Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Br J Cancer ; 108(4): 755-61, 2013 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-23322204

RESUMO

Matricellular proteins have been classified as a family of non-structural matrix proteins capable of modulating a variety of biological processes within the extracellular matrix (ECM). These proteins are expressed dynamically and their cellular functions are highly dependent upon cues from the local environment. Recent studies have shown an increasing appreciation of the key roles these ECM proteins play within the tumour microenvironment. Induced by either tumour cells or tumour stromal components, matricellular proteins initiate downstream signalling events that lead to proliferation, invasion, matrix remodelling and dissemination to pre-metastatic niches in other organs. In this review, we summarise and discuss the current knowledge of the diverse roles these proteins play within the microenvironment that influences tumour progression and potential for future therapies targeting the tumour microenvironment.


Assuntos
Proteínas da Matriz Extracelular/fisiologia , Neoplasias/patologia , Microambiente Tumoral , Matriz Extracelular/metabolismo , Humanos , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias/metabolismo , Transdução de Sinais
2.
Oncogene ; 26(16): 2353-64, 2007 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-17043653

RESUMO

Epidermal growth factor receptor (EGFR) overexpression and activation is critical in the initiation and progression of cancers, especially those of epithelial origin. EGFR activation is associated with the induction of divergent signal transduction pathways and a gamut of cellular processes; however, the cell-type and tissue-type specificity conferred by certain pathways remains to be elucidated. In the context of the esophageal epithelium, a prototype stratified squamous epithelium, EGFR overexpression is relevant in the earliest events of carcinogenesis as modeled in a three-dimensional organotypic culture system. We demonstrate that the phosphatidylinositol 3-kinase (PI3K)/AKT signaling pathway, and not the MEK/MAPK (mitogen-activated protein kinase) pathway, is preferentially activated in EGFR-mediated esophageal epithelial hyperplasia, a premalignant lesion. The hyperplasia was abolished with direct inhibition of PI3K and of AKT but not with inhibition of the MAPK pathway. With the introduction of an inducible AKT vector in both primary and immortalized esophageal epithelial cells, we find that AKT overexpression and activation is permissive for complete epithelial formation in organotypic culture, but imposes a growth constraint in cells grown in monolayer. In organotypic culture, AKT mediates changes related to cell shape and size with an expansion of the differentiated compartment.


Assuntos
Esôfago/citologia , Proteína Oncogênica v-akt/fisiologia , Diferenciação Celular , Divisão Celular , Células Cultivadas , Senescência Celular , Células Epiteliais/citologia , Receptores ErbB/fisiologia , Vetores Genéticos , Humanos , Imuno-Histoquímica , Técnicas de Cultura de Órgãos , Retroviridae
3.
J Clin Invest ; 108(5): 725-32, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11544278

RESUMO

The immortalization of human cells is a critical step in multistep carcinogenesis. Oral-esophageal carcinomas, a model system to investigate molecular mechanisms underlying squamous carcinogenesis, frequently involve cyclin D1 overexpression and inactivation of the p53 tumor suppressor. Therefore, our goal was to establish the functional role of cyclin D1 overexpression and p53 inactivation in the immortalization of primary human oral squamous epithelial cells (keratinocytes) as an important step toward malignant transformation. Cyclin D1 overexpression alone was found to induce extension of the replicative life span of normal oral keratinocytes, whereas the combination of cyclin D1 overexpression and p53 inactivation led to their immortalization. This study also demonstrates that immortalization of oral keratinocytes can be independent of telomerase activation, involving an alternative pathway of telomere maintenance (ALT).


Assuntos
Carcinoma de Células Escamosas/etiologia , Transformação Celular Neoplásica , Ciclina D1/genética , Genes p53 , Queratinócitos/fisiologia , Neoplasias Bucais/etiologia , Carcinoma de Células Escamosas/enzimologia , Ciclo Celular , Divisão Celular , Linhagem Celular , Ciclina D1/metabolismo , Humanos , Queratinócitos/citologia , Queratinócitos/enzimologia , Boca/citologia , Neoplasias Bucais/enzimologia , Mutação , Telomerase/fisiologia , Transdução Genética
4.
Oncogene ; 36(34): 4843-4858, 2017 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-28414310

RESUMO

High CD44 expression is associated with enhanced malignant potential in esophageal squamous cell carcinoma (ESCC), among the deadliest of all human carcinomas. Although alterations in autophagy and CD44 expression are associated with poor patient outcomes in various cancer types, the relationship between autophagy and cells with high CD44 expression remains incompletely understood. In transformed oesophageal keratinocytes, CD44Low-CD24High (CD44L) cells give rise to CD44High-CD24-/Low (CD44H) cells via epithelial-mesenchymal transition (EMT) in response to transforming growth factor (TGF)-ß. We couple patient samples and xenotransplantation studies with this tractable in vitro system of CD44L to CD44H cell conversion to investigate the functional role of autophagy in generation of cells with high CD44 expression. We report that high expression of the autophagy marker cleaved LC3 expression correlates with poor clinical outcome in ESCC. In ESCC xenograft tumours, pharmacological autophagy inhibition with chloroquine derivatives depletes cells with high CD44 expression while promoting oxidative stress. Autophagic flux impairment during EMT-mediated CD44L to CD44H cell conversion in vitro induces mitochondrial dysfunction, oxidative stress and cell death. During CD44H cell generation, transformed keratinocytes display evidence of mitophagy, including mitochondrial fragmentation, decreased mitochondrial content and mitochondrial translocation of Parkin, essential in mitophagy. RNA interference-mediated Parkin depletion attenuates CD44H cell generation. These data suggest that autophagy facilitates EMT-mediated CD44H generation via modulation of redox homeostasis and Parkin-dependent mitochondrial clearance. This is the first report to implicate mitophagy in regulation of tumour cells with high CD44 expression, representing a potential novel therapeutic avenue in cancers where EMT and CD44H cells have been implicated, including ESCC.


Assuntos
Autofagia/fisiologia , Receptores de Hialuronatos/metabolismo , Mitocôndrias/fisiologia , Estresse Oxidativo/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/fisiologia , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago , Humanos , Queratinócitos/metabolismo , Queratinócitos/fisiologia , Mitocôndrias/metabolismo , Oxirredução , Interferência de RNA/fisiologia , Fator de Crescimento Transformador beta/metabolismo
6.
Cancer Res ; 55(9): 1831-5, 1995 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-7537174

RESUMO

CD44 is the transmembrane adhesion molecule which binds hyaluronate. The gene encoding CD44 is found on chromosome 11p and comprises 20 exons. Differential splicing of the 10 extracellular juxtamembranous exons (v1-10) generates the major isoforms of CD44. The major CD44 isoform found on hematopoetic cells (CD44s) contains none of the variably expressed exons, while the major isoform expressed on epithelial cells [CD44(v8-10)] contains exons v8-10. Metastasis-specific isoforms of CD44 were first documented in a model of rat pancreatic adenocarcinoma [CD44(v4-7), CD44(v6-7)] and subsequently in other cancers. This study is the first characterization of CD44 isoforms in primary and metastatic human pancreatic adenocarcinomas. CD44 isoforms were analyzed in specimens of 15 primary and 6 metastatic pancreatic adenocarcinomas as well as in 6 specimens of control pancreata by two different methods. Radiolabeled reverse transcriptase-PCR coupled with 8% PAGE allowed analysis of the major isoforms of CD44, while Southern blot hybridization with [alpha-32P]dCTP-labeled probes permitted analysis for metastasis-specific CD44 isoforms containing CD44(v6) or CD44(v8-10). No differences in the expression of CD44(v8-10) and CD44s were found among the primary and metastatic pancreatic adenocarcinomas, and control specimens of pancreata. However, a novel CD44(v6) isoform was found in metastatic lesions and may represent the human homologue of the rat pancreatic adenocarcinoma metastasis-associated CD44 isoform.


Assuntos
Adenocarcinoma/química , Adenocarcinoma/secundário , Proteínas de Transporte/análise , Neoplasias Pancreáticas/química , Receptores de Superfície Celular/análise , Receptores de Retorno de Linfócitos/análise , Adenocarcinoma/genética , Adulto , Idoso , Sequência de Bases , Southern Blotting , Proteínas de Transporte/genética , Neoplasias do Sistema Digestório/secundário , Eletroforese em Gel de Poliacrilamida , Feminino , Humanos , Receptores de Hialuronatos , Isomerismo , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Pâncreas/química , Neoplasias Pancreáticas/genética , Reação em Cadeia da Polimerase , Receptores de Superfície Celular/genética , Receptores de Retorno de Linfócitos/genética , Transcrição Gênica
7.
Cancer Res ; 60(11): 2825-30, 2000 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-10850422

RESUMO

The stratified squamous epithelium is a model system in which to define molecular mechanisms underlying the switch from proliferation to differentiation. This can be achieved through the functional dissection of keratin gene promoters. Having previously established the importance of keratin 4 in maintaining the differentiated phenotype in corneal epithelial cells, we investigated the role of Sp1-mediated transactivation of the keratin 4 promoter given the role of Sp1 in differentiation and cell cycle progression. Sp1 transactivation of the keratin 4 promoter was diminished in cyclin D1-overexpressing cells, which may be mediated through a newly described direct interaction between Sp1 and cyclin D1 and opposed by a complex between Sp1 and pRB.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/genética , Ciclina D1/genética , Ciclina D1/metabolismo , Queratinas/genética , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo , Ativação Transcricional , Animais , Western Blotting , Proteínas de Ciclo Celular/genética , Linhagem Celular , Córnea/metabolismo , Epitélio/metabolismo , Citometria de Fluxo , Deleção de Genes , Regulação da Expressão Gênica , Queratinas/metabolismo , Testes de Precipitina , Regiões Promotoras Genéticas , Ligação Proteica , Coelhos , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Transfecção
8.
Cancer Res ; 60(11): 2836-9, 2000 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-10850424

RESUMO

Chromosomal allelic losses have varying frequency in breast cancer, with key regions including chromosomes 1, 3p, 7q, 9p, 16q, 17, and 22q. Recently, we have been able to map a new target region of allelic loss on chromosome 22q involved in colorectal cancer. The aim of the current investigation was to determine whether this target region may also be involved in human breast carcinogenesis. Thirty-six pairs of matched normal and tumor specimens from breast cancer patients, as well as eight breast cancer-derived cell lines, were genotyped using 17 microsatellite markers spanning chromosome 22q. Allelic deletion was found in 19 of 36 tumors (53%), and the pattern observed in those cases with partial losses was consistent with a region flanked by D22S1171 and D22S928. This interval overlaps that identified in colorectal cancer and comprises nearly 1.1 Mb. This study provides evidence of a common region of deletion on chromosome 22q13 involved in both breast and colorectal cancers and underscores the existence of putative tumor suppressor gene(s) at this location.


Assuntos
Neoplasias da Mama/genética , Cromossomos Humanos Par 22/genética , Neoplasias Colorretais/genética , Deleção de Genes , Alelos , Carcinoma Ductal de Mama/genética , Carcinoma Lobular/genética , Análise Mutacional de DNA , Feminino , Genótipo , Humanos , Repetições de Microssatélites , Modelos Genéticos , Mapeamento Físico do Cromossomo , Polimorfismo Genético , Células Tumorais Cultivadas
9.
Cancer Res ; 57(24): 5542-9, 1997 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-9407965

RESUMO

The cyclin D1 oncogene is critical in the progression of the cell cycle through the G1 phase. It is frequently overexpressed in squamous cell carcinomas originating from the head/neck and esophagus. Yet, the functional consequences of aberrant cyclin D1 overexpression are not entirely understood apart from increased cell proliferation. To address this question, we have developed a transgenic mouse model in which the EBV ED-L2 promoter targets cyclin D1 to the stratified squamous epithelium in a tissue-specific fashion to the tongue and esophagus, thereby resulting in a dysplastic phenotype. We now demonstrate that the dysplastic phenotype is associated with increased cell proliferation based on proliferating cell nuclear antigen overexpression and abnormalities in cyclin-dependent kinase 4, epidermal growth factor receptor, and p53. In aggregate, these studies suggest that alterations in certain oncogenes and tumor suppressor genes occur early during head/neck and esophageal carcinogenesis.


Assuntos
Ciclina D1/fisiologia , Receptores ErbB/fisiologia , Proteínas Proto-Oncogênicas , Proteína Supressora de Tumor p53/fisiologia , Animais , Ciclo Celular/fisiologia , Divisão Celular/fisiologia , Ciclina D1/biossíntese , Ciclina D1/genética , Quinase 4 Dependente de Ciclina , Quinases Ciclina-Dependentes/biossíntese , Epitélio/patologia , Receptores ErbB/biossíntese , Receptores ErbB/genética , Esôfago/metabolismo , Esôfago/patologia , Herpesvirus Humano 4/genética , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Fenótipo , Antígeno Nuclear de Célula em Proliferação/biossíntese , Língua/metabolismo , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/genética
10.
Cancer Res ; 61(17): 6467-73, 2001 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-11522642

RESUMO

The p53 gene family, comprising p53, p63, and p73, has overlapping and distinctive functional roles. These members share structural similarities allowing for dynamic interplay in the activation of genes that are important in development and key cellular functions, such as the induction of apoptosis. Whereas p53 is a classical tumor suppressor gene, p63 and p73 do not share this feature in cancer formation and progression. The compensation in the expression level of these members in a background that is deficient for one of them has not been examined previously. Given the importance of p63 in the development and differentiation of oral-esophageal stratified squamous epithelia and the absence of oral-esophageal tumors in p53-null mice, we postulated and describe herein that p63 expression is associated with the loss of p53 in a p53-deficient background. Both full-length and amino-truncated forms of p63 are expressed and increased in oral-esophageal epithelia of p53-null mice when compared with wild-type mice, and the induction of p21 may potentially be preserved through the increase of p63.


Assuntos
Esôfago/metabolismo , Proteínas de Membrana , Mucosa Bucal/metabolismo , Fosfoproteínas/biossíntese , Transativadores/biossíntese , Proteína Supressora de Tumor p53/deficiência , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Proteínas de Ligação a DNA , Epitélio/metabolismo , Epitélio/fisiologia , Esôfago/citologia , Esôfago/fisiologia , Genes Supressores de Tumor , Genes p53/fisiologia , Humanos , Imuno-Histoquímica , Camundongos , Mucosa Bucal/citologia , Mucosa Bucal/fisiologia , Fosfoproteínas/genética , Transativadores/genética , Fatores de Transcrição , Transfecção , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor
11.
Oncogene ; 35(41): 5337-5349, 2016 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-26923327

RESUMO

Esophageal cancer is a deadly disease, ranking sixth among all cancers in mortality. Despite incremental advances in diagnostics and therapeutics, esophageal cancer still carries a poor prognosis, and thus, there remains a need to elucidate the molecular mechanisms underlying this disease. There is accumulating evidence that a comprehensive understanding of the molecular composition of esophageal cancer requires attention to not only tumor cells but also the tumor microenvironment (TME), which contains diverse cell populations, signaling factors and structural molecules that interact with tumor cells and support all stages of tumorigenesis. In esophageal cancer, environmental exposures can trigger chronic inflammation, which leads to constitutive activation of pro-inflammatory signaling pathways that promote survival and proliferation. Antitumor immunity is attenuated by cell populations such as myeloid-derived suppressor cells and regulatory T cells, as well as immune checkpoints like programmed death-1. Other immune cells such as tumor-associated macrophages can have other pro-tumorigenic functions, including the induction of angiogenesis and tumor cell invasion. Cancer-associated fibroblasts secrete growth factors and alter the extracellular matrix to create a tumor niche and enhance tumor cell migration and metastasis. Further study of how these TME components relate to the different stages of tumor progression in each esophageal cancer subtype will lead to development of novel and specific TME-targeting therapeutic strategies, which offer considerable potential especially in the setting of combination therapy.


Assuntos
Neoplasias Esofágicas/genética , Inflamação/genética , Neovascularização Patológica/genética , Microambiente Tumoral/genética , Apoptose/genética , Apoptose/imunologia , Pontos de Checagem do Ciclo Celular/genética , Pontos de Checagem do Ciclo Celular/imunologia , Neoplasias Esofágicas/patologia , Humanos , Inflamação/imunologia , Inflamação/patologia , Invasividade Neoplásica/imunologia , Neovascularização Patológica/patologia , Linfócitos T Reguladores/imunologia
12.
Oncogene ; 35(29): 3880-6, 2016 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-26592448

RESUMO

Epidermal growth factor receptor (EGFR) signaling has a critical role in oncogenic Kras-driven pancreatic carcinogenesis. However, the downstream targets of this signaling network are largely unknown. We developed a novel model system utilizing murine primary pancreatic ductal epithelial cells (PDECs), genetically engineered to allow time-specific expression of oncogenic Kras(G12D) from the endogenous promoter. We show that primary PDECs are susceptible to Kras(G12D)-driven transformation and form pancreatic ductal adenocarcinomas in vivo after Cdkn2a inactivation. In addition, we demonstrate that activation of Kras(G12D) induces an EGFR signaling loop to drive proliferation. Interestingly, pharmacological inhibition of EGFR fails to decrease Kras(G12D)-activated ERK or PI3K signaling. Instead our data provide novel evidence that EGFR signaling is needed to activate the oncogenic and pro-proliferative transcription factor c-MYC. EGFR and c-MYC have been shown to be essential for pancreatic carcinogenesis. Importantly, our data link both pathways and thereby explain the crucial role of EGFR for Kras(G12D)-driven carcinogenesis in the pancreas.


Assuntos
Células Epiteliais/metabolismo , Receptores ErbB/metabolismo , Ductos Pancreáticos/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Animais , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Células Cultivadas , Células Epiteliais/transplante , Receptores ErbB/genética , Perfilação da Expressão Gênica/métodos , Immunoblotting , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Camundongos Transgênicos , Mutação , Ductos Pancreáticos/citologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética
13.
Oncogene ; 13(8): 1809-13, 1996 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-8895528

RESUMO

The Epstein-Barr virus (EBV), a member of the herpesviruses, is a double stranded 170 kilobase DNA virus important in many human benign and malignant conditions. It has been implicated in the pathogenesis of proliferative diseases of lymphocytes and tumors of epithelial derivation. The etiology and pathogenesis of esophageal squamous cell carcinoma (ESCC) is thought to involve a combination of genetic and environmental events which lead to epithelial cell transformation. The aim of this study was to determine whether an association exists between EBV and ESCC. DNA was extracted from 16 human ESCC cell lines and microdissected tumor specimens from 60 patients. The polymerase chain reaction was used to amplify a 400 base pair fragment corresponding to the BamH1W fragment repeat sequence of EBV. Southern blotting, utilizing an oligonucleotide probe specific for the BamH1W sequence, was used to confirm positive results and increase sensitivity of detection. 5/60 tumor samples and 1/16 ESCC cell lines were positive for the EBV sequence. Positive tumor samples were estimated to contain one copy of EBV per 20 cellular genomes. Given the role of EBV in other tumors of epithelial derivation, it is possible that EBV may contribute to the molecular pathogenesis of ESCC.


