Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncotarget ; 3(8): 833-42, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22909995

RESUMO

Multiple clinical trials are ongoing to evaluate the potential antitumor activity of human TNF variants, Fas ligand (FasL), TNF-related apoptosis inducing ligand (TRAIL) and its agonistic antibodies. These drug products act through the death receptors (DRs) TNF receptor 1 (TNFR1), Fas/CD95, DR4 (TRAIL-R1) and/or DR5 (TRAIL-R2), respectively. Therefore, characterization of the level and localization of DR expression in cancer cells is important for DR-targeted therapy. In this study, we examined the subcellular distribution of the four DRs in a panel of 10 human breast cancer cell lines by western blots and flow cytometry and 50 human breast tumors by immunohistochemistry. Despite their total protein expressions, the DRs were found to be absent on the surface of some cell lines. Consistent with this result, all four DRs were found to be mostly expressed in the cytoplasm and/or the nucleus of primary breast tumors (n=50). We further determined the growth inhibition activity (GI50) of the death ligands, recombinant human TNFα, FasL and TRAIL, and found a correlation with the subcellular localization of the corresponding DRs. These results demonstrate an aberrant expression of the death receptors in breast cancer cells, and suggest that the lack of surface DRs appears to be predictive of tumor resistance to DR-targeted therapies.


Assuntos
Neoplasias da Mama/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptor fas/metabolismo , Apoptose , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteína Ligante Fas/metabolismo , Proteína Ligante Fas/farmacologia , Feminino , Humanos , Ligantes , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/antagonistas & inibidores , Receptores Tipo I de Fatores de Necrose Tumoral/antagonistas & inibidores , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Receptor fas/antagonistas & inibidores
2.
Endocrinology ; 151(8): 4024-30, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20555035

RESUMO

The pancreatic alpha- and beta-cells are critical components in regulating blood glucose homeostasis via secretion of glucagon and insulin, respectively. Both cell types are typically localized in the islets of Langerhans. However, little is known about the roles of paracrine interactions that contribute to their physiological functions. The lack of suitable cell lines to study alpha- and beta-cells interactions have led us to develop an alpha-cell-specific Cre-expressing transgenic line utilizing a glucagon promoter sequence, the Glu-Cre transgenic mouse. Here, we demonstrate that the Glu-Cre could specifically and efficiently excise floxed target genes in adult islet alpha-cells. We further showed that deletion of the tumor suppressor gene, multiple endocrine neoplasia type 1 (Men1), in alpha-cells led to tumorigenesis. However, to our surprise, the lack of Men1 in alpha-cells did not result in glucagonomas but rather beta-cell insulinomas. Because deletion of the Men1 alleles was only present in alpha-cells, our data suggested that cross communication between alpha- and beta-cells contributes to tumorigenesis in the absence of Men1. Together, we believed that the new model systems described here will allow future studies to decipher cellular interactions between islet alpha- and beta-cells in a physiological context.


Assuntos
Células Secretoras de Glucagon/metabolismo , Insulinoma/genética , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogênicas/genética , Animais , Células Cultivadas , Deleção de Genes , Técnicas de Silenciamento de Genes , Glucagon/genética , Glucagon/metabolismo , Insulinoma/metabolismo , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos/genética , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo
3.
Clin Cancer Res ; 16(1): 249-59, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20028755

RESUMO

PURPOSE: The capability of microarray platform to interrogate thousands of genes has led to the development of molecular diagnostic tools for cancer patients. Although large-scale comparative studies on clinical samples are often limited by the access of human tissues, expression profiling databases of various human cancer types are publicly available for researchers. Given that mouse models have been instrumental to our current understanding of cancer progression, we aimed to test the hypothesis that novel gene signatures possessing predictability in clinical outcome can be derived by coupling genomic analyses in mouse models of cancer with publicly available human cancer data sets. EXPERIMENTAL DESIGN: We established a complex series of syngeneic metastatic animal models using a murine breast cancer cell line. Tumor RNA was hybridized on Affymetrix MouseGenome-430A2.0 GeneChips. With the use of Venn logic, gene signatures that represent metastatic competency were derived and tested against publicly available human breast and lung cancer data sets. RESULTS: Survival analyses showed that the spontaneous metastasis gene signature was significantly associated with metastasis-free and overall survival (P < 0.0005). Consequently, the six-gene model was determined and showed statistical predictability in predicting survival in breast cancer patients. In addition, the model was able to stratify poor from good prognosis for lung cancer patients in most data sets analyzed. CONCLUSIONS: Together, our data support that novel gene signature derived from mouse models of cancer can be used for predicting human cancer outcome. Our approaches set precedence that similar strategies may be used to decipher novel gene signatures for clinical utility.


Assuntos
Modelos Animais de Doenças , Perfilação da Expressão Gênica , Neoplasias Mamárias Experimentais/genética , Animais , Biomarcadores Tumorais , Neoplasias da Mama/genética , Bases de Dados Factuais , Humanos , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/mortalidade , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sequência com Séries de Oligonucleotídeos , Prognóstico , Análise de Sobrevida
4.
PLoS One ; 4(4): e4897, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19340311

RESUMO

The von Hippel-Lindau (VHL) syndrome is a pleomorphic familial disease characterized by the development of highly vascularized tumors, such as hemangioblastomas of the central nervous system, pheochromocytomas, renal cell carcinomas, cysts and neuroendocrine tumors of the pancreas. Up to 75% of VHL patients are affected by VHL-associated pancreatic lesions; however, very few reports in the published literature have described the cellular origins and biological roles of VHL in the pancreas. Since homozygous loss of Vhl in mice resulted in embryonic lethality, this study aimed to characterize the functional significance of VHL in the pancreas by conditionally inactivating Vhl utilizing the Cre/LoxP system. Specifically, Vhl was inactivated in different pancreatic cell populations distinguished by their roles during embryonic organ development and their endocrine lineage commitment. With Cre recombinase expression directed by a glucagon promoter in alpha-cells or an insulin promoter in beta-cells, we showed that deletion of Vhl is dispensable for normal functions of the endocrine pancreas. In addition, deficiency of VHL protein (pVHL) in terminally differentiated alpha-cells or beta-cells is insufficient to induce pancreatic neuroendocrine tumorigenesis. Most significantly, we presented the first mouse model of VHL-associated pancreatic disease in mice lacking pVHL utilizing Pdx1-Cre transgenic mice to inactivate Vhl in pancreatic progenitor cells. The highly vascularized microcystic adenomas and hyperplastic islets that developed in Pdx1-Cre;Vhl f/f homozygous mice exhibited clinical features similar to VHL patients. Establishment of three different, cell-specific Vhl knockouts in the pancreas have allowed us to provide evidence suggesting that VHL is functionally important for postnatal ductal and exocrine pancreas, and that VHL-associated pancreatic lesions are likely to originate from progenitor cells, not mature endocrine cells. The novel model systems reported here will provide the basis for further functional and genetic studies to define molecular mechanisms involved in VHL-associated pancreatic diseases.


Assuntos
Inativação Gênica , Pâncreas/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/fisiologia , Animais , Linhagem da Célula , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Camundongos , Pâncreas/citologia , Regulação para Cima , Proteína Supressora de Tumor Von Hippel-Lindau/genética
5.
Cancer Res ; 69(5): 1858-66, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19208834

RESUMO

Multiple endocrine neoplasia type 1 (MEN1) is an autosomal syndrome caused by mutations in the MEN1 tumor suppressor gene. Whereas the protein product of MEN1, menin, is ubiquitously expressed, somatic loss of the remaining wild-type MEN1 allele results in tumors primarily in parathyroid, pituitary, and endocrine pancreas. To understand the endocrine specificity of the MEN1 syndrome, we evaluated biallelic loss of Men1 by inactivating Men1 in pancreatic progenitor cells using the Cre-lox system. Men1 deletion in progenitor cells that differentiate into exocrine and endocrine pancreas did not affect normal pancreas morphogenesis and development. However, mice having homozygous inactivation of the Men1 in pancreas developed endocrine tumors with no exocrine tumor manifestation, recapitulating phenotypes seen in the MEN1 patients. In the absence of menin, the endocrine pancreas showed increase in cell proliferation, vascularity, and abnormal vascular structures; such changes were lacking in exocrine pancreas. Further analysis revealed that these endocrine manifestations were associated with up-regulation in vascular endothelial growth factor expression in both human and mouse MEN1 pancreatic endocrine tumors. Together, these data suggest the presence of cell-specific factors for menin and a permissive endocrine environment for MEN1 tumorigenesis in endocrine pancreas. Based on our analysis, we propose that menin's ability to maintain cellular and microenvironment integrity might explain the endocrine- restrictive nature of the MEN1 syndrome.


Assuntos
Proteínas de Homeodomínio/fisiologia , Neoplasia Endócrina Múltipla Tipo 1/etiologia , Tumores Neuroendócrinos/etiologia , Neoplasias Pancreáticas/etiologia , Proteínas Proto-Oncogênicas/fisiologia , Transativadores/fisiologia , Animais , Proliferação de Células , Humanos , Ilhotas Pancreáticas/irrigação sanguínea , Camundongos , Fator A de Crescimento do Endotélio Vascular/fisiologia
6.
Endocr Relat Cancer ; 15(1): 267-75, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18310293

RESUMO

Multiple endocrine neoplasia type 1 (MEN1) is an autosomal dominant syndrome caused by mutations in the MEN1 tumor suppressor gene. Loss of the functional second copy of the MEN1 gene causes individuals to develop multiple endocrine tumors, primarily affecting the parathyroid, pituitary, and pancreas. While it is clear that the protein encoded by MEN1, menin, suppresses endocrine tumors, its biochemical functions and direct downstream targets remain unclear. Recent studies have suggested that menin may act as a scaffold protein to coordinate gene transcription, and that menin is an oncogenic cofactor for homeobox (HOX) gene expression in hematopoietic cancer. The role of HOX genes in adult cell differentiation is still obscure, but growing evidence suggests that they may play important roles in the development of cancer. Therefore, we hypothesized that specific HOX genes were regulated by menin in parathyroid tumor development. Utilizing quantitative TaqMan RT-PCR, we compared expression profiles of the 39 HOX genes in human familial MEN1 (fMEN1) parathyroid tumors and sporadic parathyroid adenomas with normal samples. We identified a large set of 23 HOX genes whose deregulation is specific for fMEN1 parathyroid tumors, and only 5 HOX genes whose misexpression are specific for sporadic parathyroid tumor development. These findings provide the first evidence that loss of the MEN1 tumor suppressor gene is associated with deregulation of specific HOX gene expression in the development of familial human parathyroid tumors. Our results strongly reinforce the idea that abnormal expression of developmental HOX genes can be critical in human cancer progression.


Assuntos
Genes Homeobox/genética , Neoplasia Endócrina Múltipla Tipo 1/genética , Mutação/genética , Neoplasias das Paratireoides/metabolismo , Neoplasias das Paratireoides/patologia , Proteínas Proto-Oncogênicas/genética , Adolescente , Adulto , Idoso , Feminino , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasia Endócrina Múltipla Tipo 1/metabolismo , Neoplasia Endócrina Múltipla Tipo 1/patologia , Neoplasias das Paratireoides/genética , Proteínas Proto-Oncogênicas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Dev Dyn ; 236(6): 1683-93, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17440987

RESUMO

Recessive inactivating mutations in human matrix metalloproteinase 2 (MMP2, gelatinase A) are associated with syndromes that include abnormal facial appearance, short stature, and severe bone loss. Mmp2(-/-) mice have only mild aspects of these abnormalities, suggesting that MMP2 function is redundant during skeletal development in the mouse. Here, we report that Mmp2(-/-) mice with additional mutations that render type I collagen resistant to collagenase-mediated cleavage to TC(A) and TC(B) fragments (Col1a1(r/r) mice) have severe developmental defects resembling those observed in MMP2-null humans. Composite Mmp2(-/-);Col1a1(r/r) mice were born in expected Mendelian ratios but were half the size of wild-type, Mmp2(-/-), and Col1a1(r/r) mice and failed to thrive. Furthermore, composite Mmp2(-/-);Col1a1(r/r) animals had very abnormal craniofacial features with shorter snouts, bulging skulls, incompletely developed calvarial bones and unclosed cranial sutures. In addition, trabecular bone mass was reduced concomitant with increased numbers of bone-resorbing osteoclasts and osteopenia. In vitro, MMP2 had a unique ability among the collagenolytic MMPs to degrade mutant collagen, offering a possible explanation for the genetic interaction between Mmp2 and Col1a1(r). Thus, because mutations in the type I collagen gene alter the phenotype of mice with null mutations in Mmp2, we conclude that type I collagen is an important modifier gene for Mmp2. Developmental Dynamics 236:1683-1693, 2007. (c) 2007 Wiley-Liss, Inc.


Assuntos
Osso e Ossos/metabolismo , Colágeno Tipo I/metabolismo , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Animais , Densidade Óssea , Doenças Ósseas Metabólicas/genética , Doenças Ósseas Metabólicas/metabolismo , Doenças Ósseas Metabólicas/patologia , Anormalidades Craniofaciais , Edema/genética , Edema/metabolismo , Edema/patologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Articulações/anormalidades , Articulações/metabolismo , Metaloproteinase 2 da Matriz/deficiência , Camundongos , Camundongos Transgênicos
8.
Curr Protoc Cytom ; Chapter 12: Unit 12.5, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-18770816

RESUMO

Two vascular systems, cardiovascular and lymphatic, maintain appropriate interstitial and intravascular fluid volume in the body. Each is endowed with innate physiologic response capabilities activated upon tissue or organ "damage." Chronic activation following pathologic assault, however, can contribute to pathogenesis. Three-dimensional visualization of vasculature in whole tissues using confocal microscopy is a valuable tool for examining cellular and architectural changes accompanying altered vascular function. The relative affinities of plant lectins for carbohydrate moieties present on luminal surfaces of endothelial cells can be used to characterize endothelium in distinctive physiologic and pathologic states. Perivascular cells that wrap around blood endothelial cells can be visualized using antibodies immunoreactive with alpha-smooth muscle actin. Similarly, lymphatic endothelial cells can be detected by antibodies immunoreactive to the hyaluronan receptor LYVE-1. Together, these approaches allow functional and morphological analysis of blood vasculature distinct from endothelial cells within the lymphatic vascular network and surrounding support cells.


Assuntos
Vasos Sanguíneos/citologia , Células Endoteliais/citologia , Endotélio Vascular/citologia , Vasos Linfáticos/citologia , Microscopia Confocal/métodos , Animais , Vasos Sanguíneos/patologia , Células Endoteliais/patologia , Endotélio Vascular/patologia , Humanos , Processamento de Imagem Assistida por Computador , Vasos Linfáticos/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...