Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Oral Biosci ; 66(1): 217-224, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38147910

RESUMEN

OBJECTIVES: Krüppel-like factor (KLF)5, which is overexpressed in carcinomas such as oral cancer, inhibits epidermal differentiation. KLF5 induces dedifferentiation of carcinoma cells, which effectuates carcinoma progression; nevertheless, the regulatory mechanism affecting the transcription of the KLF5 gene remains ambiguous. METHODS: Transcriptional activity of the KLF5 silencer, specifically the 425-bp region (425-region), was examined using reporter assays. An additional analysis was conducted to assess the impact of the minimal essential region (MER) of KLF5 on its basal expression. The affinity of cAMP responsive element binding protein 1 (CREB1) for three potential CREB1-binding sites in the 425-region was analyzed using DNA pull-down and quantitative chromatin immunoprecipitation assays. Reporter assays employing a human oral squamous carcinoma cell line, HSC2, transfected with small interfering RNA or complementary DNA for CREB1, were performed to investigate the effect of CREB1 binding sites on MER activity. RESULTS: The 425-region exhibited no transcriptional activity and suppressed MER transcriptional activity. This region encodes three putative CREB1-binding sites, and CREB1 demonstrated equal binding affinity for all three sites. The deletion of each of these binding sites reduced CREB1 precipitation and enhanced MER activity. Endogenous CREB1 knockdown and overexpression elevated and reduced MER activity, respectively, at the intact sites. Conversely, site deletion hampered and improved MER activity upon CREB1 knockdown and overexpression, respectively. CONCLUSIONS: Suppression of KLF5 basal expression via CREB1 binding to the 425-region requires all three CREB1-binding sites to remain intact in oral carcinoma cells. Consequently, deletion of the CREB1-binding site relieves suppression of KLF5 basal expression.


Asunto(s)
Carcinoma , Neoplasias de la Boca , Humanos , Línea Celular Tumoral , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Regiones Promotoras Genéticas/genética , Sitios de Unión/genética , Neoplasias de la Boca/genética , Carcinoma/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo
2.
Tumour Biol ; 45(1): 81-94, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37694332

RESUMEN

BACKGROUND: Krüppel-like factor 5 (KLF5) is a transcription factor regulating the proliferation and differentiation of epithelial cells, and its uncontrolled expression is closely associated with carcinoma progression. Sp3 binding to the minimal essential region (MER) of KLF5 gene is critical for KLF5 basal expression, but the expression control mechanism is unknown. OBJECTIVE: This study aimed to identify a regulatory region for KLF5 basal expression and the binding protein in carcinoma cells by analyzing the promoter upstream region. METHODS: Reporter assays determined the silencer region. The protein binding to the region was identified by database analysis and ChIP assay. The protein mediating the interaction between the region and the MER was confirmed through chromosome conformation capture (3 C) on ChIP assay. The effects of the protein on KLF5 expression were analyzed using qRT-PCR and western blot. RESULTS: Reporter assay localized the 425-region from upstream KLF5 gene as the silencer. Database analysis and ChIP assay found CREB1 binding to the 425-region. CREB1 siRNA or mutation of CREB1-binding site in the 425-region increased luciferase activities and decreased the binding to 425-region. 3 C on ChIP assay showed that CREB1 mediated interaction of the 425-region and the MER. CREB1 overexpression decreased endogenous KLF5 expression and luciferase activity. CONCLUSIONS: The 425-region is the silencer of KLF5 basal expression, and CREB1 binding suppresses the expression.


Asunto(s)
Carcinoma , Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Factores de Transcripción de Tipo Kruppel , Humanos , Diferenciación Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Factores de Transcripción de Tipo Kruppel/genética , Luciferasas
3.
Biochem Biophys Res Commun ; 590: 68-74, 2022 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-34973532

RESUMEN

The mutation and deletion of high mobility group AT-hook 2 (Hmga2) gene exhibit skeletal malformation, but almost nothing is known about the mechanism. This study examined morphological anomaly of facial bone in Hmga2-/- mice and osteoblast differentiation of pre-osteoblast MC3T3-E1 cells with Hmga2 gene knockout (A2KO). Hmga2-/- mice showed the size reduction of anterior frontal part of facial bones. Hmga2 protein and mRNA were expressed in mesenchymal cells at ossification area of nasal bone. A2KO cells differentiation into osteoblasts after reaching the proliferation plateau was strongly suppressed by alizarin red and alkaline phosphatase staining analyses. Expression of osteoblast-related genes, especially Osterix, was down-regulated in A2KO cells. These results demonstrate a close association of Hmga2 with osteoblast differentiation of mesenchymal cells and bone growth. Although future studies are needed, the present study suggests an involvement of Hmga2 in osteoblast-genesis and bone growth.


Asunto(s)
Desarrollo Óseo , Diferenciación Celular , Huesos Faciales/crecimiento & desarrollo , Proteína HMGA2/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Animales , Línea Celular , Proliferación Celular , Forma de la Célula , Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteína HMGA2/genética , Ratones Noqueados
4.
Biochem Biophys Res Commun ; 542: 24-28, 2021 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-33482470

RESUMEN

Loss of mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) expression closely associates with increased aggressive behaviors of oral carcinoma cells. It emphasizes that a mechanism to suppress the expression is an important subject for understanding carcinoma progression pathway. However, nothing is known at present. This study conducted on transcriptional regulation of the gene down-regulation. Reporter assays showed the presence of the silencer region between +402 and +501 region of MALT1 gene in oral carcinoma cells. It encoded a binding site of nuclear factor-κB subunit, RELA. RELA binding to the site was confirmed by the chromatin immunoprecipitation analyses, and deletion and mutations of the site significantly decreased the RELA binding. Short interfering RNAs for RELA up-regulated reporter gene and endogenous MALT1 protein expressions, and deletion and mutations of RELA binding site increased reporter gene expression. These results demonstrated RELA-binding to the site suppresses MALT1 expression that may facilitate oral carcinoma progression.

5.
Biochem Biophys Res Commun ; 522(3): 799-804, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-31791579

RESUMEN

Mucosa-associated lymphoid tissue lymphoma translocation 1 protein (MALT1) consisting of death domain, Ig-like domains and caspase-like domain is expressed in nucleus of oral carcinoma cells, and loss of the expression closely associates with disease progression and stimulates proliferation of the cells. However, nothing is known about the molecular backgrounds. In this study, eight constructs with different domain constitution of human MALT1 and six constructs were transiently and stably transfected into oral carcinoma cell lines, respectively. The immunoblot analysis showed that constructs containing caspase-like domain was expressed in nucleus and the domain-deleted constructs in cytoplasm. Immunocytochemistry of stably transfected HSC2 oral carcinoma cells confirmed the caspase-like domain-dependent nuclear localization. Involvement of domains in proliferation of stably transfected HSC2 cells was quantified by the real-time and conventional colorimetric assays. In contrast to suppression of the proliferation by full-length wild-type MALT1, any domain-deleted constructs enhanced the proliferation. Death domain construct without caspase-like domain suppressed the proliferation when it was localized in nucleus by ligating with the nuclear localization signal. These results demonstrate that nuclear localization of MALT1 in oral carcinoma cells depends on the presence of caspase-like domain and that death domain nuclear entity is responsible for MALT1 inhibition of oral carcinoma cell proliferation. Nuclear localization of death domain led by caspase-like domain may suppress oral carcinoma progression.


Asunto(s)
Neoplasias de la Boca/patología , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/análisis , Línea Celular Tumoral , Núcleo Celular/patología , Proliferación Celular , Humanos , Dominios Proteicos
6.
Biochem Biophys Res Commun ; 509(4): 1008-1014, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30654938

RESUMEN

Tooth formation is accomplished under strict genetic programs. Although patients with chromosome 12q14 aberration shows tooth phenotype including the size and eruption timing with bone growth anomaly, its etiology is uncertain. Here, we examined expression of Hmga2, which is encoded at chromosome 12q14, in mouse tooth germs and analyzed the involvement in lower first molar (M1) and mandibular bone development. Hmga2 expression was immunohistochemically detected at enamel organ and the surrounding mesenchyme of the M1 germs. The expression was dynamically changed with gestation and rapidly decreased in postnatal mice. In Hmga2-/- mice, the M1 germs and crowns were diminished in size, and formation and eruption of molars were delayed with mandibular bone growth retardation. Hmga2 cDNA or siRNA transfection showed that Hmga2 transcriptionally up-regulates expression of stem cell factors, Sox2 and Nanog. They were co-localized with Hmga2 in the germs, but differentially distributed at enamel organ and mesenchyme in Hmga2-/- mice. These results demonstrate that Hmga2 expressed in tooth germs regulates the growth, sizing and eruption and stem cell factor expression in different compartment of the germ and associates with mandibular bone growth. Although future studies are needed, the present study demonstrates HMGA2 regulation of tooth genesis with skeletal development.


Asunto(s)
Proteína HMGA2/fisiología , Proteína Homeótica Nanog/metabolismo , Factores de Transcripción SOXB1/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica , Proteína HMGA2/análisis , Proteína HMGA2/metabolismo , Inmunohistoquímica , Mandíbula/crecimiento & desarrollo , Ratones , Diente Molar/crecimiento & desarrollo , Odontogénesis/efectos de los fármacos
7.
Biochem Biophys Res Commun ; 508(2): 405-409, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30503342

RESUMEN

An increase in the vasculature is one of most representative changes in the synovial tissue of joints in rheumatoid arthritis (RA) and is closely associated with disease progression. Although the vasculatures are believed to be a result of VE-cadherin-dependent angiogenesis and a possible therapeutic target of the disease, synovial fibroblastic cells express VE-cadherin and form tube-like structures, suggesting that vasculatures in RA synovium may not simply result from angiogenesis. This paper analyzes a mechanism of VE-cadherin expression by rheumatoid arthritic synovial fibroblast-like cells (RSFLs) and their involvement in the tube-like formation. A representative angiogenic factor, vascular endothelial growth factor (VEGF), and its binding to a predominant receptor (VEGFR2) activated VE-cadherin expression and the signaling pathways of ERK/MAPK and PI3K/AKT/mTOR. Treatment of RSFLs with signaling pathway inhibitors, VEGFR2 siRNA and a VEGF-antagonizing mimicking peptide inhibited VE-cadherin expression dose-dependently. VEGF-stimulated tube-like formation by RSFLs on Matrigel was hindered by the mimicking peptide and inhibitor treatment. This data demonstrates that RSFLs activated by VEGF binding of VEGFR2 express VE-cadherin and formed tube-like structure under the control of ERK/MAPK and PI3K/AKT/mTOR pathways suggesting that the inhibition suppresses vascular development in RA synovium.


Asunto(s)
Antígenos CD/metabolismo , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Cadherinas/metabolismo , Membrana Sinovial/metabolismo , Membrana Sinovial/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Sistema de Señalización de MAP Quinasas , Neovascularización Patológica , Fosfatidilinositol 3-Quinasas/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal , Membrana Sinovial/irrigación sanguínea , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
8.
Gene ; 601: 36-43, 2017 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-27940107

RESUMEN

Krüppel-like factor 5 (KLF5) transcriptionally controls the proliferation-differentiation balance of epithelium and is overexpressed in carcinomas. Although genomic region modifying KLF5 expression is widespread in different types of cells, the region that commonly regulates basal expression of the genes across cell-types is uncertain. In this study we determined the minimal essential region for the expression and its regulatory transcription factors using oral carcinoma cells. A reporter assay defined a 186bp region downstream of the transcription start site and a cluster of six GC boxes (GC1-GC6) as the minimal essential region. Mutation in the GC1 or GC6 regions but not other GC boxes significantly decreased the reporter expression. The decrease by the GC1 mutation was reproduced in the 2kbp full-length promoter, but not by the GC6 mutation. Additionally, specificity proteins (Sp) that can be expressed in epithelial cells and bind GC box, Sp3 co-localized with KLF5 in oral epithelium and carcinomas and chromatin immunoprecipitation analyses showed Sp3 as the prime GC1-binding protein. Inhibition of Sp-GC box binding by mithramycin A and knockdown of Sp3 by the short interfering RNA decreased expression of the reporter gene and endogenous KLF5. These data demonstrate that a 186bp region is the minimal essential region and that Sp3-GC1 binding is essential to the basal expression of KLF5.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Factor de Transcripción Sp3/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Células Epiteliales/citología , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Factores de Transcripción de Tipo Kruppel/química , Regiones Promotoras Genéticas , Unión Proteica , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción Sp3/genética , Sitio de Iniciación de la Transcripción
9.
BMC Oral Health ; 16: 13, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26847634

RESUMEN

BACKGROUND: Krüppel-like factor 4 (KLF4) is a transcription factor regulating proliferation-differentiation balance of epithelium, and down-regulated in less-differentiated and advanced oral carcinomas. Although the expression is inactivated by the promoter hypermethylation in malignant tumor cells, it remains unknown in oral carcinoma cells. METHODS: Genomic DNA isolated from nine different oral carcinoma cell lines and a normal keratinocyte line was treated with sodium bisulfite, and methylation at KLF4 gene promoter was determined by PCR direct-sequence analysis. KLF4 expression in cells cultured with or without demethylation reagent was monitored by quantitative real-time PCR and immunoblot. RESULTS: A 237-bp promoter region spanning - 718 and - 482 of KLF4 gene was hypermethylated in oral carcinoma cells that express KLF4 at a low level, but the methylation was infrequent in cells expressing KLF4 high amount. The downstream region from - 481 to +192 was not methylated in any cell lines. Demethylation treatment of cells up-regulated the expression at mRNA and protein levels. CONCLUSION: This study demonstrated that hypermethylation at a narrow range of the promoter region down-regulates KLF4 expression, and suggests that the loss of expression by the hypermethylation contributes to oral carcinoma progression.


Asunto(s)
Metilación de ADN , Factores de Transcripción de Tipo Kruppel/metabolismo , Neoplasias de la Boca/metabolismo , Regiones Promotoras Genéticas , Carcinoma/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Factor 4 Similar a Kruppel
10.
Int J Clin Exp Pathol ; 8(4): 3701-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26097551

RESUMEN

Proliferation-differentiation balance of epithelial cells is regulated by Krüppel-like factors (KLF) 4 and 5, and the unbalanced expression relates to carcinoma progression. However, little is known about the expression and role in oral carcinomas. This study examined expression of KLF4 and KLF 5 in the carcinomas by immunohistochemistry (n = 67) and the involvement in proliferation and differentiation of carcinoma cells. KLF4 was detected in keratinizing carcinoma cells and KLF5 in non-keratinizing cells. KLF4 staining declined in the patient with lymph node metastasis (P < 0.05) and in parallel with the histological dedifferentiation (P = 0.09). Exogenous overexpression of KLF4 arranged cells in a cobble-like structure with desmosomes and KLF5 elongated cells like fibroblasts without desmosomes. KLF4 suppressed fibronectin expression, and KLF5 down-regulated and degraded E-cadherin. The proliferation was not affected by KLFs. Thus, down-regulation of KLF4 and up-regulation of KLF5 may stimulate oral carcinoma progression through the dedifferentiation of carcinoma cells.


Asunto(s)
Carcinoma/metabolismo , Desdiferenciación Celular/genética , Diferenciación Celular/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Neoplasias de la Boca/metabolismo , Anciano , Cadherinas/metabolismo , Carcinoma/genética , Carcinoma/patología , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Femenino , Humanos , Factor 4 Similar a Kruppel , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Regulación hacia Arriba
11.
J Oral Pathol Med ; 44(9): 656-68, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25765182

RESUMEN

BACKGROUND: Oral squamous cell carcinoma exhibits a poor prognosis, caused by aggressive progression and early-stage metastasis to cervical lymph nodes. Here, we developed a xenograft mouse model to explore the heterogeneity of the tumor microenvironment that may govern local invasion and nodal metastasis of tumor cells. METHODS: We transplanted five oral carcinoma cell lines into the tongues of nude mice and determined tongue tumor growth and micrometastatic dissemination by serially sectioning the tongue and lymph node lesions in combination with immunohistochemistry and computer-assisted image analysis. Our morphometric analysis enabled a quantitative assessment of blood and lymphatic endothelial densities in the intratumoral and host stromal regions. RESULTS: All cell lines tested were tumorigenic in mouse tongue. The metastatic lesion-derived carcinoma cell lines (OSC19, OSC20, and HSC2) yielded a 100% nodal metastasis rate, whereas the primary tumor-derived cell lines (KOSC2 and HO-1-u-1) showed <40% metastatic potential. Immunohistochemistry showed that the individual cell lines gave rise to heterogeneous tumor architecture and phenotypes and that their micrometastatic lesions assimilated the immunophenotypic properties of the corresponding tongue tumors. Notably, OSC19 and OSC20 cells shared similar aggressive tumorigenicity in both the tongue and lymph node environments but displayed markedly diverse immunophenotypes and gene expression profiles. CONCLUSIONS: Our model facilitated comparing the tumor microenvironments in tongue and lymph node lesions. The results support that tumorigenicity and tumor architecture in the host tongue environment depend on the origin and properties of the carcinoma cell lines and that metastatic progression may take place through heterogeneous tumor-host interactions.


Asunto(s)
Carcinoma de Células Escamosas/patología , Modelos Animales de Enfermedad , Neoplasias de Cabeza y Cuello/patología , Neoplasias de la Lengua/patología , Animales , Línea Celular Tumoral , Femenino , Xenoinjertos , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Metástasis Linfática , Ratones , Ratones Desnudos , Micrometástasis de Neoplasia , Trasplante de Neoplasias , Fenotipo , Carcinoma de Células Escamosas de Cabeza y Cuello , Transcriptoma , Células Tumorales Cultivadas , Microambiente Tumoral
12.
Mol Clin Oncol ; 2(1): 19-25, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24649302

RESUMEN

Tongue carcinomas are common malignancies of the oral cavity. Understanding the molecular mechanisms behind the disease progression is a prerequisite for improving patient prognosis. Fatty acid-binding proteins (FABPs) are cytoplasmic lipid chaperones that affect cellular organization and energy production. Although their aberrant expression is involved in carcinoma progression, its role in the pathology of tongue carcinomas remains unclear. In the present study, the immunohistochemical expression of FABP4 and FABP5 in tongue carcinomas (n=58) and its involvement in the clinicopathological parameters were examined. Normal tongue epithelial cells expressed FABP5, an epidermal-type FABP, but not FABP4, an adipocyte-type FABP. The cytoplasmic staining of FABP5 was increased in carcinomas with advanced T-stage (P<0.05) and clinical stage (P<0.05). Ectopic expression of FABP4 was detected in almost all carcinomas, although its role in disease progression remains undetermined. Upregulation of FABP5 in the wounded skin of genetically normal mice indicated that microenvironmental tissue factors induce FABP5 expression. The results of the present study demonstrated the aberrant expression of FABP4 and FABP5 in tongue carcinomas and suggested the involvement of FABP5 in disease progression.

13.
PLoS One ; 8(8): e69777, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23936352

RESUMEN

The binding of p120-catenin and ß-catenin to the cytoplasmic domain of E-cadherin establishes epithelial cell-cell adhesion. Reduction and loss of catenin expression degrades E-cadherin-mediated carcinoma cell-cell adhesion and causes carcinomas to progress into aggressive states. Since both catenins are differentially regulated and play distinct roles when they dissociate from E-cadherin, evaluation of their expression, subcellular localization and the correlation with E-cadherin expression are important subjects. However, the same analyses are not readily performed on squamous cell carcinomas in which E-cadherin expression determines the disease progression. In the present study, we examined expression and subcellular localization of p120-catenin and ß-catenin in oral carcinomas (n = 67) and its implications in the carcinoma progression and E-cadherin expression using immunohitochemistry. At the invasive front, catenin-membrane-positive carcinoma cells were decreased in the dedifferentiated (p120-catenin, P < 0.05; ß-catenin, P < 0.05) and invasive carcinomas (p120-catenin, P < 0.01; ß-catenin, P < 0.05) and with the E-cadherin staining (p120-catenin, P < 0.01; ß-catenin, P < 0.01). Carcinoma cells with ß-catenin cytoplasmic and/or nuclear staining were increased at the invasive front compared to the center of tumors (P < 0.01). Although the p120-catenin isoform shift from three to one associates with carcinoma progression, it was not observed after TGF-ß, EGF or TNF-α treatments. The total amount of p120-catenin expression was decreased upon co-treatment of TGF-ß with EGF or TNF-α. The above data indicate that catenin membrane staining is a primary determinant for E-cadherin-mediated cell-cell adhesion and progression of oral carcinomas. Furthermore, it suggests that loss of p120-catenin expression and cytoplasmic localization of ß-catenin fine-tune the carcinoma progression.


Asunto(s)
Cadherinas/metabolismo , Cateninas/metabolismo , Membrana Celular/metabolismo , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Boca/metabolismo , beta Catenina/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Persona de Mediana Edad , Neoplasias de la Boca/patología , Transporte de Proteínas/efectos de los fármacos , Catenina delta
14.
Int J Oncol ; 43(3): 729-36, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23799590

RESUMEN

Progression of oral carcinomas associates with aberrant activation and inactivation of molecules that work in established or unknown pathways. Although mucosa­associated lymphoid tissue 1 (MALT1) expressed in normal oral epithelium is inactivated in the aggressive subset of carcinomas with worse prognosis, phenotypic changes of carcinoma cells upon the loss of expression is unknown. We performed a proteomic analysis to identify MALT1­regulated proteins in oral carcinoma cells. Four different keratins were included in the ten most abundantly changed proteins. K8/18 were upregulated in MALT1 stably­expressing carcinoma cells and K5/14 in MALT1­marginal control cells. K8/18 upregulation and K5/14 downregulation were MALT1 dose­dependent and observed in a series of oral carcinoma cells. MALT1 suppressed cell proliferation (0.52-fold, P<0.01) and its dominant-negative form stimulated it (1.33-fold, P<0.01). The decreased proliferation associated with reduction of cyclin D1, which was recovered by the short interfering RNA against MALT1. Taken together, loss of MALT1 expression alters keratin expression and enhances proliferation of carcinoma cells, and may progress oral carcinomas into the advanced state.


Asunto(s)
Carcinoma/genética , Caspasas/genética , Queratinas/biosíntesis , Neoplasias de la Boca/genética , Proteínas de Neoplasias/genética , Carcinoma/patología , Caspasas/biosíntesis , Línea Celular Tumoral , Proliferación Celular , Ciclina D1/biosíntesis , Epitelio/metabolismo , Epitelio/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Queratinas/genética , Queratinas/metabolismo , Neoplasias de la Boca/patología , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas , Membrana Mucosa/metabolismo , Membrana Mucosa/patología , Proteínas de Neoplasias/biosíntesis , Proteómica/métodos , Regulación hacia Arriba
15.
Odontology ; 101(2): 150-5, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22752732

RESUMEN

Oral carcinoma patients with inactivation of mucosa-associated lymphoid tissue 1 (MALT1) expression worsen their prognoses. Although the genetic mutation could be responsible for the inactivation, no information is available at present. In the present study, genomic DNA of oral carcinoma cells (HOC313, TSU, HSC2, HSC3, KOSC2, KOSC3, SCCKN, OSC19, Ca9.22, and Ho1u1 cells) and normal gingival fibroblasts (GF12 cells) derived from a Japanese population were amplified by polymerase chain reaction using primer sets spanning MALT1 exons, and nucleotide substitutions were analyzed by the single strand conformation polymorphism analysis. The substitutions were commonly observed in all cells, which express MALT1 at various levels. The substitutions at exons 1 and 9 were located at the 5' untranslated region and replaced (336)Asp to Asn, respectively, and others were positioned at the introns. Among the intronic substitutions, four were matched with the single nucleotide polymorphisms (SNPs) registered at the database. Since all cells were derived from a Japanese population, all substitutions detected are the SNPs. Absence of the carcinoma cell-specific mutation suggests that the inactivation of MALT1 expression but not the mutation promotes oral carcinoma progression.


Asunto(s)
Caspasas/genética , Encía/metabolismo , Neoplasias de la Boca/genética , Proteínas de Neoplasias/genética , Polimorfismo de Nucleótido Simple , Secuencia de Bases , Cartilla de ADN , Exones , Fibroblastos/metabolismo , Encía/patología , Humanos , Neoplasias de la Boca/patología , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas , Reacción en Cadena de la Polimerasa
16.
PLoS One ; 7(10): e47899, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23110125

RESUMEN

The cadherin switch from E-cadherin to N-cadherin is considered as a hallmark of the epithelial-mesenchymal transition and progression of carcinomas. Although it enhances aggressive behaviors of adenocarcinoma cells, the significance and role of cadherin switch in squamous cell carcinomas (SCCs) are largely controversial. In the present study, we immunohistochemically examined expression of E-cadherin and N-cadherin in oral SCCs (n = 63) and its implications for the disease progression. The E-cadherin-positive carcinoma cells were rapidly decreased at the invasive front. The percentage of carcinoma cells stained E-cadherin at the cell membrane was reduced in parallel with tumor dedifferentiation (P<0.01) and enhanced invasion (P<0.01). In contrast, N-cadherin-positive cells were very limited and did not correlate with the clinicopathological parameters. Mouse tongue tumors xenotransplantated oral SCC cell lines expressing both cadherins in vitro reproduced the reduction of E-cadherin-positive carcinoma cells at the invasive front and the negligible expression of N-cadherin. These results demonstrate that the reduction of E-cadherin-mediated carcinoma cell-cell adhesion at the invasive front, but not the cadherin switch, is an important determinant for oral SCC progression, and suggest that the environments surrounding carcinoma cells largely affect the cadherin expression.


Asunto(s)
Cadherinas/metabolismo , Carcinoma de Células Escamosas/fisiopatología , Regulación Neoplásica de la Expresión Génica/fisiología , Neoplasias de la Boca/fisiopatología , Adulto , Anciano , Anciano de 80 o más Años , Análisis de Varianza , Animales , Adhesión Celular/fisiología , Línea Celular Tumoral , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica/fisiopatología , Reacción en Cadena en Tiempo Real de la Polimerasa
17.
Odontology ; 100(2): 149-55, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21607592

RESUMEN

RAS overexpression and its active mutations are involved in malignant tumorigenesis. However, the mutation rates in oral carcinoma cells differ between populations. In the present study, genomic DNA of oral carcinoma cells (HOC313, TSU, HSC2, HSC3, KOSC2, KOSC3, SCCKN, OSC19, Ca9.22, and Ho1u1 cells) or normal gingival fibroblasts (GF12 cells) derived from a Japanese population were amplified by polymerase chain reaction using primer sets, spanning HRAS and KRAS exons. Nucleotide substitutions were analyzed by single strand conformation polymorphism. In contrast to no substitutions in KRAS, nine different substitutions were detected in HRAS. Of the nine, six substitutions were located at intron 1 (HSC2 and HSC3 cells) or intron 2 (HSC3, SCCKN and Ca9.22 cells), and one each of exon 1 (all cells), exon 2 (HOC313, TSU, HSC2 and HSC3 cells) and the 5' upstream region (all cells). Substitutions at exons 1 and 2 did not affect the amino acid sequence; the exon 1 substitution was positioned at the 5' untranslated region, which may be a single nucleotide polymorphism (SNP) sequence because all the cells were isolated from a Japanese population, and the mutations at exon 2 was a silent mutation. A substitution at the 5' upstream region was an SNP. These data demonstrate that SNPs and point mutations observed in HRAS do not change the amino acid sequence, and suggest that the mutations affecting the amino acid sequence may be a rare event in oral carcinomas of the Japanese population.


Asunto(s)
Carcinoma/genética , Genes ras/genética , Neoplasias de la Boca/genética , Mutación/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas/genética , Proteínas ras/genética , Regiones no Traducidas 5'/genética , Empalme Alternativo/genética , Secuencia de Aminoácidos , Línea Celular Tumoral , Células Cultivadas , Análisis Mutacional de ADN , Exones/genética , Fibroblastos/metabolismo , Variación Genética/genética , Encía/citología , Encía/metabolismo , Humanos , Intrones/genética , Japón , Mutación Puntual/genética , Polimorfismo de Nucleótido Simple/genética , Polimorfismo Conformacional Retorcido-Simple/genética , Eliminación de Secuencia/genética
18.
Am J Pathol ; 177(2): 598-607, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20595636

RESUMEN

Developmentally expressed genes are believed to play a central role in tissue repair after injury; however, in lung disease their role has not been established. This study demonstrates that SFRP1, an inhibitor of Wnt signaling normally expressed during lung embryogenesis, is induced in the lungs of emphysema patients and in two murine models of the disease. SFRP1 was found to be essential for alveolar formation as Sfrp1(-/-) mice exhibited aberrant Wnt signaling, mesenchymal proliferation, and impaired alveoli formation. In contrast, SFRP1 activated ERK and up-regulated MMP1 and MMP9 without altering TIMP1 production when expressed in human lung epithelial cells. These findings demonstrate that SFRP1 promotes normal alveolar formation in lung development, although its expression in the adult up-regulates proteins that can cause tissue destruction. Thus, SFRP1 induction during tissue injury is unlikely to contribute to the repair response but rather is a participatory factor in the pathogenesis of emphysema and tissue destruction.


Asunto(s)
Enfisema/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Pulmón , Proteínas de la Membrana/metabolismo , Organogénesis/fisiología , Animales , Línea Celular , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Pulmón/embriología , Pulmón/crecimiento & desarrollo , Pulmón/patología , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/fisiología , Humo , Nicotiana/efectos adversos , Proteínas Wnt/metabolismo , Proteína Wnt-5a , beta Catenina/metabolismo
19.
Cancer Res ; 69(18): 7216-23, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19738055

RESUMEN

Mucosa-associated lymphoid tissue 1 (MALT1), which is located in a genomic region that encodes unknown tumor suppressor gene(s), activates nuclear factor-kappaB in lymphocyte lineages. However, its expression and role in the pathology of malignant tumors of epithelial origin is not known. In the present study, we examined MALT1 expression and its implications for the pathology of oral carcinomas. Immunostaining localized MALT1 in the nucleus of normal oral epithelial cells, but the expression was absent in 45.0% of carcinomas (49 of 109 cases) especially at the invasive front. The loss of expression was correlated with tumor recurrence (P = 0.007) and poor patient survival (P < 0.001), and it was an independent prognostic determinant (P < 0.001). MALT1-negative carcinomas exhibited microsatellite instability at the MALT1 locus and a specific cytosine methylation positioned at -256 from the gene, and the expression was recovered by demethylation treatment. In contrast to lymphocyte lineages, carcinoma cells showed MALT1 located at the nucleus independent of its domain structures, and its loss of expression induced the epithelial-mesenchymal transition. These results show that MALT1 is expressed in the nucleus of oral epithelial cells and that its expression is epigenetically inactivated during tumor progression, suggesting that the detection of MALT1 expression is a useful predictive and prognostic determinant in the clinical management of oral carcinomas.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Caspasas/biosíntesis , Neoplasias de la Boca/enzimología , Proteínas de Neoplasias/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Caspasas/genética , Caspasas/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Metilación de ADN , Activación Enzimática , Femenino , Inestabilidad Genómica , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Regiones Promotoras Genéticas
20.
Gene ; 445(1-2): 17-25, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19559773

RESUMEN

Determining binding sites of transcription factors is important for understanding the transcriptional control of target genes. Although a transcription factor GATA3 plays a pivotal role in Th2 lymphocyte development, its physiological role is not clearly defined because the target genes remain largely unknown. In this study, we modified chromatin immunoprecipitation (ChIP), and isolated 121 GATA3 binding sites and 83 different annotated target genes. Re-ChIP analysis using anti-GATA3 and anti-RNA polymerase II mAbs and chromosome conformation capture assay demonstrate that GATA3-bound fragments interact with basal transcriptional units of target genes. GATA3 regulation of target genes under the control of binding fragments was confirmed by reporter assay and quantification of target gene mRNA expression in the presence of GATA inhibitor or short interfering RNA against GATA3. These data demonstrate that GATA3 binds to regulatory elements and controls target gene expression through physical interaction with core promoter regions.


Asunto(s)
Factor de Transcripción GATA3/metabolismo , Células Jurkat , Secuencia de Bases , Sitios de Unión/genética , Células Cultivadas , Inmunoprecipitación de Cromatina/métodos , Regulación Leucémica de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Unión Proteica , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...