Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Oncogene ; 36(39): 5484-5496, 2017 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-28534513

RESUMEN

Protein disulfide isomerase a4 (PDIA4) is implicated in the growth and death of tumor cells; however, its molecular mechanism and therapeutic potential in cancer are unclear. Here, we found that PDIA4 expression was upregulated in a variety of tumor cell lines and human lung adenocarcinoma tissues. Knockdown and overexpression of PDIA4 in tumor cells showed that PDIA4 facilitated cell growth via the reduction of caspases 3 and 7 activity. Consistently, Lewis lung carcinoma cells overexpressing PDIA4 grew faster than did parental cells in tumor-bearing mice, as shown by a reduced survival rate, increased tumor size and metastasis, and decreased cell death and caspases 3 and 7 activity. PDIA4 knockdown resulted in opposite outcomes. Moreover, results obtained in mice with spontaneous hepatoma indicated that PDIA4 deficiency significantly reduced hepatic tumorigenesis and cyst formation and increased mouse survival, tumor death, and caspases 3 and 7 activity. Mechanistic studies illustrated that PDIA4 negatively regulated tumor cell death by inhibiting degradation and activation of procaspases 3 and 7 via their mutual interaction in a CGHC-dependent manner. Finally, we found that 1,2-dihydroxytrideca-5,7,9,11-tetrayne, a PDIA4 inhibitor, reduced tumor development via enhancement of caspase-mediated cell death in TSA tumor-bearing mice. These findings characterize PDIA4 as a negative regulator of cancer cell apoptosis and suggest that PDIA4 is a potential therapeutic target for cancer.


Asunto(s)
Caspasas/metabolismo , Precursores Enzimáticos/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Animales , Línea Celular Tumoral , Femenino , Células HEK293 , Células Hep G2 , Humanos , Células Jurkat , Células MCF-7 , Masculino , Melanoma Experimental , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
2.
Oncogene ; 36(31): 4405-4414, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28368406

RESUMEN

The epithelial-mesenchymal transition (EMT) is an important process in the progression of cancer. However, its occurrence and mechanism of regulation are not fully understood. We propose a regulatory pathway in which spermatogenic leucine zipper 1 (SPZ1) promotes EMT through its transactivating ability in increasing TWIST1 expression. We compared the expression of SPZ1 and TWIST1 in specimens of hepatocarcinoma cells (HCCs) and non-HCCs. Expression of SPZ1 exhibited a tumor-specific expression pattern and a high correlation with patients' survival time, tumor size, tumor number and progression stage. Moreover, forced expression and knockdown of SPZ1 in hepatoma cells showed that SPZ1 was able to regulate the cellular proliferation, invasion, and tumorigenic activity in a TWIST1-dependent manner in vitro and in vivo. These data demonstrate that SPZ1, a newly dscribed molecule, transactivates TWIST1 promoters, and that this SPZ1-TWIST axis mediates EMT signaling and exerts significant regulatory effects on tumor oncogenesis.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/fisiología , Carcinoma Hepatocelular/patología , Transición Epitelial-Mesenquimal , Neoplasias Hepáticas/patología , Proteínas Nucleares/fisiología , Proteína 1 Relacionada con Twist/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Carcinogénesis , Carcinoma Hepatocelular/etiología , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Neoplasias Hepáticas/etiología , Masculino , Persona de Mediana Edad , Proteínas Nucleares/genética , Proteína 1 Relacionada con Twist/genética
3.
Cell Death Dis ; 4: e921, 2013 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-24232097

RESUMEN

Oxidative stress and reactive oxygen species (ROS) are associated with diseases such as cancer, cardiovascular complications, inflammation and neurodegeneration. Cellular defense systems must work constantly to control ROS levels and to prevent their accumulation. We report here that the Jun dimerization protein 2 (JDP2) has a critical role as a cofactor for transcription factors nuclear factor-erythroid 2-related factor 2 (Nrf2) and small Maf protein family K (MafK) in the regulation of the antioxidant-responsive element (ARE) and production of ROS. Chromatin immunoprecipitation-quantitative PCR (qPCR), electrophoresis mobility shift and ARE-driven reporter assays were carried out to examine the role of JDP2 in ROS production. JDP2 bound directly to the ARE core sequence, associated with Nrf2 and MafK (Nrf2-MafK) via basic leucine zipper domains, and increased DNA-binding activity of the Nrf2-MafK complex to the ARE and the transcription of ARE-dependent genes. In mouse embryonic fibroblasts from Jdp2-knockout (Jdp2 KO) mice, the coordinate transcriptional activation of several ARE-containing genes and the ability of Nrf2 to activate expression of target genes were impaired. Moreover, intracellular accumulation of ROS and increased thickness of the epidermis were detected in Jdp2 KO mice in response to oxidative stress-inducing reagents. These data suggest that JDP2 is required to protect against intracellular oxidation, ROS activation and DNA oxidation. qPCR demonstrated that several Nrf2 target genes such as heme oxygenase-1, glutamate-cysteine ligase catalytic and modifier subunits, the notch receptor ligand jagged 1 and NAD(P)H dehydrogenase quinone 1 are also dependent on JDP2 for full expression. Taken together, these results suggest that JDP2 is an integral component of the Nrf2-MafK complex and that it modulates antioxidant and detoxification programs by acting via the ARE.


Asunto(s)
Factor de Transcripción MafK/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Represoras/metabolismo , Western Blotting , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Técnica del Anticuerpo Fluorescente , Glutatión/metabolismo , Células Hep G2 , Humanos , Factor de Transcripción MafK/genética , Factor 2 Relacionado con NF-E2/genética , Unión Proteica , ARN Interferente Pequeño , Proteínas Represoras/genética
4.
Cell Death Dis ; 4: e907, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24201806

RESUMEN

The androgen receptor (AR) has a critical role in promoting androgen-dependent and -independent apoptosis in testicular cells. However, the molecular mechanisms that underlie the ligand-independent apoptosis, including the activity of AR in testicular stem cells, are not completely understood. In the present study, we generated induced pluripotent stem cells (iPSCs) from bovine testicular cells by electroporation of octamer-binding transcription factor 4 (OCT4). The cells were supplemented with leukemia inhibitory factor and bone morphogenetic protein 4, which maintained and stabilized the expression of stemness genes and pluripotency. The iPSCs were used to assess the apoptosis activity following exposure to phthalate esters, including di (2-ethyhexyl) phthalates, di (n-butyl) phthalate, and butyl benzyl phthalate. Phthalate esters significantly reduced the expression of AR in iPSCs and induced a higher ratio of BAX/BCL-2, thereby favoring apoptosis. Phthalate esters also increased the expression of cyclin-dependent kinase inhibitor 1 (p21(Cip1)) in a p53-dependent manner and enhanced the transcriptional activity of p53. The forced expression of AR and knockdown of p21(Cip1) led to the rescue of the phthalate-mediated apoptosis. Overall, this study suggests that testicular iPSCs are a useful system for screening the toxicity of environmental disruptors and examining their effect on the maintenance of stemness and pluripotency, as well as for identifying the iPSC signaling pathway(s) that are deregulated by these chemicals.


Asunto(s)
Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Ácidos Ftálicos/farmacología , Receptores Androgénicos/metabolismo , Testículo/citología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Bovinos , Reprogramación Celular/efectos de los fármacos , Reprogramación Celular/genética , Células Madre Pluripotentes Inducidas/citología , Masculino , Receptores Androgénicos/genética , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor
5.
Curr Mol Med ; 12(10): 1340-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23016591

RESUMEN

Human amnion-derived cells possess great potential for the repair of human neural disorders, and recent studies have broadened the spectrum for applications because they exhibit the characteristics of multipotent stem cells. These cells express embryonic stem cell markers such as Oct4, Nanog, Sox2, SSEA-3, SSEA-4 and Rex1, and can differentiate into multiple primary germ layers both in vitro and in vivo. Moreover, induced pluripotent stem cells have been generated from amnion-derived cells by virus-mediated delivery of three or four pluripotency-relating transcription factors or by the introduction of only one transcription factor with electroporation. Because human amnion-derived cells are readily available, less likely to contain genetic aberrations and can be reprogrammed earlier and more efficiently than differentiated cells, they can be ideal resources as the donor pluripotent stem cells for therapeutic purposes. We discuss here the highlights of recent studies and potential applications of human amnion-derived multipotent stem cells to stem cell biology as well as to regenerative medicine in the field of aging, heart disease, diabetes and neural disorders.


Asunto(s)
Amnios/citología , Células Madre Pluripotentes Inducidas , Células Madre Multipotentes , Antígenos de Carbohidratos Asociados a Tumores/metabolismo , Diferenciación Celular , Células Madre Embrionarias , Hepatocitos/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Miocitos Cardíacos/metabolismo , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Osteoblastos/metabolismo , Regeneración , Factores de Transcripción SOXB1/metabolismo , Antígenos Embrionarios Específico de Estadio/metabolismo
6.
Curr Mol Med ; 11(6): 439-52, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21663590

RESUMEN

Abnormal and prolonged inflammatory reaction is seen in a wide variety of disorders, and high level of Tumor Necrosis Factor alpha (TNF-α) has been linked to these disorders. Therefore, modulation of TNF-α expression is important in the regulation of inflammatory disorders. In our previous study, we have shown that a transcription factor LPS-induced TNF factor (LITAF) significantly induces TNF-α production. Furthermore, we found that p53 and its synthetic peptide 162-motif specifically downregulate LITAF/TNF-α gene expression in human cells in vitro. Thus, in the present study, the role of p53 in regulating TNF-α-mediated inflammation was investigated. Our data showed that a synthetic peptide, named 162-motif, corresponding to this region functions independently from p53 to cause a significant suppression of TNF-α gene expression in mouse primary macrophages. The 162-motif, when delivered into cells and organs, reduces serum TNF-α level in mice and prevents TNF-α-induced lung lesions and endotoxic shock. Our findings highlight the regulation of LITAF/TNF-α by p53 and its short peptide 162-motif. These in vitro and in vivo observations serve to pave the way for pharmacotherapeutic approaches in the treatment of inflammatory diseases.


Asunto(s)
Proteínas Nucleares/metabolismo , Choque Séptico/metabolismo , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células Cultivadas , Proteínas de Unión al ADN , Regulación hacia Abajo , Regulación de la Expresión Génica , Humanos , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Pulmón/metabolismo , Pulmón/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Regiones Promotoras Genéticas , Choque Séptico/patología , Factores de Transcripción/genética , Transfección , Factor de Necrosis Tumoral alfa/genética
7.
Oncogene ; 29(47): 6245-56, 2010 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-20802531

RESUMEN

We report here a novel role for Jun dimerization protein-2 (JDP2) as a regulator of the progression of normal cells through the cell cycle. To determine the role of JDP2 in vivo, we generated Jdp2-knockout (Jdp2KO) mice by targeting exon-1 to disrupt the site of initiation of transcription. The epidermal thickening of skin from the Jdp2KO mice after treatment with 12-O-tetradecanoylphorbol 13-acetate (TPA) proceeded more rapidly than that of control mice, and more proliferating cells were found at the epidermis. Fibroblasts derived from embryos of Jdp2KO mice proliferated faster and formed more colonies than fibroblasts from wild-type mice. JDP2 was recruited to the promoter of the gene for cyclin-A2 (ccna2) at the AP-1 site. Cells lacking Jdp2 had elevated levels of cyclin-A2 mRNA. Furthermore, reintroduction of JDP2 resulted in the repression of transcription of ccna2 and of cell-cycle progression. Thus, transcription of the gene for cyclin-A2 appears to be a direct target of JDP2 in the suppression of cell proliferation.


Asunto(s)
Ciclo Celular/genética , Ciclina A2/genética , Regulación hacia Abajo , Proteínas Represoras/metabolismo , Animales , Apoptosis/genética , Línea Celular , Proliferación Celular , Ciclina A2/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Epidermis/lesiones , Epidermis/metabolismo , Epidermis/patología , Técnicas de Inactivación de Genes , Genes Reporteros/genética , Ratones , Regiones Promotoras Genéticas/genética , Proteínas Represoras/deficiencia , Proteínas Represoras/genética
8.
Cancer Gene Ther ; 16(2): 126-36, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18818710

RESUMEN

Combination therapy with replicative oncolytic viruses is a recent topic in innovative cancer therapy, but few studies have examined the efficacy of oncolytic adenovirus plus replication-deficient adenovirus carrying a suicide gene. We aim to evaluate whether an E1A, E1B double-restricted oncolytic adenovirus, AxdAdB-3, can improve the efficacy for gallbladder cancers (GBCs) of the replication-deficient adenovirus-based herpes simplex virus thymidine kinase (HSVtk)/ganciclovir (GCV) therapy directed by the carcinoembryonic antigen (CEA) promoter. Cytopathic effects of AxdAdB-3 plus AxCEAprTK (an adenovirus expressing HSVtk directed by CEA promoter) or AxCAHSVtk (an adenovirus expressing HSVtk directed by a nonspecific CAG promoter) with GCV administration were examined in several GBC lines and normal cells. Efficacy in vivo was tested in severe combined immunodeficiency disease mice with GBC xenografts. Addition of AxdAdB-3 (1 multiplicity of infection, MOI) significantly enhanced the cytopathic effects of AxCEAprTK (10 MOI)/GCV on GBC cells. The augmented effect was attributable to the replication of the AxCEAprTK and also to the enhanced CEA promoter activity, which was presumably transactivated by E1A. In normal cells, AxdAdB-3 (20 MOI) plus AxCEAprTK (200 MOI)/GCV was not cytopathic, whereas AxdAdB-3 (1 MOI) plus AxCAHSVtk (10 MOI)/GCV was significantly toxic. Low-dose AxdAdB-3 (2 x 10(7) PFU, plaque-forming unit) plus AxCEAprTK (2 x 10(8) PFU)/GCV significantly suppressed the growth of GBC xenografts as compared with either AxdAdB-3 (2 x 10(7) PFU)/GCV or AxCEAprTK (2 x 10(9) PFU)/GCV alone. E1A, E1B double-restricted replicating adenovirus at low dose significantly augmented the efficacy of CEA promoter-directed HSVtk/GCV therapy without obvious toxicity to normal cells, suggesting a potential use of this combination for treating GBC and other CEA-producing malignancies.


Asunto(s)
Adenoviridae/metabolismo , Proteínas E1A de Adenovirus/metabolismo , Proteínas E1B de Adenovirus/metabolismo , Neoplasias de la Vesícula Biliar/terapia , Terapia Genética/métodos , Viroterapia Oncolítica , Replicación Viral , Adenoviridae/genética , Proteínas E1A de Adenovirus/genética , Proteínas E1B de Adenovirus/genética , Animales , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/metabolismo , Línea Celular Tumoral , Femenino , Neoplasias de la Vesícula Biliar/genética , Neoplasias de la Vesícula Biliar/virología , Células HeLa , Humanos , Ratones , Ratones SCID
9.
J Biol Chem ; 276(43): 39903-10, 2001 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-11546757

RESUMEN

Mannosylerythritol lipid (MEL), a novel extracellular glycolipid from yeast, was found to inhibit the proliferation of mouse melanoma B16 cells in a dose-dependent manner and to induce the apoptosis of B16 cells at concentrations higher than 10 microm (Zhao, X., Wakamatsu, Y., Shibahara, M., Nomura, N., Geltinger, C., Nakahara, T., Murata, T., and Yokoyama, K. K. (1999) Cancer Res. 59, 482-486). We show here that exposure of B16 cells to MEL (5 microm) for 2 days resulted in an increase of the levels of differentiation-associated markers of melanoma cells such as melanogenesis and tyrosinase activity, which were accompanied by morphological changes. The MEL-induced differentiation of B16 cells at this concentration was closely associated with arrest of the cell cycle at G(1) phase, but no significant population of apoptotic cells was identified. Expression of protein kinase Calpha (PKCalpha) was enhanced after exposure of B16 cells to MEL for 48 h. Antisense oligodeoxynucleotides against the mouse gene for PKCalpha prevented MEL-induced melanogenesis in B16 cells. Conversely, the effects of the expression of a constitutively active form of PKCalpha mimicked the effects of MEL on B16 cells. These data suggest that MEL, a yeast-derived glycolipid, triggers the differentiation of B16 melanoma cells through a signaling pathway that involves PKCalpha.


Asunto(s)
Antineoplásicos/farmacología , Glucolípidos/farmacología , Isoenzimas/metabolismo , Melanoma Experimental/tratamiento farmacológico , Proteína Quinasa C/metabolismo , Animales , Antígenos de Diferenciación , Ciclo Celular/efectos de los fármacos , Diferenciación Celular , Melaninas/biosíntesis , Ratones , Imitación Molecular , Monofenol Monooxigenasa/biosíntesis , Oligonucleótidos Antisentido/farmacología , Fosforilación , Proteína Quinasa C-alfa , Células Tumorales Cultivadas
10.
J Histochem Cytochem ; 49(9): 1177-82, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11511686

RESUMEN

Proteinase K is widely used in methods for detection of transcripts in biological specimens by in situ hybridization (ISH). However, treatment with proteinase K hampers detection of RNA and protein simultaneously. We have developed a method for double staining of transcripts and proteins by ISH and IHC staining in imaginal discs and embryos of Drosophila. Instead of treatment with proteinase K, samples are treated with ethanol plus xylene and with acetone. Acetone renders cell membranes permeable to probes and antibodies without damaging tissue integrity, whereas treatment with proteinase K sometimes damages tissues. Treatment of samples with acetone allows hybridization of probe with transcripts in tissue. It is also effective for immunological staining of samples after ISH with a riboprobe. Thus, our method allows detection not only of transcripts but also of specific proteins in relatively intact single samples. (J Histochem Cytochem 49:1177-1182, 2001)


Asunto(s)
Proteínas de Drosophila , Proteínas de Insectos/metabolismo , ARN/metabolismo , Acetona , Animales , Permeabilidad de la Membrana Celular , Drosophila , Embrión no Mamífero , Etanol , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica/métodos , Hibridación in Situ/métodos , Indicadores y Reactivos , Proteínas de Insectos/genética , Larva , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/metabolismo , Proteína Wnt1 , Xilenos
11.
Biochem Biophys Res Commun ; 286(5): 1087-97, 2001 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-11527412

RESUMEN

Expression of the DCC (deleted in colorectal cancer) protein is strongly induced during the neural differentiation of mouse P19 embryonal carcinoma (EC) cells that occurs when these cells are treated with retinoic acid (RA). Myc-associated zinc finger protein (MAZ) is a DNA-binding protein that is widely expressed and functions in human, mouse and hamster cells as an activator, an initiator or a terminator of transcription. However, the biological functions of MAZ remain elusive. We report here that MAZ associates with the cytoplasmic domain of the DCC protein in vivo and in vitro. Yeast two-hybrid assays confirmed this association. An immunofluorescence study demonstrated that DCC protein is expressed at elevated levels in neuron-like P19 EC cells, in particular in axons, in which the MAZ protein is also expressed. We found that MAZ was translocated from the nucleus to the cytoplasm during the RA-induced terminal differentiation of P19 EC cells with resultant loss of the ability of MAZ to bind to the ME1a1 site of the c-myc promoter. Taken together, our observations imply that the DCC protein might play a critical role as a signaling molecule in the regulation of the transcriptional activity of MAZ during the neural differentiation of P19 EC cells.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Neuronas/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Sitios de Unión , Northern Blotting , Western Blotting , Diferenciación Celular , Línea Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Receptor DCC , Proteínas de Unión al ADN , Genes myc/genética , Humanos , Inmunohistoquímica , Microscopía Fluorescente , Modelos Biológicos , Datos de Secuencia Molecular , Plásmidos/metabolismo , Pruebas de Precipitina , Regiones Promotoras Genéticas , Unión Proteica , Estructura Terciaria de Proteína , ARN/metabolismo , ARN Mensajero/metabolismo , Receptores de Superficie Celular , Proteínas Recombinantes de Fusión/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Transducción de Señal , Factores de Tiempo , Transcripción Genética , Transfección , Tretinoina/metabolismo , Técnicas del Sistema de Dos Híbridos , Xenopus , Dedos de Zinc
12.
Biochem Biophys Res Commun ; 281(4): 1041-4, 2001 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-11237769

RESUMEN

To study the Hoxa-13 gene in the liver, we examined its expression by RT-PCR in various liver cell lines, rat livers under different conditions, and human primary hepatocellular carcinomas (HCCs). The gene was found to be expressed in cell lines originating from liver stem-like cells, but not in cell lines originating from hepatocytes and bile duct epithelia. Expression was induced in rat livers after treatment with d-galactosamine, which is known to induce oval cell proliferation, but not after a two-thirds partial hepatectomy (2/3 PH) where induction of oval cell proliferation is thought not to occur. Expression of the gene correlated with human HCC samples associated with Hepatitis B or C virus infection in this small series. These results suggest that the Hoxa-13 gene may provide a potentially useful tool for elucidation of mechanisms involved in lineage-specific differentiation and carcinogenesis of liver stem cells.


Asunto(s)
Carcinoma Hepatocelular/genética , Proteínas de Homeodominio/genética , Neoplasias Hepáticas/genética , Animales , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/virología , Línea Celular , Galactosamina/farmacología , Regulación Neoplásica de la Expresión Génica , Hepacivirus , Hepatectomía , Virus de la Hepatitis B , Hepatitis Viral Humana/genética , Hepatitis Viral Humana/virología , Humanos , Hígado/citología , Hígado/efectos de los fármacos , Hígado/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
13.
J Biol Chem ; 276(23): 19897-904, 2001 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-11259406

RESUMEN

The transcription factors Sp1 and MAZ (Myc-associated zinc finger protein) contain several zinc finger motifs, and each functions as both a positive and a negative regulator of gene expression. In this study, we characterized the extremely GC-rich promoter of the human gene for MAZ, which is known as a housekeeping gene. Unique symmetrical motifs in the promoter region (nucleotides -383 to -334) were essential for the expression of the gene for MAZ, whereas an upstream silencer element (nucleotides -784 to -612) was found to act in a position-dependent but orientation-independent manner. Sp1 and MAZ bound to the same cis-elements in the GC-rich promoter, apparently sharing DNA-binding sites. The relative extent of binding of Sp1 and MAZ to these cis-elements corresponded to the extent of negative regulation of the expression of the gene for MAZ in various lines of cells. Furthermore, novel repressive domains in both Sp1 (amino acids 622-788) and MAZ (amino acids 127-292) were identified. Suppression by Sp1 and suppression by MAZ were independent phenomena; histone deacetylases were involved in the autorepression by MAZ itself, whereas DNA methyltransferase 1 was associated with suppression by Sp1. Our results indicate that both deacetylation and methylation might be involved in the regulation of expression of a single gene via the actions of different zinc finger proteins that bind to the same cis-elements.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Proteínas Proto-Oncogénicas c-myc/fisiología , Factor de Transcripción Sp1/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Animales , Secuencia de Bases , Línea Celular , ADN , Proteínas de Unión al ADN , Histona Desacetilasas/metabolismo , Humanos , Datos de Secuencia Molecular , Pruebas de Precipitina , Regiones Promotoras Genéticas , Dedos de Zinc
14.
Eur J Biochem ; 268(2): 374-83, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11168372

RESUMEN

Rat pheochromocytoma PC12 cells undergo neuronal differentiation in response to nerve growth factor (NGF). The differentiation involves protein kinase cascades that include the kinases MEK and ERK, as well as activation of the transcription factors c-Jun and c-Fos. We show here, that exposure of PC12 cells to mannosylerythritol lipid (MEL), a yeast extracellular glycolipid, enhances the activity of acetylcholinesterase and interrupts the cell cycle at the G1 phase, with resulting outgrowth of neurites and partial cellular differentiation. Treatment with MEL stimulates the phosphorylation of ERK to a similar extent as treatment with NGF, although, the appearance of phosphorylated ERK is somewhat delayed. Both the MEL-induced outgrowth of neurites and the increase in the activity of acetylcholinesterase are prevented by PD98059, a specific inhibitor of MEK. Northern blotting analysis of c-jun transcripts and analysis of transcription in PC12 cells of a c-jun/CAT reporter construct demonstrated a significant increase in the rate of transcription of the c-jun gene upon treatment with MEL. The sequence elements required for the MEL-mediated activation of transcription of the c-jun gene are located between nucleotides -126 and -79 in the 5' flanking region. Our results suggest that MEL induces characteristics of neuronal differentiation in PC12 cells, with transactivation of the c-jun gene, via an ERK-related signal cascade that is partially overlapping the pathways activated in response to NGF. These results might provide the groundwork for the use of microbial extracellular glycolipids as novel reagents for the treatment of cancer cells.


Asunto(s)
Eritritol/análogos & derivados , Glucolípidos/farmacología , Sistema de Señalización de MAP Quinasas , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neuronas/metabolismo , Acetilcolinesterasa/biosíntesis , Animales , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Eritritol/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos , Neuritas , Neuronas/citología , Células PC12 , Proteínas Proto-Oncogénicas c-jun/biosíntesis , Proteínas Proto-Oncogénicas c-jun/genética , Ratas , Transcripción Genética , Proteínas Quinasas p38 Activadas por Mitógenos
15.
Hepatology ; 32(4 Pt 1): 721-7, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11003615

RESUMEN

Human chromosome 8p23 is known as a region that is associated with loss of heterozygosity (LOH), which is frequently deleted in hepatocellular carcinoma (HCC) tissues. We report here the characterization of a gene for a liver-related putative tumor suppressor (LPTS) localized at 8p23, that was isolated by allelic-loss mapping and positional candidate cloning. The expression of the gene for LPTS was ubiquitous in normal human tissues, albeit at relatively low levels, whereas levels appeared to be significantly reduced, or sometimes undetectable in HCC cells and neoplastic tissues. Thus, it appeared that LPTS might be involved in the control of cell proliferation. Indeed, we observed the significant suppression of growth and growth arrest of SMMC-7721 HCC cells after introduction of the gene for LPTS. We also used antisense oligodeoxynucleotides (AS-ODNs) to suppress the expression of LPTS in normal liver cells L02. Several AS-ODNs specific for LPTS mRNA significantly enhanced cell growth, whereas control oligodeoxynucleotides (ODNs) did not. Our results suggest that LPTS might be a growth-inhibitory protein in human hepatocytes.


Asunto(s)
Carcinoma Hepatocelular/genética , Cromosomas Humanos Par 8 , Genes Supresores de Tumor , Neoplasias Hepáticas/genética , Pérdida de Heterocigocidad , Secuencia de Aminoácidos , Secuencia de Bases , Carcinoma Hepatocelular/patología , División Celular , Humanos , Neoplasias Hepáticas/patología , Datos de Secuencia Molecular
16.
Cytotechnology ; 33(1-3): 123-30, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19002819

RESUMEN

Mannosylerythritol lipid (MEL), an extracellularglycolipid from yeast, induces the differentiation ofHL-60 promyelocytic leukemia cells towardsgranulocytes. We show here that MEL is also a potentinhibitor of the proliferation of mouse melanoma B16cells. Flow-cytometric analysis of the cell cycle ofMEL-treated B16 cells revealed the accumulation ofcells in the sub-G(0)/G(1) phase, which is a hallmark ofcells undergoing apoptosis. Treatment of B16 cellsfor 24 h with phorbol 12-myristate 13-acetate (PMA),an activator of protein kinase C (PKC), did notinterfere with the growth and survival of the cells,but it effectively counteracted the MEL-induced growtharrest and apoptosis. The activity of PKC was reducedin B16 cells treated with MEL at a concentration atwhich MEL induced apoptosis. However, incubation withPMA in addition to MEL reversed this reduction in theactivity of PKC. These results suggest thatconverging signaling pathways are triggeredindependently by MEL and PMA and that the signalsmight both be mediated by PKC.

17.
Cytotechnology ; 33(1-3): 247-51, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19002832

RESUMEN

We report here that a microbial extracellular glycolipid,mannosylerythritol lipid (MEL), induces the outgrowth ofneurites from and enhances the activity of acetylcholinesterase(AChE) in PC12 pheochromocytoma cells. Furthermore, treatment ofPC12 cells with MEL increased levels of galactosylceramide(Galbeta1-1'Cer; GalCer). Exposure of PC12 cells to exogenous GalCer caused the dose-dependent outgrowth ofneurites. By contrast, treatment of PC12 cells with nerve growthfactor (NGF) did not increase the level of GalCer in the cells. The neurite-related morphological changes induced by GalCerdifferend from those induced by NGF, indicating differencesbetween the signal transduction pathways triggered by NGF and by GalCer.

18.
Cytotechnology ; 33(1-3): 259-64, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19002834

RESUMEN

Four analogs of succinoyl trehalose lipid-3 (STL-3)with saturated even-number or odd-number carbonchains, and unsaturated or halogenated fatty acidswere examined for their ability to inhibit the growthand induce the differentiation of HL-60 humanpromyelocytic leukemia cells. The optimalconcentration of STL-3 at which such activities wererecognized was closed to the critical micelleconcentration of STL-3. Analog of STL-3 witheven-number or odd-number carbon chain and unsaturatedfatty acids strongly inhibited growth and induced thedifferentiation of HL-60 cells, as evaluated in termsof nitroblue tetrazilium-reducing activity and theappearance of the CD36 antigen. An analog of STL-3with halogenated fatty acids significantly inhibitedproliferation but only induced the differentiation ofHL-60 cells. Our results indicate that the effects ofSTL-3 and its analogs on HL-60 cells depend on thestructure of the hydrophobic moiety of STL-3.

19.
J Mol Med (Berl) ; 77(6): 481-94, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10475063

RESUMEN

We have characterized an element (differentiation response element, DRE) in the promoter region of the c-jun gene that is both necessary and sufficient for retinoic acid (RA) and adenovirus early region (E1A) mediated up-regulation of c-jun gene expression during the differentiation of F9 cells. The DRF complex, which binds specifically to DRE, is composed of the E1A-associated protein p300 and the activation transcription factor-2 (ATF-2) as a DNA-binding subunit of the DRF. The molecular association of p300 and ATF-2 enhances the transcription of the c-jun gene, which requires protein kinase C alpha mediated phosphorylation of Ser-121 of ATF-2 within its p300 interaction domain. We used antisense oligodeoxynucleotides (AS-ODNs) capable of binding specifically to the mRNA for either p300 or CBP to examine the individual roles of p300 and CBP during the RA-induced differentiation, exit from the cell cycle, and apoptosis of F9 cells. F9 cells treated with AS-ODNs specific for p300 mRNA became resistant to RA-induced differentiation, while cells incubated with AS-ODNs specific for CBP mRNA were still able to differentiate. Despite their similarities p300 and CBP appear to have distinct functions during the differentiation of F9 cells. These results suggest that ATF-2 and p300 cooperate in the control of transcription by forming a protein complex in response to RA or E1A, and that the phosphorylation of ATF-2 and p300 is probably a signaling event in the pathway that leads to the transactivation of the c-jun gene in F9 cell differentiation.


Asunto(s)
Diferenciación Celular/genética , Proteínas Nucleares/metabolismo , Transactivadores/metabolismo , Animales , Secuencia de Bases , Proteína de Unión a CREB , Diferenciación Celular/fisiología , División Celular/genética , División Celular/fisiología , Proteína p300 Asociada a E1A , Regulación de la Expresión Génica , Ratones , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Fosforilación , Transactivadores/genética , Células Tumorales Cultivadas
20.
Biochem Biophys Res Commun ; 262(1): 198-205, 1999 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-10448092

RESUMEN

Myc-associated zinc finger protein (MAZ) is a transcription factor that contains proline-rich, alanine repeats and six C(2)H(2)-type zinc finger motifs, as well as five putative sites of phosphorylation by casein kinase II (CKII). Site-specific mutagenesis of MAZ revealed that the serine residue at position 480 was the major site of phosphorylation by CKII both in vitro and in vivo. Phosphorylation of MAZ by CKII at this serine residue was required for maximum binding of MAZ to the pyrimidine-rich DNA of the nuclease-hypersensitive element (NHE) in the 5'-end promoter region of the c-myc gene. Mutation of serine at position 480 to alanine eliminated the DNA-binding activity of MAZ to this element. Moreover, the mutated MAZ was unable to enhance the expression of luciferase encoded by a c-myc promoter/luciferase reporter gene in HeLa cells in the presence of CKII. These results suggest that phosphorylation of the serine residue at position 480 of MAZ by CKII can control the function of MAZ by altering its DNA-binding activity.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/metabolismo , Sustitución de Aminoácidos , Quinasa de la Caseína II , Núcleo Celular/metabolismo , ADN/genética , ADN/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Genes Reporteros , Genes myc/genética , Células HeLa , Humanos , Fosforilación , Fosfoserina/metabolismo , Regiones Promotoras Genéticas/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Elementos de Respuesta/genética , Factores de Transcripción/química , Factores de Transcripción/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA