diminished by melatonin. The study provides evidence of melatonin preventing the damage mediated by plasticizers on the reproductive system in females and CEBPB may serve as a downstream target factor of melatonin in the process.


Asunto(s)
Dietilhexil Ftalato , Melatonina , Ácidos Ftálicos , Embarazo , Femenino , Humanos , Animales , Ratones , Melatonina/farmacología , Plastificantes/toxicidad , Dietilhexil Ftalato/toxicidad , Histonas , Oocitos , Proteína beta Potenciadora de Unión a CCAAT/farmacología
3.
Int J Cardiol ; 376: 115-124, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36716974

RESUMEN

OBJECTIVE: The study aims to investigate the effect of developmental endothelial locus-1(DEL-1) expression in atherosclerotic plaque formation and its mechanism. METHODS: Human left coronary arteries were collected to detect the DEL-1 expression. The ApoE-/- mice were used to establish the atherosclerosis mice model. The left coronary artery and mouse aorta were stained with HE, Oil Red O, and Movat staining. The DEL-1 levels, chemokines CXC chemokine receptor 4 (CXCR4) and its ligand stromal cell-derived factor-1alpha (SDF-1α), pathway protein glycogen synthase kinase-3ß (GSK-3ß), CCAAT enhanced binding protein ß (C/EBPß), and downstream inflammatory factors (C-X-C motif chemokine 2 (MIP-2or CXCL2), macrophage inflammatory protein-1alpha (MIP-1α or CCL3),Tumor Necrosis Factor alpha (TNF-α) were detected by immunoblotting and immunohistochemistry. Pearson correlation coefficient was used to analyze the correlation between DEL-1 gene expression and inflammatory factors in the lesion group and the correlation between DEL-1 gene expression and structure-related indexes. RESULTS: Compared with Control group(CON), the intravascular plaque area was widened, accompanied by narrowed lumens. The number of plaque foam cells was significantly increased in the high fat and high cholesterol (AS group) or AAV9-eGFP group (P < 0.05). Compared to CON, the enhanced fluorescence intensity of DEL-1 with CD68 in the AS or AAV9-eGFP groups. Diminished fluorescence of DEL-1 with CD68 expression in AAV9-CXCR4 group compared to AS group or AAV9-eGFP group. The DEL-1 and its downstream proteins in AS group or AAV9-eGFP group were mainly accumulated in the macrophage cytoplasm. The DEL-1 expression level was significantly and positively correlated with plaque area, lumen stenosis, plaque foam cell count, TNFα, CXCL2, and CCL3 levels. CONCLUSION: DEL-1 inhibition decreases macrophagic inflammatory factors involved in atherosclerotic plaque formation.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Ratones , Humanos , Animales , Placa Aterosclerótica/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/farmacología , Ratones Noqueados para ApoE , Aterosclerosis/metabolismo , Transducción de Señal , Macrófagos/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
4.
Metab Syndr Relat Disord ; 20(10): 576-583, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36318489

RESUMEN

Background: Metabolic syndrome (MetS) is a group of signs and symptoms that are associated with a higher risk of type 2 diabetes mellitus and cardiovascular diseases. The major risk factor for developing MetS is abdominal obesity, which is caused by an increase in adipocyte size or quantity. Increased adipocyte quantity is a result of differentiation of stem cells into adipose tissue. Numerous studies have investigated the expression of key transcription factors, including PPARG and CEBPB during adipocyte differentiation in murine cells such as 3T3-L1 cell lines. To better understand the expression changes during the process of fat accumulation in adipose-derived stem cells (ASCs), we compared the expression of DYRK1B, PPARG, and ẟB in ASCs between the patient (harboring DYRK1B R102C) and control (healthy individuals) groups. Methods: Gene expression was evaluated on the eighth day before induction and days 1, 5, and 15 postinduction. The pluripotent capacity of ASCs and the potential for differentiation into adipocytes were confirmed by flow cytometry analysis of surface markers (CD34, CD44, CD105, and CD90), and Oil Red O staining, respectively. The Expression of DYRK1B, PPARG, and CEBPB were assessed by real-time-polymerase chain reaction in patients and normal individuals. The effects of AZ191, a potent small molecule inhibitor on DYRK1B and CEBPB expression in patients' samples were studied. Result: The expression of DYRK1B kinase and transcription factors (CEBPB and PPARG) are higher in ASCs harboring DYRK1B R102C compared with noncarriers on days 5 and 15 during adipocyte differentiation. These proteins may be helpful to elucidate the mechanisms underlying obesity and obesity-related disorders like MetS. Furthermore, the new compound AZ191 exhibited inhibitory activity toward DYRK1B and CEBPB. We suggest that AZ191 may be helpful in defining the potential roles of DYRK1B and CEBPB in adipogenesis.


Asunto(s)
Adipogénesis , Diabetes Mellitus Tipo 2 , Animales , Humanos , Ratones , Adipocitos/metabolismo , Adipogénesis/genética , Tejido Adiposo/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/farmacología , Diabetes Mellitus Tipo 2/metabolismo , Obesidad/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , PPAR gamma/farmacología , Células Madre/metabolismo , Factores de Transcripción , Quinasas DyrK
5.
Arterioscler Thromb Vasc Biol ; 20(12): 2559-65, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11116053

RESUMEN

Type II secreted phospholipase A(2) (sPLA(2)) releases precursors of important inflammatory lipid mediators from phospholipids. Some observations have indicated that the sPLA(2), which has been implicated in chronic inflammatory conditions such as arthritis, contributes to atherosclerosis in the arterial wall. sPLA(2) was not detected in control vascular smooth muscle cells (VSMC). Treatment of VSMC with agents that increase intracellular cAMP (eg, forskolin, dibutyryl [db]-cAMP) resulted in a time- and concentration-dependent increase in sPLA(2) gene expression. Semiquantitative reverse transcriptase polymerase chain reaction (RT-PCR) showed a marked dose-dependent inhibition of forskolin-induced mRNA by protein kinase A inhibitor. Electrophoretic mobility shift analysis of nuclear proteins from forskolin-treated and db-cAMP-treated VSMC with C/EBP consensus oligonucleotides and C/EBP oligonucleotides from the rat promoter revealed greater binding than in control VSMC. Incubation of VSMC with H89, a specific protein kinase inhibitor, also blocked the binding of nuclear C/EBP to the C/EBP site of the rat promoter induced by db-cAMP and forskolin. Binding was unchanged with the use of CRE consensus oligonucleotides. Antibodies revealed the specific formation of C/EBP/DNA complexes, the majority of which were supershifted by C/EBP-ss and -delta antibodies. Functional activation of C/EBP was confirmed by a luciferase reporter gene assay. A construct comprising 4 tandem repeat copies of the C/EBP element from the rat sPLA(2) promoter linked to luciferase was transcriptionally activated in VSMC by cotransfection with expression vector for the protein kinase A catalytic subunit. It was also significantly activated in transfected VSMC treated by forskolin or db-cAMP. H89 inhibited this activations. We therefore conclude that the increases in sPLA(2) mRNA and enzyme activity produced by cAMP-elevating agents is controlled by a mechanism involving nuclear C/EBP-ss and -delta acting through a protein kinase A signaling pathway.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/farmacología , Músculo Liso Vascular/efectos de los fármacos , Fosfolipasas A/genética , Sulfonamidas , Factores de Transcripción , Animales , Bucladesina/farmacología , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/farmacología , Proteína delta de Unión al Potenciador CCAAT , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/farmacología , Células Cultivadas , Colforsina/farmacología , Proteína Quinasa Tipo II Dependiente de AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Inducción Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Isoquinolinas/farmacología , Luciferasas/genética , Masculino , Músculo Liso Vascular/enzimología , Oligonucleótidos/farmacología , Fosfolipasas A/biosíntesis , ARN Mensajero/análisis , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Transcripción Genética/efectos de los fármacos , Transfección
6.
Mol Endocrinol ; 15(2): 228-40, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11158330

RESUMEN

Beta-casein gene transcription is controlled primarily by a composite response element (CoRE) that integrates signaling from the lactogenic hormones, PRL, insulin, and hydrocortisone, in mammary epithelial cells. This CoRE contains binding sites for STAT5 (signal transducer and activator of transcription 5) and C/EBPbeta (CCAAT/enhancer-binding protein-beta) and several half-sites for glucocorticoid receptor (GR). To examine how interactions among these three transcription factors might regulate beta-casein gene transcription, a COS cell reconstitution system was employed. Cooperative transactivation was observed when all three factors were expressed, but unexpectedly was not seen between STAT5 and C/EBPbeta in the absence of full-length, transcriptionally active GR. Cooperativity required the amino-terminal transactivation domain of C/EBPbeta, and neither C/EBPalpha nor C/EBPdelta was able to substitute for C/EBPbeta when cotransfected with STAT5 and GR. Different GR determinants were needed for transcriptional cooperation between STAT5 and GR as compared with those required for all three transcription factors. These studies provide some new insights into the mechanisms responsible for high level, tissue-specific expression conferred by the beta-casein CoRE.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/farmacología , Caseínas/genética , Proteínas de Unión al ADN/farmacología , Proteínas de la Leche , Receptores de Glucocorticoides/fisiología , Transactivadores/farmacología , Transcripción Genética/efectos de los fármacos , Animales , Sitios de Unión , Proteína beta Potenciadora de Unión a CCAAT/química , Proteína beta Potenciadora de Unión a CCAAT/genética , Células COS , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Vectores Genéticos , Mutagénesis , Plásmidos , Receptores de Glucocorticoides/química , Receptores de Glucocorticoides/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/metabolismo , Factor de Transcripción STAT5 , Relación Estructura-Actividad , Transactivadores/genética , Activación Transcripcional , Transfección , Proteínas Supresoras de Tumor
7.
Acta Biomater ; 11: 191-203, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25257317

RESUMEN

Adipose-derived stem cells (ADSCs) are promising for cartilage repair due to their easy accessibility and chondrogenic potential. Although chondrogenesis of transforming growth factor-ß (TGF-ß) mediated mesenchymal stem cells (MSCs) is well established in vitro, clinical tissue engineering requires effective and controlled delivery of TGF-ß in vivo. In this work, a self-assembled peptide scaffold was employed to construct cartilages in vivo through the chondrogenesis from ADSCs controlled by recombinant fusion protein LAP-MMP-mTGF-ß3 that was transfected by lentiviral vectors. During this course, the addition of matrix metalloproteinases (MMPs) can trigger the release of mTGF-ß3 from the recombinant fusion protein of LAP-MMP-mTGF-ß3 in the combined scaffolds, thus stimulating the differentiation of ADSCs into chondrogenesis. The specific expression of cartilage genes was analyzed by real-time polymerase chain reaction and Western blot. The expression of chondrocytic markers was obviously upregulated to a higher level compared to the one by commonly used TGF-ß3 alone. After 3 weeks of in vitro culturing, the hybrids with differentiated chondrogenesis were then injected subcutaneously into nude mice and retrieved after 4 weeks of culturing in vivo. Histological analysis also confirmed that the recombinant fusion protein was more effective for the formation of cartilage matrix than the cases either with TGF-ß3 alone or without LAP-MMP-mTGF-ß3 (P<0.05). This study demonstrates that controlled local delivery of the LAP-MMP-mTGF-ß3 constructs can accelerate differentiation of ADSCs into the cartilage in vivo, which indicates the great potential of this hybrid in rapid therapy of osteoarthritis.


Asunto(s)
Tejido Adiposo/metabolismo , Diferenciación Celular/efectos de los fármacos , Condrocitos/metabolismo , Condrogénesis/efectos de los fármacos , Células Madre/metabolismo , Factor de Crecimiento Transformador beta3/farmacología , Tejido Adiposo/citología , Animales , Antígenos de Diferenciación/biosíntesis , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/farmacología , Células Cultivadas , Condrocitos/citología , Gelatinasas/genética , Gelatinasas/farmacología , Ratones , Osteoartritis/terapia , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Células Madre/citología , Factor de Crecimiento Transformador beta3/genética
8.
Virus Res ; 89(1): 53-63, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12367750

RESUMEN

To examine the mechanism of HIV-1 regulation by NF-IL6 in activated human cells, we selected a Jurkat cell line that did not contain endogenous NF-IL6. In this cellular environment, we evaluated the effect of exogenous NF-IL6 on transcription mediated by native and deleted LTR sequences. In Jurkat cells stimulated with LPS and PMA, LTR-mediated transcription was enhanced by NF-IL6. The results of deletion studies revealed a central role for the basal LTR region and the TATA element in the LTR, in upregulation of reporter gene expression by NF-IL6 in activated cells. In the selected cellular environment, regulation of transcription by NF-IL6 was not evident in studies of promoter regions of other genes. The results implied that the basal region of HIV-1 LTR includes molecular properties that support activation of HIV-1 by NF-IL6 in stimulated cells.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/farmacología , Regulación Viral de la Expresión Génica , VIH-1/metabolismo , Células Jurkat/virología , Transcripción Genética/efectos de los fármacos , Duplicado del Terminal Largo de VIH/genética , VIH-1/genética , Humanos , Activación de Linfocitos , Regiones Promotoras Genéticas
9.
Mol Cell Endocrinol ; 172(1-2): 57-67, 2001 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-11165040

RESUMEN

The chum salmon insulin-like growth factor II (IGF-II) gene is highly expressed in liver tissue. In this study we demonstrate that two transcription factors, Sp1 and C/EBPbeta, are involved in the enhanced expression of the salmon IGF-II gene. The presence of the fish homolog for C/EBPbeta in salmon liver RNA was confirmed by Northern blotting. The sIGF-II promoter was activated up to 20-fold by co-transfection with C/EBPbeta. The functional importance of four out of the five putative C/EBPbeta binding sites was demonstrated with mutational analysis in transient transfection assays. The transcription factor Sp1 binds to two sites within the salmon IGF-II promoter. Interestingly, mutation of the Sp1 binding sites decreases not only the basal IGF-II promoter activity but also the C/EBPbeta-induced transactivation. These results demonstrate that liver-enriched C/EBPbeta and ubiquitously expressed Sp1 each activate the sIGF-II promoter and that Sp1 is required for full transactivation of the sIGF-II gene by C/EBPbeta. This suggests that C/EBPbeta and Sp1 act in synergy.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/farmacología , Factor I del Crecimiento Similar a la Insulina/genética , Oncorhynchus keta/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Factor de Transcripción Sp1/farmacología , Animales , Secuencia de Bases , Sitios de Unión/genética , Northern Blotting , Sinergismo Farmacológico , Hígado/metabolismo , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , ARN Mensajero/análisis , Activación Transcripcional/efectos de los fármacos
10.
Cancer Res ; 67(23): 11272-83, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18056453

RESUMEN

CCAAT/enhancer binding protein beta (C/EBP beta) is a transcription factor implicated in the control of development, differentiation, and proliferation of mammary epithelial cells. However, it remains unclear how C/EBP beta is involved in tumor suppression through its interaction with specific downstream genes in breast cancer. Tumor cells overexpress serine proteases, which play crucial roles in tumor invasion and metastasis. Elafin is an endogenous serine protease inhibitor and is transcriptionally down-regulated in most tumor cell lines. In this study, we show that C/EBP beta is differentially expressed in normal versus tumor cell lines and normal adjacent versus tumor tissues obtained from breast cancer patients. We identified elafin as a downstream effector of C/EBP beta and show that elafin is also differentially regulated between normal and tumor cells. The mechanism by which C/EBP beta regulates elafin expression is through its direct interaction with the elafin promoter. There are three C/EBP beta binding sites involved in the elafin promoter activity, and the overexpression of C/EBP beta transactivates the elafin gene through these sites in tumor cells. RNA interference studies in normal cells further evidenced the requirement of the C/EBP beta for the elafin expression and negative feedback loop between C/EBP beta and elafin. We suggest that elafin is a novel substrate of C/EBP beta, and alterations in C/EBP beta isoforms result in their differential binding to the elafin promoter, leading to the altered expression of the elafin between normal and tumor cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Mama/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/farmacología , Elafina/genética , Transcripción Genética , Adulto , Anciano , Anciano de 80 o más Años , Sitios de Unión , Northern Blotting , Western Blotting , Mama/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Núcleo Celular/metabolismo , Células Cultivadas , Inmunoprecipitación de Cromatina , Elafina/metabolismo , Elementos de Facilitación Genéticos , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Luciferasas/metabolismo , Persona de Mediana Edad , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas , Procesamiento Proteico-Postraduccional , ARN Mensajero , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Células Tumorales Cultivadas
11.
Virology ; 299(2): 256-265, 2002 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12202228

RESUMEN

DNA-binding and functional assays examined the role played by NF-IL6 in regulation of HIV-1 transcription in human monocyte/macrophages (U937 cells), stimulated with LPS+PMA. When incubated with nuclear extracts from stimulated cells, a region (-189/-147), containing the major NF-IL6-binding sequence and the USF site, interacted selectively with USF1 and USF2. Anti-C/EBPbeta reacted poorly with the complexes produced with the wild-type probe. In contrast, complex formation with NF-IL6 was clearly evident in experiments analyzing a probe containing an insertion in the USF site. In functional assays, increasing concentrations of a decoy against NF-IL6 reduced gene expression from the LTR of the wild-type HIV-1 variant, supporting a critical role for NF-IL6 in regulation of HIV-1 transcription in stimulated monocyte/macrophages. The decoy also reduced gene expression from a deletion construct lacking NF-IL6-binding sequences. The results implied that in LPS+PMA-stimulated monocyte/macrophages, the endogenous NF-IL6 could act via a site-independent pathway in upregulation of HIV-1 transcription. Analysis of a short DNA segment, containing the -189/-147 region, suggested functional interactions of NF-IL6 and USF. In activated cells exogenous NF-IL6 enhanced dramatically gene expression through a short DNA segment containing the NF-kappaB sites, supporting functional interactions of NF-IL6 and NF-kappaB.


Asunto(s)
VIH-1/genética , Macrófagos/virología , Monocitos/virología , Transcripción Genética , Proteína beta Potenciadora de Unión a CCAAT/farmacología , Duplicado del Terminal Largo de VIH , Humanos , Lipopolisacáridos/farmacología , Activación de Macrófagos , Acetato de Tetradecanoilforbol/farmacología , Células U937
12.
Biol Pharm Bull ; 23(12): 1424-9, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11145170

RESUMEN

CCAAT/enhancer-binding protein delta (C/EBPdelta) transcription factor is rapidly induced at an early stage of acute phase response. We previously reported that this induction was mainly mediated by acute phase response factor/signal transducers and activators of transcription 3 (APRF/STAT3). Furthermore, the high expression level of C/EBPdelta is maintained by autoregulation mechanisms through the C/EBPdelta binding sites located downstream of C/EBPdelta gene. Thereafter, the expression of C/EBPdelta gene decreases rapidly to the basal level. However, these mechanisms are still unknown. According to both transfection and DNA binding analyses, liver-enriched inhibitory protein (LIP), the shorter form of C/EBPbeta and C/EBP-homologous protein 10 (CHOP10), were found to inhibit C/EBPdelta gene expression. DNA binding analysis has further indicated that both LIP and CHOP10 form heterodimers with C/EBPdelta, and inhibit the binding of C/EBPdelta homodimer to the C/EBPdelta binding sites located downstream of C/EBPdelta gene. Taken together, these findings indicated that the maintained expression of C/EBPdelta gene by autoregulation was inhibited and decreased to the basal level as a result of the competition of other C/EBP family proteins. Thus, C/EBPdelta gene expression is mediated by the gene regulation circuit through the downstream C/EBPdelta binding sites.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/fisiología , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/fisiología , Regulación de la Expresión Génica/fisiología , Factores de Transcripción/fisiología , Animales , Sitios de Unión , Unión Competitiva , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/farmacología , Proteína delta de Unión al Potenciador CCAAT , Proteínas Potenciadoras de Unión a CCAAT/antagonistas & inhibidores , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/farmacología , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Dimerización , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratas , Factor de Transcripción STAT3 , Transactivadores/metabolismo , Transactivadores/fisiología , Factor de Transcripción CHOP , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/farmacología , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/fisiología , Transfección , Células Tumorales Cultivadas
13.
Blood ; 99(6): 2032-6, 2002 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11877276

RESUMEN

CCAAT/enhancer-binding proteins (C/EBPs) are critical transcriptional regulators of differentiation of hematopoietic cells. Previous studies have shown that targeted disruption of the C/ebpalpha gene results in a lack of granulocytic differentiation with an arrest at the stage of immature myeloblasts. By using a gene replacement strategy in which C/EBPbeta was expressed from the C/ebpalpha gene locus of C/EBPalpha-null mice, we have evaluated the ability of C/EBPbeta to function for C/EBPalpha in directing differentiation along the granulocytic pathway. We show that the morphology and the differential cell counts of the bone marrow and peripheral blood cells from C/EBPbeta knockin mice are indistinguishable from those of their wild-type littermates, indicating that hematopoiesis occurs normally in these animals. Additionally, we analyzed expression of 21 myeloid-specific genes, including markers for distinct stages of granulocytic differentiation, and found no significant differences in their levels of expression in the bone marrow of C/EBPbeta knockin and wild-type mice. These results imply that C/EBPbeta can substitute for C/EBPalpha during hematopoiesis when expressed from the C/ebpalpha gene locus.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/biosíntesis , Hematopoyesis/genética , Animales , Células Sanguíneas/citología , Células Sanguíneas/efectos de los fármacos , Células Sanguíneas/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/farmacología , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Granulocitos/citología , Hematopoyesis/efectos de los fármacos , Ratones , Ratones Noqueados , ARN Mensajero/análisis
14.
J Nutr ; 132(11): 3314-24, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12421845

RESUMEN

Indole-3-carbinol (I3C) and its dimer 3,3'-diindolylmethane (DIM), obtained from dietary consumption of cruciferous vegetables, have multiple biochemical activities. Both compounds have been effective clinically in treating precancerous lesions of the cervix and laryngeal papillomas, pathologies with a human papillomavirus (HPV) component. Using cDNA microarrays, we examined early changes in gene expression after treatment with 100 micro mol/L DIM in C33A and CaSki cervical cancer cells and in an immortalized human epithelial cell line (HaCat), as well as in normal human foreskin keratinocytes (HFK). Multiple analyses were done after treating C33A cells for 6 h; other analyses included 4- and 12-h treatments of C33A and 6-h treatments of CaSki, HaCat and HFK cells. DIM consistently altered the expression of >100 genes at least twofold. Many of the stimulated genes encode transcription factors and proteins involved in signaling, stress response and growth. Results were comparable between transformed cells with and without integrated HPV sequences, and many of the same genes were induced in these cancer-derived cells and in noncancer cells. Eight genes encoding bZip proteins were among the most consistently and robustly induced, including the stress-associated immediate early gene GADD153 (>50 fold in C33A) and nuclear factor-interleukin 6 (NF-IL6), also known as c/EBPbeta, (>5 fold in C33A), which has been shown to reduce expression of HPV oncogenes. Induction of GADD153, NF-IL6 and ATF3 was confirmed by Western analysis. In functional analyses, DIM not only suppressed transcription of a luciferase gene driven by the HPV11 upstream regulatory region (URR) in C33A, CaSki, HaCat and HFK cells from >2-fold to 37-fold depending on the type of cells, but also reduced endogenous transcription of HPV16 oncogenes to undetectable levels in CaSki cells as determined by an RNase protection assay. Ectopic expression of GADD153 or NF-IL6 suppressed transcription in a dose-dependent manner driven by the HPV11 URR in C33A, CaSki, HaCat and HFK cells. These results identify unexpected ways in which dietary I3C and DIM invoke cellular responses and are consistent with a potential antiviral effect of DIM on keratinocytes, but they do not explain the differential sensitivity of transformed keratinocytes to apoptosis by DIM.


Asunto(s)
Expresión Génica/efectos de los fármacos , Indoles/farmacología , Queratinocitos/metabolismo , Western Blotting , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/farmacología , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/farmacología , Células Cultivadas , Femenino , Humanos , Luciferasas/genética , Papillomaviridae/efectos de los fármacos , Papillomaviridae/genética , Regiones Promotoras Genéticas , Secuencias Reguladoras de Ácidos Nucleicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción CHOP , Factores de Transcripción/genética , Factores de Transcripción/farmacología , Transcripción Genética/efectos de los fármacos , Transfección , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/virología
15.
Genes Dev ; 16(8): 908-12, 2002 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-11959839

RESUMEN

Fibroblast growth factors (FGFs) are important intercellular signaling molecules in developmental processes. Here, we show that FGF10 is secreted by cultured preadipocytes and that prevention of FGF10 signaling inhibits the expression of C/EBPbeta and the subsequent differentiation of these cells. An active form of C/EBPbeta rescued differentiation of the cells in which FGF10 signaling was blocked. Development of white adipose tissue and the expression of C/EBPbeta in this tissue of FGF10 knockout mice were markedly reduced, and the ability of embryonic fibroblasts derived from FGF10 knockout mice to differentiate into adipocytes was impaired. Therefore, FGF10 plays an important role in adipogenesis, at least partly by contributing to the expression of C/EBPbeta through an autocrine/paracrine mechanism.


Asunto(s)
Tejido Adiposo/embriología , Tejido Adiposo/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Tejido Adiposo/citología , Animales , Comunicación Autocrina , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Factor 10 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/deficiencia , Factores de Crecimiento de Fibroblastos/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Glucocorticoides/farmacología , Insulina/farmacología , Ratones , Ratones Noqueados , Comunicación Paracrina , Inhibidores de Fosfodiesterasa/farmacología , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
16.
Biochem Biophys Res Commun ; 285(3): 811-9, 2001 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-11453665

RESUMEN

CCAAT/enhancer binding protein (C/EBP) family members are known to transactivate the gene encoding cytosolic phosphoenolpyruvate carboxykinase (PEPCK; EC 4.1.1.32) in hepatocytes via promoter proximal C/EBP response elements. PEPCK is also expressed in adipocytes; however, fibroblasts that are homozygous null for C/EBPbeta cannot express PEPCK when induced to differentiate into adipocytes (Tanaka et al., EMBO J. 16, 7432-7443, 1997). This along with our previous observation that an upstream adipocyte-specific enhancer contains multiple putative C/EBP binding elements suggested the possibility that C/EBPbeta transactivates the PEPCK gene in adipocytes via distal elements. We report here that C/EBPbeta transactivates a PEPCK-luciferase chimera in transient transfection assays. C/EBPbeta acted independently of peroxisome proliferator-activated receptor gamma (PPARgamma) which is required for function of the enhancer. C/EBPbeta in nuclear extracts and recombinant C/EBPbeta bound three of the putative C/EBP-binding elements within the enhancer. C/EBPbeta binding to these three elements was strongly cooperative. However, mutation of all three elements did not affect reporter transactivation by C/EBPbeta suggesting that additional elements participate in PEPCK regulation or that the effects of C/EBPbeta are indirect.


Asunto(s)
Adipocitos/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Elementos de Facilitación Genéticos/genética , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Adipocitos/citología , Adipocitos/efectos de los fármacos , Animales , Sitios de Unión/efectos de los fármacos , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/farmacología , Línea Celular , Núcleo Celular/metabolismo , Elementos de Facilitación Genéticos/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Humanos , Ratones , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Homología de Secuencia de Ácido Nucleico , Factores de Transcripción/metabolismo , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA