Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Mod Pathol ; 34(2): 438-444, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32792597

RESUMO

Diagnostic testing of pancreatic cyst fluid obtained by endoscopic ultrasound-guided fine needle aspiration (EUS-FNA) has traditionally utilized elevated carcinoembryonic antigen (CEA) (≥192 ng/ml) and cytomorphologic examination to differentiate premalignant mucinous from benign pancreatic cystic lesions (PCLs). Molecular testing for KRAS/GNAS mutations has been shown to improve accuracy of detecting mucinous PCLs. Using a targeted next-generation sequencing (NGS) panel, we assess the status of PCL-associated mutations to improve understanding of the key diagnostic variables. Molecular analysis of cyst fluid was performed on 108 PCLs that had concurrent CEA and/or cytological analysis. A 48-gene NGS assay was utilized, which included genes commonly mutated in mucinous PCLs such as GNAS, KRAS, CDKN2A, and TP53. KRAS and/or GNAS mutations were seen in 59 of 68 (86.8%) cases with multimodality diagnosis of a mucinous PCL. Among 31 patients where surgical histopathology was available, the sensitivity, specificity, and diagnostic accuracy of NGS for the diagnosis of mucinous PCL was 88.5%, 100%, and 90.3%, respectively. Cytology with mucinous/atypical findings were found in only 29 of 62 cases (46.8%), with fluid CEA elevated in 33 of 58 cases (56.9%). Multiple KRAS mutations at different variant allele frequencies were seen in seven cases favoring multiclonal patterns, with six of them showing at least two separate PCLs by imaging. Among the 6 of 10 cases with GNAS + /KRAS- results, uncommon, non-V600E exon 11/15 hotspot BRAF mutations were identified. The expected high degree of accuracy of NGS detection of KRAS and/or GNAS mutations for mucinous-PCLs, as compared with CEA and cytological examination, was demonstrated. Multiple KRAS mutations correlated with multifocal cysts demonstrated by radiology. In IPMNs that lacked KRAS mutations, the concurring BRAF mutations with GNAS mutations supports an alternate mechanism of activation in the Ras pathway.


Assuntos
Biomarcadores/análise , Cisto Pancreático/diagnóstico , Cisto Pancreático/genética , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Idoso , Líquido Cístico/química , Análise Mutacional de DNA/métodos , Aspiração por Agulha Fina Guiada por Ultrassom Endoscópico , Feminino , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Cisto Pancreático/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Sensibilidade e Especificidade , Transdução de Sinais
2.
Dis Model Mech ; 8(9): 1121-7, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26092125

RESUMO

Scaffold attachment factors SAFB1 and SAFB2 are multifunctional proteins that share >70% sequence similarity. SAFB1-knockout (SAFB1(-/-)) mice display a high degree of lethality, severe growth retardation, and infertility in male mice. To assess the in vivo role of SAFB2, and to identify unique functions of the two paralogs, we generated SAFB2(-/-) mice. In stark contrast to SAFB1(-/-), SAFB2(-/-) offspring were born at expected Mendelian ratios and did not show any obvious defects in growth or fertility. Generation of paralog-specific antibodies allowed extensive expression analysis of SAFB1 and SAFB2 in mouse tissues, showing high expression of both SAFB1 and SAFB2 in the immune system, and in hormonally controlled tissues, with especially high expression of SAFB2 in the male reproductive tract. Further analysis showed a significantly increased testis weight in SAFB2(-/-) mice, which was associated with an increased number of Sertoli cells. Our data suggest that this is at least in part caused by alterations in androgen-receptor function and expression upon deletion of SAFB2. Thus, despite a high degree of sequence similarity, SAFB1(-/-) and SAFB2(-/-) mice do not totally phenocopy each other. SAFB2(-/-) mice are viable, and do not show any major defects, and our data suggest a role for SAFB2 in the differentiation and activity of Sertoli cells that deserves further study.


Assuntos
Proteínas de Transporte/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a RNA/genética , Animais , Peso Corporal , Proteínas de Transporte/fisiologia , Diferenciação Celular , Proteínas de Ligação a DNA/fisiologia , Modelos Animais de Doenças , Éxons , Feminino , Deleção de Genes , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Proteínas de Ligação a RNA/fisiologia , Reprodução , Células de Sertoli/citologia , Transdução de Sinais
3.
Mol Endocrinol ; 28(6): 846-59, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24678732

RESUMO

Transcription of the HER2 oncogene can be repressed by estrogen (E2). We now show that, a splice isoform of the nuclear receptor coactivator AIB1, AIB1-Δ4, is able to reverse E2 repression of HER2 gene expression in breast cancer cells. The first 224 amino acids of AIB1 that are absent in AIB1-Δ4, bind a co-repressor, ANCO1. Using chromatin immunoprecipitation assay approaches in MCF7 and BT474 cell lines, we demonstrate that AIB1 and AIB1-Δ4 can bind to the E2 regulatory site in the first intron of the HER2 gene, after E2 treatment, but only full-length AIB1 recruits ANCO1. Consistent with E2-induced chromatin repression, the AIB1-ANCO1 complex recruits HDAC3 and HDAC4 to the intronic estrogen response element and the proximal promoter acquires the repressive chromatin mark H3K9me3 and loses H3K4me1. In contrast, AIB1-Δ4 does not recruit ANCO 1, HDAC3, or HDAC4 and the proximal promoter retains activation marks of H3K4me1. In cell lines with low levels of ANCO1 (T47D), E2 does not repress HER2 gene transcription but the repressive response can be restored by overexpression of ANCO1. ANCO1 can also repress other E2-responsive genes, indicating that AIB1, AIB1-Δ4 and ANCO1 are important determinants of endocrine and growth factor responsiveness in breast cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Coativador 3 de Receptor Nuclear/fisiologia , Receptor ErbB-2/genética , Proteínas Repressoras/fisiologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Montagem e Desmontagem da Cromatina , Estradiol/fisiologia , Feminino , Células HEK293 , Histona Desacetilases/metabolismo , Humanos , Células MCF-7 , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/metabolismo , Oncogenes , Ligação Proteica , Receptor ErbB-2/metabolismo , Elementos de Resposta , Transdução de Sinais , Transcrição Gênica
4.
Sci Transl Med ; 6(229): 229ra41, 2014 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-24670685

RESUMO

Resistance to aromatase inhibitors (AIs) is a major clinical problem in the treatment of estrogen receptor (ER)-positive breast cancer. In two breast cancer cell line models of AI resistance, we identified widespread DNA hyper- and hypomethylation, with enrichment for promoter hypermethylation of developmental genes. For the homeobox gene HOXC10, methylation occurred in a CpG shore, which overlapped with a functional ER binding site, causing repression of HOXC10 expression. Although short-term blockade of ER signaling caused relief of HOXC10 repression in both cell lines and breast tumors, it also resulted in concurrent recruitment of EZH2 and increased H3K27me3, ultimately transitioning to increased DNA methylation and silencing of HOXC10. Reduced HOXC10 in vitro and in xenografts resulted in decreased apoptosis and caused antiestrogen resistance. Supporting this, we used paired primary and metastatic breast cancer specimens to show that HOXC10 was reduced in tumors that recurred during AI treatment. We propose a model in which estrogen represses apoptotic and growth-inhibitory genes such as HOXC10, contributing to tumor survival, whereas AIs induce these genes to cause apoptosis and therapeutic benefit, but long-term AI treatment results in permanent repression of these genes via methylation and confers resistance. Therapies aimed at inhibiting AI-induced histone and DNA methylation may be beneficial in blocking or delaying AI resistance.


Assuntos
Neoplasias da Mama/genética , Reprogramação Celular/genética , Resistencia a Medicamentos Antineoplásicos/genética , Epigênese Genética/genética , Estrogênios/farmacologia , Proteínas de Homeodomínio/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Inibidores da Aromatase/farmacologia , Inibidores da Aromatase/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Reprogramação Celular/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Histonas/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Células MCF-7 , Camundongos , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Regiões Promotoras Genéticas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Mol Endocrinol ; 27(7): 1113-27, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23660594

RESUMO

The oncogene nuclear receptor coactivator amplified in breast cancer 1 (AIB1) is a transcriptional coactivator that is overexpressed in various types of human cancers. However, the molecular mechanisms controlling AIB1 expression in the majority of cancers remain unclear. In this study, we identified a novel interacting protein of AIB1, forkhead-box protein G1 (FoxG1), which is an evolutionarily conserved forkhead-box transcriptional corepressor. We show that FoxG1 expression is low in breast cancer cell lines and that low levels of FoxG1 are correlated with a worse prognosis in breast cancer. We also demonstrate that transient overexpression of FoxG1 can suppress endogenous levels of AIB1 mRNA and protein in MCF-7 breast cancer cells. Exogenously expressed FoxG1 in MCF-7 cells also leads to apoptosis that can be rescued in part by AIB1 overexpression. Using chromatin immunoprecipitation, we determined that FoxG1 is recruited to a region of the AIB1 gene promoter previously characterized to be responsible for AIB1-induced, positive autoregulation of transcription through the recruitment of an activating, multiprotein complex, involving AIB1, E2F transcription factor 1, and specificity protein 1. Increased FoxG1 expression significantly reduces the recruitment of AIB1, E2F transcription factor 1 and E1A-binding protein p300 to this region of the endogenous AIB1 gene promoter. Our data imply that FoxG1 can function as a pro-apoptotic factor in part through suppression of AIB1 coactivator transcription complex formation, thereby reducing the expression of the AIB1 oncogene.


Assuntos
Apoptose , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Fatores de Transcrição Forkhead/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Coativador 3 de Receptor Nuclear/genética , Proteínas Repressoras/metabolismo , Transcrição Gênica , Apoptose/genética , Regulação para Baixo/genética , Fator de Transcrição E2F1/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Células MCF-7 , Modelos Biológicos , Coativador 3 de Receptor Nuclear/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica/genética , Estabilidade Proteica , Fator de Transcrição Sp1/metabolismo
6.
Biochem Biophys Res Commun ; 408(4): 516-22, 2011 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-21527249

RESUMO

Sumoylation is an emerging modification associated with a variety of cellular processes including the regulation of transcriptional activities of nuclear receptors and their coregulators. As SUMO modifications are often associated with transcriptional repression, we examined if sumoylation was involved in modulation of the transcriptional repressive activity of scaffold attachment factor B1. Here we show that SAFB1 is modified by both the SUMO1 and SUMO2/3 family of proteins, on lysine's K231 and K294. Further, we demonstrate that SAFB1 can interact with PIAS1, a SUMO E3 ligase which mediates SAFB1 sumoylation. Additionally, SENP1 was identified as the enzyme desumoylating SAFB1. Mutation of the SAFB1 sumoylation sites lead to a loss of transcriptional repression, at least in part due to decreased interaction with HDAC3, a known transcriptional repressor and SAFB1 binding partner. In summary, the transcriptional repressor SAFB1 is modified by both SUMO1 and SUMO2/3, and this modification is necessary for its full repressive activity.


Assuntos
Proteínas Correpressoras/metabolismo , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Proteínas Associadas à Matriz Nuclear/metabolismo , Receptores de Estrogênio/metabolismo , Sumoilação , Sequência de Aminoácidos , Linhagem Celular Tumoral , Proteínas Correpressoras/genética , Sequência Consenso , Regulação da Expressão Gênica , Células HEK293 , Humanos , Proteínas de Ligação à Região de Interação com a Matriz/genética , Dados de Sequência Molecular , Proteínas Associadas à Matriz Nuclear/genética , Proteínas Inibidoras de STAT Ativados/metabolismo , Receptores de Estrogênio/genética , Proteína SUMO-1 , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Transcrição Gênica , Ubiquitinas
7.
Mol Cell Biol ; 30(2): 399-412, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19917725

RESUMO

Activation of estrogen receptor alpha (ERalpha) results in both induction and repression of gene transcription; while mechanistic details of estrogen induction are well described, details of repression remain largely unknown. We characterized several ERalpha-repressed targets and examined in detail the mechanism for estrogen repression of Reprimo (RPRM), a cell cycle inhibitor. Estrogen repression of RPRM is rapid and robust and requires a tripartite interaction between ERalpha, histone deacetylase 7 (HDAC7), and FoxA1. HDAC7 is the critical HDAC needed for repression of RPRM; it can bind to ERalpha and represses ERalpha's transcriptional activity--this repression does not require HDAC7's deacetylase activity. We further show that the chromatin pioneer factor FoxA1, well known for its role in estrogen induction of genes, is recruited to the RPRM promoter, is necessary for repression of RPRM, and interacts with HDAC7. Like other FoxA1 recruitment sites, the RPRM promoter is characterized by H3K4me1/me2. Estrogen treatment causes decreases in H3K4me1/me2 and release of RNA polymerase II (Pol II) from the RPRM proximal promoter. Overall, these data implicate a novel role for HDAC7 and FoxA1 in estrogen repression of RPRM, a mechanism which could potentially be generalized to many more estrogen-repressed genes and hence be important in both normal physiology and pathological processes.


Assuntos
Proteínas de Ciclo Celular/genética , Receptor alfa de Estrogênio/metabolismo , Regulação da Expressão Gênica , Glicoproteínas/genética , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Histona Desacetilases/metabolismo , Proteínas de Ciclo Celular/antagonistas & inibidores , Linhagem Celular Tumoral , Metilação de DNA/efeitos dos fármacos , Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio/efeitos dos fármacos , Estrogênios/farmacologia , Fulvestranto , Glicoproteínas/antagonistas & inibidores , Fator 3-alfa Nuclear de Hepatócito/efeitos dos fármacos , Histona Desacetilases/efeitos dos fármacos , Histonas/efeitos dos fármacos , Histonas/metabolismo , Humanos , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/fisiologia , RNA Polimerase II/efeitos dos fármacos , RNA Polimerase II/metabolismo , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/fisiologia
8.
J Cell Biochem ; 109(2): 312-9, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20014070

RESUMO

The examination of scaffold attachment factor B1 (SAFB1) and its multiple functions and tasks in cellular processes provides insight into its role in diseases, such as cancer. SAFB1 is a large multi-domain protein with well-described functions in transcriptional repression, and RNA splicing. It is ubiquitously expressed, and has been shown to be important in numerous cellular processes including cell growth, stress response, and apoptosis. SAFB1 is part of a protein family with at least two other family members, SAFB2 and the SAFB-like transcriptional modulator SLTM. The goal of this prospect article is to summarize known functions of SAFB1, and its roles in cellular processes, but also to speculate on less well described, novel attributes of SAFB1, such as a potential role in chromatin organization. This timely review shows aspects of SAFB1, which are proving to have a complexity far greater than was previously thought.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Proteínas de Ligação à Região de Interação com a Matriz/fisiologia , Proteínas Associadas à Matriz Nuclear/fisiologia , Proteínas de Ligação a RNA/fisiologia , Receptores de Estrogênio/fisiologia , Animais , Apoptose , Proliferação de Células , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina , Regulação da Expressão Gênica , Humanos , Proteínas de Ligação à Região de Interação com a Matriz/química , Camundongos , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Associadas à Matriz Nuclear/química , Especificidade de Órgãos , Estrutura Terciária de Proteína , Receptores de Estrogênio/química , Transdução de Sinais/fisiologia , Estresse Fisiológico/fisiologia , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA