Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Nature ; 543(7643): 122-125, 2017 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-28178237

RESUMO

Human cells have twenty-three pairs of chromosomes. In cancer, however, genes can be amplified in chromosomes or in circular extrachromosomal DNA (ecDNA), although the frequency and functional importance of ecDNA are not understood. We performed whole-genome sequencing, structural modelling and cytogenetic analyses of 17 different cancer types, including analysis of the structure and function of chromosomes during metaphase of 2,572 dividing cells, and developed a software package called ECdetect to conduct unbiased, integrated ecDNA detection and analysis. Here we show that ecDNA was found in nearly half of human cancers; its frequency varied by tumour type, but it was almost never found in normal cells. Driver oncogenes were amplified most commonly in ecDNA, thereby increasing transcript level. Mathematical modelling predicted that ecDNA amplification would increase oncogene copy number and intratumoural heterogeneity more effectively than chromosomal amplification. We validated these predictions by quantitative analyses of cancer samples. The results presented here suggest that ecDNA contributes to accelerated evolution in cancer.


Assuntos
Variações do Número de Cópias de DNA/genética , Evolução Molecular , Amplificação de Genes/genética , Heterogeneidade Genética , Modelos Genéticos , Neoplasias/genética , Oncogenes/genética , Cromossomos Humanos/genética , Análise Citogenética , Análise Mutacional de DNA , Genoma Humano/genética , Humanos , Metáfase/genética , Neoplasias/classificação , RNA Mensageiro/análise , RNA Neoplásico/genética , Reprodutibilidade dos Testes , Software
2.
Nat Immunol ; 10(5): 504-13, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19363483

RESUMO

Foxo transcription factors regulate cell cycle progression, cell survival and DNA-repair pathways. Here we demonstrate that deficiency in Foxo3 resulted in greater expansion of T cell populations after viral infection. This exaggerated expansion was not T cell intrinsic. Instead, it was caused by the enhanced capacity of Foxo3-deficient dendritic cells to sustain T cell viability by producing more interleukin 6. Stimulation of dendritic cells mediated by the coinhibitory molecule CTLA-4 induced nuclear localization of Foxo3, which in turn inhibited the production of interleukin 6 and tumor necrosis factor. Thus, Foxo3 acts to constrain the production of key inflammatory cytokines by dendritic cells and to control T cell survival.


Assuntos
Células Dendríticas/imunologia , Fatores de Transcrição Forkhead/imunologia , Ativação Linfocitária/imunologia , Linfócitos T/imunologia , Animais , Apresentação de Antígeno/imunologia , Antígenos CD/imunologia , Antígenos CD/metabolismo , Infecções por Arenaviridae/imunologia , Western Blotting , Antígeno CTLA-4 , Células Dendríticas/metabolismo , Citometria de Fluxo , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/metabolismo , Interleucina-6/imunologia , Interleucina-6/metabolismo , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Congênicos , Camundongos Transgênicos , Transporte Proteico/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
3.
Diabetes ; 58(6): 1275-82, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19289458

RESUMO

OBJECTIVE: Forkhead box O (FoxO) transcription factors represent evolutionarily conserved targets of insulin signaling, regulating metabolism and cellular differentiation in response to changes in nutrient availability. Although the FoxO1 isoform is known to play a key role in adipogenesis, its physiological role in differentiated adipose tissue remains unclear. RESEARCH DESIGN AND METHODS: In this study, we analyzed the phenotype of FoxO1 haploinsufficient mice to investigate the role of FoxO1 in high-fat diet-induced obesity and adipose tissue metabolism. RESULTS: We showed that reduced FoxO1 expression protects mice against obesity-related insulin resistance with marked improvement not only in hepatic insulin sensitivity but also in skeletal muscle insulin action. FoxO1 haploinsufficiency also resulted in increased peroxisome proliferator-activated receptor (PPAR)gamma gene expression in adipose tissue, with enhanced expression of PPARgamma target genes known to influence metabolism. Moreover, treatment of mice with the PPARgamma agonist rosiglitazone caused a greater improvement in in vivo insulin sensitivity in FoxO1 haploinsufficient animals, including reductions in circulating proinflammatory cytokines. CONCLUSIONS: These findings indicate that FoxO1 proteins negatively regulate insulin action and that their effect may be explained, at least in part, by inhibition of PPARgamma function.


Assuntos
Tecido Adiposo/fisiologia , Gorduras na Dieta/farmacologia , Fatores de Transcrição Forkhead/deficiência , Hipoglicemiantes/farmacologia , Resistência à Insulina/fisiologia , PPAR gama/genética , Tiazolidinedionas/farmacologia , Animais , Proteína Forkhead Box O1 , Deleção de Genes , Técnica Clamp de Glucose , Masculino , Camundongos , Fenótipo , RNA Mensageiro/genética , Valores de Referência , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
Nat Immunol ; 9(12): 1388-98, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18978794

RESUMO

The transcription factors Foxo1, Foxo3 and Foxo4 modulate cell fate 'decisions' in diverse systems. Here we show that Foxo1-dependent gene expression was critical at many stages of B cell differentiation. Early deletion of Foxo1 caused a substantial block at the pro-B cell stage due to a failure to express interleukin 7 receptor-alpha. Foxo1 inactivation in late pro-B cells resulted in an arrest at the pre-B cell stage due to lower expression of the recombination-activating genes Rag1 and Rag2. Deletion of Foxo1 in peripheral B cells led to fewer lymph node B cells due to lower expression of L-selectin and failed class-switch recombination due to impaired upregulation of the gene encoding activation-induced cytidine deaminase. Thus, Foxo1 regulates a transcriptional program that is essential for early B cell development and peripheral B cell function.


Assuntos
Linfócitos B/citologia , Diferenciação Celular/imunologia , Fatores de Transcrição Forkhead/imunologia , Animais , Linfócitos B/imunologia , Southern Blotting , Proteínas de Ligação a DNA/imunologia , Proteínas de Ligação a DNA/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Expressão Gênica/imunologia , Rearranjo Gênico do Linfócito B/genética , Proteínas de Homeodomínio/imunologia , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Mutantes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/imunologia , Células-Tronco/metabolismo , Transcrição Gênica/imunologia
5.
Cell Cycle ; 7(7): 837-41, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18414034

RESUMO

Human cancer cells frequently harbor chromosomal translocations that create chimeric fusion genes. The t(2;13) translocation is characteristic of the pediatric muscle tumor, alveolar rhabdomyosarcoma, and produces the chimeric transcription factor, PAX3-FOXO1, that contains the DNA binding elements of PAX3 and the transcriptional activation domain of FOXO1. Experiments designed to determine how PAX3-FOXO1 expression contributes to the development of muscle cell-derived tumors resulted in the discovery that the fusion protein misregulates gene expression and interrupts myogenic differentiation through a unique gain of function mechanism. These results yield new insight into how tumor-associated genetic alterations increase the likelihood of cancer formation and may lead to new therapeutic approaches.


Assuntos
Diferenciação Celular/fisiologia , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Fatores de Transcrição Box Pareados/metabolismo , Rabdomiossarcoma Alveolar/metabolismo , Western Blotting , Linhagem Celular , Proteína Forkhead Box O1 , Humanos , Imunoprecipitação , Modelos Biológicos , Mioblastos/citologia , Fator de Transcrição PAX3 , Ubiquitinação
6.
Genome Res ; 18(1): 46-59, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18042645

RESUMO

By integrating genome-wide maps of RNA polymerase II (Polr2a) binding with gene expression data and H3ac and H3K4me3 profiles, we characterized promoters with enriched activity in mouse embryonic stem cells (mES) as well as adult brain, heart, kidney, and liver. We identified approximately 24,000 promoters across these samples, including 16,976 annotated mRNA 5' ends and 5153 additional sites validating cap-analysis of gene expression (CAGE) 5' end data. We showed that promoters with CpG islands are typically non-tissue specific, with the majority associated with Polr2a and the active chromatin modifications in nearly all the tissues examined. By contrast, the promoters without CpG islands are generally associated with Polr2a and the active chromatin marks in a tissue-dependent way. We defined 4396 tissue-specific promoters by adapting a quantitative index of tissue-specificity based on Polr2a occupancy. While there is a general correspondence between Polr2a occupancy and active chromatin modifications at the tissue-specific promoters, a subset of them appear to be persistently marked by active chromatin modifications in the absence of detectable Polr2a binding, highlighting the complexity of the functional relationship between chromatin modification and gene expression. Our results provide a resource for exploring promoter Polr2a binding and epigenetic states across pluripotent and differentiated cell types in mammals.


Assuntos
Mapeamento Cromossômico , Ilhas de CpG/fisiologia , Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica/fisiologia , Genoma/fisiologia , Regiões Promotoras Genéticas/fisiologia , Animais , Diferenciação Celular/fisiologia , Cromatina/genética , Cromatina/metabolismo , Células-Tronco Embrionárias/citologia , Feminino , Camundongos , Especificidade de Órgãos/fisiologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/fisiologia , RNA Polimerase II/genética , RNA Polimerase II/metabolismo
7.
Proc Natl Acad Sci U S A ; 104(46): 18085-90, 2007 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-17986608

RESUMO

The chimeric protein PAX3-FOXO1, resulting from a translocation between chromosomes 2 and 13, is the most common genetic aberration in the alveolar subtype of the human skeletal muscle tumor, rhabdomyosarcoma. To understand how PAX3-FOXO1 contributes to tumor development, we isolated and characterized muscle cells from transgenic mice expressing PAX3-FOXO1 under control of the PAX3 promoter. We demonstrate that these myoblasts are unable to complete myogenic differentiation because of an inability to up-regulate p57Kip2 transcription. This defect is caused by reduced levels of the EGR1 transcriptional activator resulting from a direct, destabilizing interaction with PAX3-FOXO1. Neither PAX3 nor FOXO1 share the ability to regulate p57Kip2 transcription. Thus, the breakage and fusion of the genes encoding these transcription factors creates a unique chimeric protein that controls a key cell-cycle and -differentiation regulator.


Assuntos
Inibidor de Quinase Dependente de Ciclina p57/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Fatores de Transcrição Forkhead/fisiologia , Regulação da Expressão Gênica/fisiologia , Fatores de Transcrição Box Pareados/fisiologia , Animais , Ensaio de Desvio de Mobilidade Eletroforética , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Humanos , Hidrólise , Camundongos , Camundongos Transgênicos , Neoplasias Musculares/genética , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/genética , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Rabdomiossarcoma/genética
8.
Cell ; 128(2): 235-7, 2007 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-17254960

RESUMO

The FoxO transcription factors have been implicated in many processes including tumor suppression and cell death. In this issue, two groups now report on mice that conditionally lack the three predominant FoxO transcription factors. Demonstrate that FoxOs are critical for the long-term maintenance of hematopoietic stem cells, and show that FoxOs suppress the formation of hemangiomas and lymphomas in mice.


Assuntos
Transformação Celular Neoplásica/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Hemangioma/genética , Células-Tronco Hematopoéticas/metabolismo , Linfoma/genética , Proteínas Supressoras de Tumor/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Transformação Celular Neoplásica/genética , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Regulação Neoplásica da Expressão Gênica/genética , Hemangioma/metabolismo , Humanos , Linfoma/metabolismo , Proteínas Supressoras de Tumor/genética
9.
Transgenic Res ; 15(5): 595-614, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16952014

RESUMO

The t(2;13) chromosomal translocation is found in the majority of human alveolar rhabdomyosarcomas (RMS). The resulting PAX3-FKHR fusion protein contains PAX3 DNA-binding domains fused to the potent transactivation domain of FKHR, suggesting that PAX3-FKHR functions to deregulate PAX3-specific target genes and signaling pathways. We previously developed transgenic mice expressing PAX3-FKHR under the control of mouse Pax3 regulatory sequences to test this hypothesis. We reported that PAX3-FKHR interferes with normal Pax3 developmental functions, with mice exhibiting neural tube and neural crest abnormalities that mimic those found in Pax3-deficient Splotch mice. Here we expanded those studies to show that developmental expression of PAX3-FKHR results in aberrant myogenesis in the developing somites and neural tube, leading to ectopic skeletal muscle formation in the mature spinal cord. Gene expression profiling indicated that PAX3-FKHR expression in the developing neural tube induces a myogenic pattern of gene expression at the expense of the normal neurogenic program. Somite defects in PAX3-FKHR transgenic animals resulted in skeletal malformations that included rib fusions and mis-attachments. As opposed to the neural tube defects, the severity of the rib phenotype was rescued by reducing Pax3 levels through mating with Splotch mice. Embryos from the transgenic line expressing the highest levels of PAX3-FKHR had severe neural tube defects, including exencephaly, and almost half of the embryos died between gestational ages E13.5-E15.5. Nearly all of the embryos that survived to term died after birth due to severe spina bifida, rather than the absence of a muscular diaphragm. These studies reveal a prominent role for PAX3-FKHR in disrupting Pax3 functions and in deregulating skeletal muscle development, suggesting that this fusion protein plays a critical role in the pathogenesis of alveolar RMS by influencing the commitment and differentiation of the myogenic cell lineage.


Assuntos
Coristoma/genética , Fatores de Transcrição Forkhead/genética , Desenvolvimento Muscular/genética , Defeitos do Tubo Neural/genética , Fatores de Transcrição Box Pareados/genética , Proteínas Recombinantes de Fusão/genética , Animais , Diferenciação Celular/genética , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/fisiologia , Humanos , Camundongos , Camundongos Transgênicos , Músculo Esquelético/citologia , Defeitos do Tubo Neural/patologia , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/fisiologia , Proteínas Recombinantes de Fusão/fisiologia , Rabdomiossarcoma Alveolar/etiologia , Rabdomiossarcoma Alveolar/genética , Rabdomiossarcoma Alveolar/patologia , Somitos/patologia
10.
Exp Gerontol ; 41(8): 709-17, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16806782

RESUMO

The FOXO family of transcription factors has been implicated in several cellular processes including cell cycle arrest, cell death and protection from stress stimuli. FOXO function is influenced by multiple signaling pathways. Many of these pathways are known to be misregulated in cancer. Perturbation of FOXO function leads to uncontrolled cell proliferation and accumulation of DNA damage. It is becoming clear that active FOXO proteins play an important role in keeping cells in check and inactivation of FOXO proteins is associated with characteristics of cancer cells. FOXO proteins may represent new therapeutic targets for a broad spectrum of cancers.


Assuntos
Transformação Celular Neoplásica/genética , Fatores de Transcrição Forkhead/fisiologia , Neoplasias/genética , Acetilação , Animais , Antineoplásicos/farmacologia , Dano ao DNA , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica , Proteínas de Neoplasias/fisiologia , Fosforilação , Ubiquitina/metabolismo
11.
Nat Genet ; 38(5): 589-93, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16642023

RESUMO

The total mass of islets of Langerhans is reduced in individuals with type 2 diabetes, possibly contributing to the pathogenesis of this condition. Although the regulation of islet mass is complex, recent studies have suggested the importance of a signaling pathway that includes the insulin or insulin-like growth factor-1 receptors, insulin receptor substrate and phosphatidylinositol (PI) 3-kinase. 3-Phosphoinositide-dependent protein kinase 1 (PDK1) is a serine-threonine kinase that mediates signaling downstream of PI 3-kinase. Here we show that mice that lack PDK1 specifically in pancreatic beta cells (betaPdk1-/- mice) develop progressive hyperglycemia as a result of a loss of islet mass. The mice show reductions in islet density as well as in the number and size of cells. Haploinsufficiency of the gene for the transcription factor Foxo1 resulted in a marked increase in the number, but not the size, of cells and resulted in the restoration of glucose homeostasis in betaPdk1-/- mice. These results suggest that PDK1 is important in maintenance of pancreatic cell mass and glucose homeostasis.


Assuntos
Diabetes Mellitus Experimental/genética , Ilhotas Pancreáticas/enzimologia , Ilhotas Pancreáticas/patologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Animais , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Experimental/patologia , Camundongos , Camundongos Knockout , Transdução de Sinais
12.
Science ; 307(5716): 1720-1, 2005 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-15779114
13.
14.
Cell ; 117(4): 421-6, 2004 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-15137936

RESUMO

Forkhead transcription factors of the FoxO subfamily are emerging as a shared component among pathways regulating diverse cellular functions, such as differentiation, metabolism, proliferation, and survival. Their transcriptional output is controlled via a two-tiered mechanism of phosphorylation and acetylation. Modest alterations of this balance can result in profound effects. The gamut of phenotypes runs from protection against diabetes and predisposition to neoplasia, conferred by FoxO loss of function, to increased cellular survival and a marked catabolic response associated with gain of function.


Assuntos
Transformação Celular Neoplásica/genética , Metabolismo Energético/genética , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Acetilação , Animais , Diferenciação Celular/genética , Sobrevivência Celular/genética , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Genes Letais/genética
15.
Mol Cell ; 14(4): 416-8, 2004 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-15149589

RESUMO

Two recent reports reveal new roles for FoxO proteins in cell proliferation and tumorigenesis. Seoane and colleagues show that FoxO proteins play key roles in the TGFbeta-dependent activation of p21Cip1 by partnering with Smad3 and Smad4. FoxG1, a protein from a distinct Fox subfamily, binds FoxO/Smad complexes and blocks p21Cip1 expression. These interactions establish a relationship between the PI3K pathway, FoxG1, and the TGFbeta/Smad pathways. The second report identifies IkappaB kinase as a negative regulator of FoxO proteins, suggesting a mechanism for relieving negative regulation of cell cycle and promoting tumor cell proliferation.


Assuntos
Transformação Celular Neoplásica/genética , Transdução de Sinais/genética , Fatores de Transcrição/genética , Animais , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/genética , Divisão Celular/genética , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Proteínas de Ligação a DNA/genética , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Mitocondriais/genética , Proteínas do Tecido Nervoso/genética , Fosfatidilinositol 3-Quinases/genética , Fator de Crescimento Transformador beta/genética
16.
Proc Natl Acad Sci U S A ; 101(9): 2975-80, 2004 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-14978268

RESUMO

Genetic analysis in Caenorhabditis elegans has uncovered essential roles for DAF-16 in longevity, metabolism, and reproduction. The mammalian orthologs of DAF-16, the closely-related FOXO subclass of forkhead transcription factors (FKHR/FOXO1, FKHRL1/FOXO3a, and AFX/FOXO4), also have important roles in cell cycle arrest, apoptosis and stress responses in vitro, but their in vivo physiological roles are largely unknown. To elucidate their role in normal development and physiology, we disrupted each of the Foxo genes in mice. Foxo1-null embryos died on embryonic day 10.5 as a consequence of incomplete vascular development. Foxo1-null embryonic and yolk sac vessels were not well developed at embryonic day 9.5, and Foxo1 expression was found in a variety of embryonic vessels, suggesting a crucial role of this transcription factor in vascular formation. On the other hand, both Foxo3a- and Foxo4-null mice were viable and grossly indistinguishable from their littermate controls, indicating dispensability of these two members of the Foxo transcription factor family for normal vascular development. Foxo3a-null females showed age-dependent infertility and had abnormal ovarian follicular development. In contrast, histological analyses of Foxo4-null mice did not identify any consistent abnormalities. These results demonstrate that the physiological roles of Foxo genes are functionally diverse in mammals.


Assuntos
Desenvolvimento Embrionário e Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Variação Genética , Fatores de Transcrição/genética , Animais , Feminino , Morte Fetal , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead , Infertilidade Feminina/genética , Masculino , Camundongos , Família Multigênica , Neovascularização Fisiológica/genética , Ovário/embriologia , Deleção de Sequência , Saco Vitelino/fisiologia
17.
Proc Natl Acad Sci U S A ; 101(2): 615-20, 2004 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-14722351

RESUMO

Prostate tumors are complex entities composed of malignant cells mixed and interacting with nonmalignant cells. However, molecular analyses by standard gene expression profiling are limited because spatial information and nontumor cell types are lost in sample preparation. We scored 88 prostate specimens for relative content of tumor, benign hyperplastic epithelium, stroma, and dilated cystic glands. The proportions of these cell types were then linked in silico to gene expression levels determined by microarray analysis, revealing unique cell-specific profiles. Gene expression differences for malignant and nonmalignant epithelial cells (tumor versus benign hyperplastic epithelium) could be identified without being confounded by contributions from stroma that dominate many samples or sacrificing possible paracrine influences. Cell-specific expression of selected genes was validated by immunohistochemistry and quantitative PCR. The results provide patterns of gene expression for these three lineages with relevance to pathogenetic, diagnostic, and therapeutic considerations.


Assuntos
Perfilação da Expressão Gênica , Neoplasias da Próstata/genética , Humanos , Imuno-Histoquímica , Masculino , Hibridização de Ácido Nucleico , Neoplasias da Próstata/patologia
18.
Genomics ; 82(6): 628-36, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14611804

RESUMO

Human SSX was first identified as the gene involved in the t(X;18) translocation in synovial sarcoma. SSX is a multigene family, with 9 complete genes on chromosome Xp11. Normally expressed almost exclusively in testis, SSX mRNA is expressed in various human tumors, defining SSX as a cancer/testis antigen. We have now cloned the mouse ortholog of SSX. Mouse SSX genes can be divided into Ssxa and Ssxb subfamilies based on sequence homology. Ssxa has only one member, whereas 12 Ssxb genes, Ssxb1 to Ssxb12, were identified by cDNA cloning from mouse testis and mouse tumors. Both Ssxa and Ssxb are located on chromosome X and show tissue-restricted mRNA expression to testis among normal tissues. All putative human and mouse SSX proteins share conserved KRAB and SSX-RD domains. Mouse tumors were found to express some, but not all, Ssxb genes, similar to the SSX activation in human tumors.


Assuntos
Expressão Gênica , Família Multigênica/genética , Proteínas de Neoplasias/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras/genética , Testículo/metabolismo , Cromossomo X/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Southern Blotting , Mapeamento Cromossômico , Componentes do Gene , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , RNA Mensageiro/genética , Mapeamento por Restrição , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Análise de Sequência de DNA , Homologia de Sequência , Células Tumorais Cultivadas
19.
Dev Cell ; 4(1): 119-29, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12530968

RESUMO

An outstanding question in adipocyte biology is how hormonal cues are relayed to the nucleus to activate the transcriptional program that promotes adipogenesis. The forkhead transcription factor Foxo1 is regulated by insulin via Akt-dependent phosphorylation and nuclear exclusion. We show that Foxo1 is induced in the early stages of adipocyte differentiation but that its activation is delayed until the end of the clonal expansion phase. Constitutively active Foxo1 prevents the differentiation of preadipocytes, while dominant-negative Foxo1 restores adipocyte differentiation of fibroblasts from insulin receptor-deficient mice. Further, Foxo1 haploinsufficiency protects from diet-induced diabetes in mice. We propose that Foxo1 plays an important role in the integration of hormone-activated signaling pathways with the complex transcriptional cascade that promotes adipocyte differentiation.


Assuntos
Adipócitos/citologia , Adipócitos/metabolismo , Diferenciação Celular , Fatores de Transcrição/metabolismo , Células 3T3 , Tecido Adiposo/crescimento & desenvolvimento , Tecido Adiposo/metabolismo , Animais , Tamanho Celular , Diabetes Mellitus/induzido quimicamente , Diabetes Mellitus/metabolismo , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/farmacologia , Fibroblastos , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead , Regulação da Expressão Gênica , Resistência à Insulina , Camundongos , Mutação/genética , Fosforilação , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Fatores de Transcrição/genética
20.
Cancer Res ; 62(23): 6981-9, 2002 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-12460916

RESUMO

We have recently characterized T24T, an invasive and metastatic variant of the T24 human bladder cell line, resulting in a model for bladder cancer progression. To gain additional insight into the repertoire of genetic changes that may be responsible for the invasive and metastatic phenotype, we used spectral karyotyping (SKY) in combination with comparative genomic hybridization (CGH) in these cells. To assess the functional significance of the genetic differences found between the two cell lines, we have developed a positional expression profiling (PEP) method for comparing gene expression data obtained from oligonucleotide microarrays based upon chromosomal position. Using SKY and CGH, we were able to define the genetic changes in the cell lines, and in addition, resolve the identity of all marker chromosomes from our initial karyotyping and G-band analysis. PEP analysis revealed important similarities and differences when compared with the cytogenetic data, allowing insights of how genomic structural changes affect gene expression on a regional scale. The shape of the expression profiles for chromosomes 8, 12, and X correlated well with the numerical imbalances revealed by CGH and SKY, whereas regions like 10q, gained in T24T compared with T24, was not associated with changes in gene expression. Furthermore, we have shown that 12p, a region of agreement between CGH and PEP harbors RhoGDI2, a candidate gene, the expression of which inversely correlates with bladder tumor progression, demonstrating the usefulness of this multimodal approach in identifying promising genetic changes that may be responsible for the invasive phenotype.


Assuntos
Aberrações Cromossômicas , Neoplasias da Bexiga Urinária/genética , Perfilação da Expressão Gênica , Ligação Genética , Humanos , Cariotipagem , Invasividade Neoplásica , Metástase Neoplásica , Hibridização de Ácido Nucleico , Análise de Sequência com Séries de Oligonucleotídeos , Células Tumorais Cultivadas , Neoplasias da Bexiga Urinária/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...