Assuntos
Carcinoma de Células Escamosas/virologia , DNA Viral/metabolismo , Neoplasias Esofágicas/virologia , Herpesvirus Humano 4/isolamento & purificação , Carcinoma de Células Escamosas/genética , Neoplasias Esofágicas/genética , Herpesvirus Humano 4/genética , Humanos , Reação em Cadeia da Polimerase , Células Tumorais Cultivadas
14.
Oncogene ; 12(6): 1239-45, 1996 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-8649826

RESUMO

We and others have recently shown that p16 can potently and specifically inhibit progression through the G1 phase of the replicative cycle in cells that express the retinoblastoma protein (pRB). However, none of these studies examined cell types in which p16 has been firmly implicated in tumorigenesis. We predicted that such cells would show sensitivity to p16 inhibition, perhaps conferred by proteins in addition to or other than pRB. Intragenic, inactivating mutations of p16 have been found at significant frequency in primary tumors derived from squamous epithelial cells of the esophagus (ESCC). We therefore examined p16 function in ESCC lines and in primary squamous epithelial cells cultured from mouse skin. We find that seven of eight ESCC lines tested are inhibited by p16 and fail to express the protein endogenously. The lone p16-resistant line expresses endogenous p16 but lacks functional pRB. Primary squamous epithelial cells are also inhibited by p16. These data suggest that squamous epithelial cells are generally sensitive to inhibition by a regulatory pathway that involves p16 and pRB, and that, by the time of establishment in culture, there is nearly universal inactivation of this pathway in ESCCs.


Assuntos
Carcinoma de Células Escamosas/patologia , Proteínas de Transporte/fisiologia , Transformação Celular Neoplásica/patologia , Queratinócitos/fisiologia , Neoplasias Cutâneas/patologia , Células 3T3/patologia , Células 3T3/fisiologia , Animais , Proteínas de Transporte/genética , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina , Células Epiteliais , Epitélio/fisiologia , Éxons , Humanos , Queratinócitos/citologia , Camundongos , Camundongos Endogâmicos SENCAR , Microinjeções , Proteína do Retinoblastoma/fisiologia , Sensibilidade e Especificidade , Pele/citologia , Fenômenos Fisiológicos da Pele
15.
Oncogene ; 19(15): 1941-9, 2000 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-10773884

RESUMO

The ets family of transcription factors comprises many members which contribute to diverse cellular functions that vary depending upon the cell- and tissue-type context. Recently, different groups have identified a novel member of the ets family that is epithelial-specific. Variably called ESE-1, ERT, jen, ESX, this gene is designated currently as ELF3. In order to understand transcriptional regulatory mechanisms mediated by ELF3, we investigated its effect on the human keratin 4 gene promoter based upon the role of keratin 4 in early differentiation of the esophageal squamous epithelium. Interestingly, ELF3 suppressed basal keratin 4 promoter activity in both esophageal and cervical epithelial cancer cell lines, a novel result, while simultaneously activating the late-differentiation linked SPRR2A promoter. Furthermore, serial deletion constructs of the keratin 4 promoter continued to be suppressed by ELF3, a phenomenon that was only partially rescued by ELF3 ets domain mutants, but completely abrogated by deletion of the ELF3 pointed domain. These results suggest that ELF3 may have dual functions in the transcriptional regulation of genes involved in squamous epithelial differentiation. One of these functions may not be exclusively mediated through DNA binding in the context of transcriptional suppression of the keratin 4 promoter.


Assuntos
Proteínas de Ligação a DNA , Células Epiteliais/fisiologia , Queratinas/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Fatores de Transcrição/fisiologia , Sequência de Aminoácidos , Carcinoma de Células Escamosas/metabolismo , Diferenciação Celular , Neoplasias Esofágicas/metabolismo , Células HeLa , Humanos , Queratinas/genética , Dados de Sequência Molecular , Mutação , Proteínas Proto-Oncogênicas c-ets , Células Tumorais Cultivadas
16.
Oncogene ; 10(3): 619-22, 1995 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-7845688

RESUMO

MTS-1 is a candidate tumor suppressor gene on chromosome 9p21-22, a region frequently observed to have loss of heterozygosity in esophagus squamous cell carcinomas and pancreatic ductal adenocarcinomas. In order to determine whether MTS-1 sequences are deleted or mutated in cell lines derived from these cancers, we performed PCR amplification of MTS-1 exons 1 and 2. In this fashion, we found that 67% of esophagus squamous cancer cell lines have deletions of both exons 1 and 2, and 50% of pancreatic cancer cell lines have similar deletions. Furthermore, an additional 30% of pancreatic cancer cell lines harbored point mutations or microdeletions based on DNA sequencing. MTS-1 encodes p16, an inhibitor of cyclin-dependent kinase 4 (cdk4) which complexes with cyclin D1. Our data suggest that MTS-1 deletions and mutations may play an important role in the molecular pathogenesis of esophagus squamous cell and pancreatic cancers.


Assuntos
Adenocarcinoma/genética , Carcinoma de Células Escamosas/genética , Proteínas de Transporte/genética , Neoplasias Esofágicas/genética , Genes Supressores de Tumor , Neoplasias Pancreáticas/genética , Sequência de Bases , Inibidor p16 de Quinase Dependente de Ciclina , Éxons/genética , Deleção de Genes , Humanos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Células Tumorais Cultivadas
17.
Oncogene ; 14(10): 1185-90, 1997 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-9121767

RESUMO

Cyclin D1 in cooperation with its major catalytic partners, cyclin-dependent kinases cdk4 and cdk6, facilitates progression through the G1 phase of the eukaryotic cell cycle, in part through phosphorylation of the retinoblastoma protein. Cyclin D1's oncogenic properties have been suggested by its cooperation with ras or adenovirus E1a to transform cultured cells, as well its overexpression in transgenic mice that leads to breast cancer. Activated by a number of different mechanisms in human cancers, the cyclin D1 gene is frequently amplified in squamous epithelial cancers derived from the head/neck and esophageal regions. In order to study the functional consequences of cyclin D1 overexpression in these squamous epithelial specific sites, we have linked the Epstein-Barr virus ED-L2 promoter to the human cyclin D1 cDNA and utilized this transgene to generate founder lines. This transgene is transcribed specifically in the tongue, esophagus and forestomach, all sharing a stratified squamous epithelium. The transgene protein product localizes to the basal and suprabasal compartments of these squamous epithelial tissues, and mice from different lines develop dysplasia, a prominent precursor to carcinoma, by 16 months of age in contrast to age-matched wild-type mice. This transgenic model is useful in demonstrating cyclin D1 may be a tumor initiating event in aero-upper digestive squamous epithelial tissues.


Assuntos
Transformação Celular Neoplásica/genética , Ciclinas/genética , Neoplasias Esofágicas/genética , Herpesvirus Humano 4/genética , Proteínas Oncogênicas/genética , Lesões Pré-Cancerosas/genética , Regiões Promotoras Genéticas/fisiologia , Neoplasias Gástricas/genética , Neoplasias da Língua/genética , Animais , Southern Blotting , Ciclina D1 , Ciclinas/metabolismo , DNA Complementar/genética , Neoplasias Esofágicas/patologia , Esôfago/metabolismo , Esôfago/patologia , Mucosa Gástrica/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Proteínas Oncogênicas/metabolismo , Fenótipo , Lesões Pré-Cancerosas/patologia , Regiões Promotoras Genéticas/genética , Estômago/patologia , Neoplasias Gástricas/patologia , Língua/metabolismo , Língua/patologia , Neoplasias da Língua/patologia
18.
Oncogene ; 18(1): 59-66, 1999 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-9926920

RESUMO

We previously described the oral-esophageal tissue-specific expression of cyclin D1 with the Epstein-Barr virus ED-L2 promoter in transgenic mice, and resulting dysplasia. Given the evidence for an interplay between environmental and genetic factors in esophageal squamous carcinogenesis, the aim of this study was to determine the potential cooperation of the nitrosamine compound N-nitrosomethylbenzylamine (NMBA), an esophageal specific carcinogen, in the cyclin D1 transgenic mice. NMBA was first demonstrated to induce dysplasia in two strains of inbred mice, C57BL/6 and FVB/N. Subcutaneous NMBA was then administrated to wild type and transgenic mice beginning at 4 weeks of age. Mice were monitored for the duration of the study for general appearance, activity and weight, and were euthanized at 12 and 15 months. Histopathologic analysis revealed increased severity of dysplasia in cyclin D1 mice treated with NMBA compared with treated age-matched wild-type mice and untreated mice. There was also increased proliferating cell nuclear antigen (PCNA) expression in the esophagi of NMBA treated cyclin D1 mice. Taken together, these findings suggest that a genetic alteration, specifically cyclin D1 overexpression and a chemical carinogen, NMBA, may cooperate to increase the severity of esophageal squamous dysplasia, a prominent precursor to carcinoma.


Assuntos
Carcinógenos/farmacologia , Ciclina D1/farmacologia , Dimetilnitrosamina/análogos & derivados , Neoplasias Esofágicas/etiologia , Neoplasias de Células Escamosas/etiologia , Animais , Divisão Celular , Ciclina D1/biossíntese , Ciclina D1/genética , Dimetilnitrosamina/farmacologia , Epitélio/patologia , Neoplasias Esofágicas/patologia , Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias de Células Escamosas/patologia
19.
J Clin Oncol ; 22(23): 4730-6, 2004 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-15570073

RESUMO

PURPOSE: In March 2001, the National Colorectal Cancer Research Alliance (NCCRA) and OncoLink (http://www.oncolink.org) established a database to facilitate patient enrollment onto clinical trials. This study describes the population registering with the database and identifies discrepancies between individuals registering through the Internet and those registering through a telephone call center. METHODS: Participants registered with the NCCRA/OncoLink database through the Internet or a telephone call center. All participants entering the database completed a questionnaire regarding basic demographics, colon cancer risk factors, and indicated how they became aware of the database. Comparisons were made between individuals registering through the Internet and those registering through the telephone call center. RESULTS: A total of 2,162 participants registered during the first 16 months of the database. Most patients registered through the Internet rather than the telephone call center (88% v 12%; P < .001). More females than males registered (73% v 27%; P < .001). The majority (89%) were white. Participants registering through the Internet were younger than those registering through the call center (mean, 48.8 v 55.0 years; P < .001). There was no difference between the two groups with regard to sex or ethnicity. CONCLUSION: The Internet has the potential to increase the likelihood that interested individuals find appropriate clinical trials. Some of the discrepancies that are known to exist for access to the Internet were also seen for those registering with the database through the Internet. Despite these differences, the potential to increase clinical trial enrollment with this type of Internet-based database is high.


Assuntos
Ensaios Clínicos como Assunto , Neoplasias Colorretais/terapia , Internet , Sistema de Registros , Telefone , Adolescente , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Neoplasias Colorretais/diagnóstico , Bases de Dados Factuais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Seleção de Pacientes , Probabilidade , Pesquisa , Sensibilidade e Especificidade , Fatores Sexuais , Sociedades Médicas , Estados Unidos
20.
Arch Intern Med ; 148(7): 1583-4, 1988 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-3260095

RESUMO

A patient with systemic lupus erythematosus developed gluten-sensitive enteropathy, or celiac sprue. The patient's histocompatibility antigens included HLA-B8 and HLA-DR3, previously found to have a high frequency in gluten-sensitive enteropathy and possibility increased as well in systemic lupus erythematosus. Such histocompatibility antigens are common to a variety of autoimmune disorders. An immune basis for the association is discussed herein.


Assuntos
Doença Celíaca/complicações , Lúpus Eritematoso Sistêmico/complicações , Doença Celíaca/genética , Doença Celíaca/imunologia , Feminino , Antígenos HLA/genética , Antígeno HLA-B8 , Antígenos HLA-DR/genética , Antígeno HLA-DR3 , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Pessoa de Meia-Idade
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa