Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Int J Cancer ; 139(4): 899-907, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27062469

RESUMO

Centromere protein-A (CENP-A), a histone-H3 variant, plays an essential role in cell division by ensuring proper formation and function of centromeres and kinetochores. Elevated CENP-A expression has been associated with cancer development. This study aimed to establish whether elevated CENP-A expression can be used as a prognostic and predictive cancer biomarker. Molecular profiling of CENP-A in human cancers was investigated using genomic, transcriptomic and patient information from databases, including COSMIC, Oncomine, Kaplan-Meier plotter and cBioPortal. A network of CENP-A co-expressed genes was derived from cBioPortal and analyzed using Ingenuity Pathway Analysis (IPA) and Oncomine protocols to explore the function of CENP-A and its predictive potential. Transcriptional and post-transcriptional regulation of CENP-A expression was analyzed in silico. It was found that CENP-A expression was elevated in 20 types of solid cancer compared with normal counterparts. Elevated CENP-A expression highly correlated with cancer progression and poor patient outcome. Genomic analysis indicated that the elevated CENP-A expression was not due to alterations in the sequence or copy number of the CENP-A gene. Furthermore, CENP-A can be regulated by key oncogenic proteins and tumor-suppressive microRNAs. CENP-A co-expression network analysis indicated that CENP-A function is associated with cell cycle progression. Oncomine analysis showed a strong correlation between elevated CENP-A expression and oncolytic response of breast cancer patients to taxane-based chemotherapy. In conclusion, elevated CENP-A expression is coupled to malignant progression of numerous types of cancer. It may be useful as a biomarker of poor patient prognosis and as a predictive biomarker for taxane-based chemotherapy.


Assuntos
Autoantígenos/genética , Biomarcadores Tumorais , Proteínas Cromossômicas não Histona/genética , Expressão Gênica , Neoplasias/genética , Neoplasias/mortalidade , Proteína Centromérica A , Bases de Dados Genéticas , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Estimativa de Kaplan-Meier , Mutação , Metástase Neoplásica , Neoplasias/metabolismo , Prognóstico , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Transdução de Sinais
2.
Mol Cancer ; 14: 46, 2015 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-25744782

RESUMO

BACKGROUND: Long non-coding RNAs (lncRNAs) can orchestrate oncogenic or tumor-suppressive functions in cancer biology. Accordingly, PCGEM1 and PRNCR1 were implicated in progression of prostate cancer (PCa) as transcriptional co-regulators of the androgen receptor (AR). However, these findings were recently refuted asserting that neither gene physically binds to the AR. Despite evidence for differing AR transcriptional programs in vivo and in vitro, studies investigating AR-regulation of these genes hitherto have only been conducted in vitro. Here, we further examine the relevance of PCGEM1 and PRNCR1 in PCa, and their relationship with AR signaling, using patient-derived xenograft models. FINDINGS: RNA sequencing of two distinct androgen-dependent models shows PCGEM1 to be considerably expressed, while PRNCR1 showed scant basal expression. PCGEM1 was sharply down-regulated following castration and up-regulated upon AR activation in vivo. However, we found no parallel evidence following AR stimulation in vitro. A PCGEM1-associated gene expression signature (PES) was significantly repressed in response to androgen ablation therapy and in hormone-refractory versus hormone-naïve PCa patients. Furthermore, we found PCGEM1 was uniformly distributed in PCa cell nucleus and cytoplasm which remained unaltered upon AR transcriptional activation. PCGEM1 was up-regulated in primary PCa but not in metastasized PCa. Accordingly, the PES was significantly down-regulated in advanced and higher grade PCa patients from multiple independent studies. CONCLUSION: Our results demonstrate PCGEM1 as an in vivo androgen-regulated transcript with potential nuclear and/or cytoplasmic function(s). Importantly, the clinical expression profile of PCGEM1 implicates it in the early stages of PCa warranting further research in this direction.


Assuntos
Neoplasias da Próstata/genética , RNA Longo não Codificante/genética , Receptores Androgênicos/genética , Androgênios/genética , Animais , Linhagem Celular Tumoral , Núcleo Celular/genética , Citoplasma/genética , Regulação para Baixo/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Masculino , Camundongos , Transdução de Sinais/genética , Transcrição Gênica/genética , Ativação Transcricional/genética , Regulação para Cima/genética
3.
Cancer Metastasis Rev ; 33(1): 1-16, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24346158

RESUMO

Since the discovery of microRNAs, non-coding RNAs (NC-RNAs) have increasingly attracted the attention of cancer investigators. Two classes of NC-RNAs are emerging as putative metastasis-related genes: long non-coding RNAs (lncRNAs) and small nucleolar RNAs (snoRNAs). LncRNAs orchestrate metastatic progression through several mechanisms, including the interaction with epigenetic effectors, splicing control and generation of microRNA-like molecules. In contrast, snoRNAs have been long considered "housekeeping" genes with no relevant function in cancer. However, recent evidence challenges this assumption, indicating that some snoRNAs are deregulated in cancer cells and may play a specific role in metastasis. Interestingly, snoRNAs and lncRNAs share several mechanisms of action, and might synergize with protein-coding genes to generate a specific cellular phenotype. This evidence suggests that the current paradigm of metastatic progression is incomplete. We propose that NC-RNAs are organized in complex interactive networks which orchestrate cellular phenotypic plasticity. Since plasticity is critical for cancer cell metastasis, we suggest that a molecular interactome composed by both NC-RNAs and proteins orchestrates cancer metastasis. Interestingly, expression of lncRNAs and snoRNAs can be detected in biological fluids, making them potentially useful biomarkers. NC-RNA expression profiles in human neoplasms have been associated with patients' prognosis. SnoRNA and lncRNA silencing in pre-clinical models leads to cancer cell death and/or metastasis prevention, suggesting they can be investigated as novel therapeutic targets. Based on the literature to date, we critically discuss how the NC-RNA interactome can be explored and manipulated to generate more effective diagnostic, prognostic, and therapeutic strategies for metastatic neoplasms.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias/genética , RNA não Traduzido/genética , Transcriptoma , Biomarcadores Tumorais/genética , Humanos , Modelos Genéticos , Metástase Neoplásica , Neoplasias/patologia , RNA Longo não Codificante/genética , RNA Nucleolar Pequeno/genética
4.
Prostate ; 73(16): 1747-60, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24038102

RESUMO

BACKGROUND: Androgen signalling through the androgen receptor (AR) plays a critical role in prostate cancer (PCa) initiation and progression. Estrogen in synergy with androgen is essential for cell growth of the normal and malignant prostate. However, the exact role that estrogen and the estrogen receptor play in prostate carcinogenesis remains unclear. We have previously demonstrated the metastasis-promoting effect of an estrogen receptor beta (ERß) agonist (genistein) in a patient-derived PCa xenograft model mimicking localized and metastatic disease. METHODS: To test the hypothesis that the tumor-promoting activity of genistein was due to its estrogenic properties, we treated the xenograft-bearing mice with genistein and an anti-estrogen compound (ICI 182, 780) and compared the differential gene expression using microarrays. RESULTS: Using a second xenograft model which was derived from another patient, we showed that genistein promoted disease progression in vivo and ICI 182, 780 inhibited metastatic spread. The microarray analysis revealed that the metallothionein (MT) gene family was differentially expressed in tumors treated by these compounds. Using qRT-PCR, the differences in expression levels were validated in the metastatic and non-metastatic LTL313 PCa xenograft tumor lines, both of which were originally derived from the same PCa patient. CONCLUSIONS: Together our data provide evidence that genistein stimulates and ICI 182, 780 inhibits metastatic progression, suggesting that these effects may be mediated by ERß signalling.


Assuntos
Antineoplásicos/uso terapêutico , Progressão da Doença , Estradiol/análogos & derivados , Antagonistas de Estrogênios/uso terapêutico , Genisteína/uso terapêutico , Metástase Neoplásica/tratamento farmacológico , Neoplasias da Próstata/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Modelos Animais de Doenças , Estradiol/farmacologia , Estradiol/uso terapêutico , Antagonistas de Estrogênios/farmacologia , Receptor beta de Estrogênio/efeitos dos fármacos , Fulvestranto , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genisteína/farmacologia , Humanos , Masculino , Metalotioneína/genética , Metalotioneína/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias da Próstata/patologia , RNA Interferente Pequeno/farmacologia , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Genome Res ; 20(8): 1037-51, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20551221

RESUMO

The liver and pancreas share a common origin and coexpress several transcription factors. To gain insight into the transcriptional networks regulating the function of these tissues, we globally identify binding sites for FOXA2 in adult mouse islets and liver, PDX1 in islets, and HNF4A in liver. Because most eukaryotic transcription factors bind thousands of loci, many of which are thought to be inactive, methods that can discriminate functionally active binding events are essential for the interpretation of genome-wide transcription factor binding data. To develop such a method, we also generated genome-wide H3K4me1 and H3K4me3 localization data in these tissues. By analyzing our binding and histone methylation data in combination with comprehensive gene expression data, we show that H3K4me1 enrichment profiles discriminate transcription factor occupied loci into three classes: those that are functionally active, those that are poised for activation, and those that reflect pioneer-like transcription factor activity. Furthermore, we demonstrate that the regulated presence of H3K4me1-marked nucleosomes at transcription factor occupied promoters and enhancers controls their activity, implicating both tissue-specific transcription factor binding and nucleosome remodeling complex recruitment in determining tissue-specific gene expression. Finally, we apply these approaches to generate novel insights into how FOXA2, PDX1, and HNF4A cooperate to drive islet- and liver-specific gene expression.


Assuntos
Loci Gênicos , Fator 3-beta Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/genética , Histonas/genética , Proteínas de Homeodomínio/genética , Ilhotas Pancreáticas/metabolismo , Fígado/metabolismo , Nucleossomos/genética , Transativadores/genética , Animais , Sequência de Bases , Sítios de Ligação , Perfilação da Expressão Gênica , Fator 3-beta Nuclear de Hepatócito/metabolismo , Fator 4 Nuclear de Hepatócito/metabolismo , Histonas/metabolismo , Proteínas de Homeodomínio/metabolismo , Camundongos , Dados de Sequência Molecular , Nucleossomos/metabolismo , Sequências Reguladoras de Ácido Nucleico , Transativadores/metabolismo
6.
Int J Mol Sci ; 14(4): 7757-70, 2013 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-23574937

RESUMO

MicroRNAs (miRNAs) have emerged as key regulators of numerous biological processes, and increasing evidence suggests that circulating miRNAs may be useful biomarkers of clinical disease. In this study, we sought to identify plasma miRNAs that differentiate patients with metastatic castration resistant prostate cancer (mCRPC) from those with localized prostate cancer (PCa). Pooled plasma samples from patients with localized PCa or mCRPC (25 per group) were assayed using the Exiqon miRNA qPCR panel, and the differential expression of selected candidates was validated using qRT-PCR. We identified 63 miRNAs upregulated in mCRPC versus localized PCa, while only four were downregulated. Pearson's correlation analysis revealed two highly correlated groups: one consisting of miR-141, miR375 and miR-200c and the other including miR151-3p, miR423-3p, miR-126, miR152 and miR-21. A third group, containing miR-16 and miR-205, showed less correlation. One miRNA from each group (miR-141, miR151-3p and miR-16) was used for logistic regression analysis and proved to increase the sensitivity of the prostate-specific antigen (PSA) test alone. While no miRNA alone differentiated localized PCa and mCRPC, combinations had greater sensitivity and specificity. The expression of these 10 candidates was assayed for association with clinical parameters of disease progression through the cBio portal. Our results demonstrate that plasma levels of selected miRNAs are potential biomarkers to differentiate localized PCa and mCRPC.


Assuntos
Biomarcadores Tumorais/sangue , Regulação Neoplásica da Expressão Gênica , MicroRNAs/sangue , Neoplasias da Próstata/sangue , RNA Neoplásico/sangue , Regulação para Cima , Idoso , Idoso de 80 Anos ou mais , Castração , Feminino , Humanos , Calicreínas/sangue , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/cirurgia , Sensibilidade e Especificidade
7.
Mol Cancer ; 11: 52, 2012 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-22867098

RESUMO

Early prostate cancer (PCa) is generally treatable and associated with good prognosis. After a variable time, PCa evolves into a highly metastatic and treatment-refractory disease: castration-resistant PCa (CRPC). Currently, few prognostic factors are available to predict the emergence of CRPC, and no curative option is available. Epigenetic gene regulation has been shown to trigger PCa metastasis and androgen-independence. Most epigenetic studies have focused on DNA and histone methyltransferases. While DNA methylation leads to gene silencing, histone methylation can trigger gene activation or inactivation, depending on the target amino acid residues and the extent of methylation (me1, me2, or me3). Interestingly, some histone modifiers are essential for PCa tumor-initiating cell (TIC) self-renewal. TICs are considered the seeds responsible for metastatic spreading and androgen-independence. Histone Lysine Demethylases (KDMs) are a novel class of epigenetic enzymes which can remove both repressive and activating histone marks. KDMs are currently grouped into 7 major classes, each one targeting a specific methylation site. Since their discovery, KDM expression has been found to be deregulated in several neoplasms. In PCa, KDMs may act as either tumor suppressors or oncogenes, depending on their gene regulatory function. For example, KDM1A and KDM4C are essential for PCa androgen-dependent proliferation, while PHF8 is involved in PCa migration and invasion. Interestingly, the possibility of pharmacologically targeting KDMs has been demonstrated. In the present paper, we summarize the emerging role of KDMs in regulating the metastatic potential and androgen-dependence of PCa. In addition, we speculate on the possible interaction between KDMs and other epigenetic effectors relevant for PCa TICs. Finally, we explore the role of KDMs as novel prognostic factors and therapeutic targets. We believe that studies on histone demethylation may add a novel perspective in our efforts to prevent and cure advanced PCa.


Assuntos
Histona Desmetilases/metabolismo , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/genética , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Histona Desmetilases/antagonistas & inibidores , Histona Desmetilases/genética , Humanos , Masculino , Prognóstico , Neoplasias da Próstata/tratamento farmacológico
8.
Blood ; 115(1): 38-46, 2010 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-19861679

RESUMO

The Additional sex combs like 1 (Asxl1) gene is 1 of 3 mammalian homologs of the Additional sex combs (Asx) gene of Drosophila. Asx is unusual because it is required to maintain both activation and silencing of Hox genes in flies and mice. Asxl proteins are characterized by an amino terminal homology domain, by interaction domains for nuclear receptors, and by a C-terminal plant homeodomain protein-protein interaction domain. A recent study of patients with myelodysplastic syndrome (MDS) and chronic myelomonocytic leukemia (CMML) revealed a high incidence of truncation mutations that would delete the PHD domain of ASXL1. Here, we show that Asxl1 is expressed in all hematopoietic cell fractions analyzed. Asxl1 knockout mice exhibit defects in frequency of differentiation of lymphoid and myeloid progenitors, but not in multipotent progenitors. We do not detect effects on hematopoietic stem cells, or in peripheral blood. Notably, we do not detect severe myelodysplastic phenotypes or leukemia in this loss-of-function model. We conclude that Asxl1 is needed for normal hematopoiesis. The mild phenotypes observed may be because other Asxl genes have redundant function with Asxl1, or alternatively, MDS or oncogenic phenotypes may result from gain-of-function Asxl mutations caused by genomic amplification, gene fusion, or truncation of Asxl1.


Assuntos
Hematopoese/genética , Leucemia/genética , Mutação/genética , Síndromes Mielodisplásicas/genética , Proteínas Repressoras/genética , Animais , Linfócitos B/citologia , Linfócitos B/metabolismo , Contagem de Células , Linhagem da Célula , Células Cultivadas , Citometria de Fluxo , Marcação de Genes , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Camundongos Mutantes , Células Mieloides/patologia , Esplenomegalia/patologia , Linfócitos T/citologia , Linfócitos T/metabolismo , Timo/citologia
9.
J Immunol ; 184(6): 2805-13, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20154203

RESUMO

Although several groups have investigated the role of SHIP in macrophage (M) development and function, SHIP's contribution to the generation, maturation, and innate immune activation of dendritic cells (DCs) is poorly understood. We show herein that SHIP negatively regulates the generation of DCs from bone marrow precursors in vitro and in vivo, as illustrated by the enhanced expansion of DCs from SHIP(-/-) GM-CSF cultures, as well as increased numbers of DCs in the spleens of SHIP-deficient mice. Interestingly, however, these SHIP(-/-) DCs display a relatively immature phenotype and secrete substantially lower levels of IL-12 after TLR ligand stimulation than wild type DCs. This, in turn, leads to a dramatically reduced stimulation of Ag-specific T cell proliferation and Th1 cell responses in vitro and in vivo. This immature phenotype of SHIP(-/-) DCs could be reversed with the PI3K inhibitors LY294002 and wortmannin, suggesting that SHIP promotes DC maturation by reducing the levels of the PI3K second messenger phosphatidylinositol-3,4,5-trisphosphate. These results are consistent with SHIP being a negative regulator of GM-CSF-derived DC generation but a positive regulator of GM-CSF-derived DC maturation and function.


Assuntos
Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Monoéster Fosfórico Hidrolases/fisiologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Diferenciação Celular/genética , Proliferação de Células , Células Cultivadas , Células Dendríticas/citologia , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Inibidores do Crescimento/deficiência , Inibidores do Crescimento/genética , Inibidores do Crescimento/fisiologia , Imunidade Inata/genética , Inositol Polifosfato 5-Fosfatases , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Monoéster Fosfórico Hidrolases/deficiência , Monoéster Fosfórico Hidrolases/genética , Células-Tronco/citologia , Células-Tronco/imunologia , Células-Tronco/metabolismo , Receptores Toll-Like/fisiologia , Regulação para Cima/genética , Regulação para Cima/imunologia
10.
Nat Med ; 8(9): 943-9, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12161749

RESUMO

The hematopoietic-restricted protein Src homology 2-containing inositol-5-phosphatase (SHIP) blunts phosphatidylinositol-3-kinase-initiated signaling by dephosphorylating its major substrate, phosphatidylinositol-3,4,5-trisphosphate. As SHIP(-/-) mice contain increased numbers of osteoclast precursors, that is, macrophages, we examined bones from these animals and found that osteoclast number is increased two-fold. This increased number is due to the prolonged life span of these cells and to hypersensitivity of precursors to macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor-kappa B ligand (RANKL). Similar to pagetic osteoclasts, SHIP(-/-) osteoclasts are enlarged, containing upwards of 100 nuclei, and exhibit enhanced resorptive activity. Moreover, as in Paget disease, serum levels of interleukin-6 are markedly increased in SHIP(-/-) mice. Consistent with accelerated resorptive activity, 3D trabecular volume fraction, trabecular thickness, number and connectivity density of SHIP(-/-) long bones are reduced, resulting in a 22% loss of bone-mineral density and a 49% decrease in fracture energy. Thus, SHIP negatively regulates osteoclast formation and function and the absence of this enzyme results in severe osteoporosis.


Assuntos
Osteoclastos/patologia , Osteoporose/patologia , Osteoporose/fisiopatologia , Monoéster Fosfórico Hidrolases/deficiência , Animais , Apoptose/genética , Densidade Óssea , Reabsorção Óssea/genética , Proteínas de Transporte/farmacologia , Contagem de Células , Células Cultivadas , Interleucina-6/sangue , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/efeitos dos fármacos , Glicoproteínas de Membrana/farmacologia , Camundongos , Camundongos Mutantes , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Monoéster Fosfórico Hidrolases/genética , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B
11.
BMC Dev Biol ; 8: 81, 2008 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-18778483

RESUMO

BACKGROUND: The full-length mammalian homologs of groucho, Tle1, 2, 3, and 4, act as transcriptional corepressors and are recruited by transcription factors containing an eh1 or WRPW/Y domain. Many transcription factors critical to pancreas development contain a Gro/TLE interaction domain and several have been shown to require Gro/TLE interactions for proper function during neuronal development. However, a detailed analysis of the expression patterns of the Gro/TLE proteins in pancreas development has not been performed. Moreover, little is known about the ability of Gro/TLE proteins to interact with transcription factors in the pancreas. RESULTS: We describe the expression of Gro/TLE family members, and of 34 different transcription factors that contain a Gro/TLE interaction motif, in the pancreas utilizing nine SAGE libraries created from the developing and adult pancreas, as well as the GenePaint database. Next, we show the dynamic expression of Tle1, 2, 3, 4, 5 and 6 during pancreas development by qRT-PCR. To further define the cell-type specificity of the expression of these proteins we use immunofluorescence to co-localize them with Pdx1 at embryonic day 12.5 (E12.5), Ngn3 at E14.5, Pdx1, Nkx2-2, Insulin, Glucagon, Pancreatic polypeptide and Somatostatin at E18.5, as well as Insulin and Glucagon in the adult. We then show that Tle2 can interact with Nkx2-2, Hes1, Arx, and Nkx6-1 which are all critical factors in pancreas development. Finally, we demonstrate that Tle2 modulates the repressive abilities of Arx in a beta-cell line. CONCLUSION: Although Tle1, 2, 3, and 4 show overlapping expression in pancreatic progenitors and in the adult islet, the expression of these factors is restricted to different cell types during endocrine cell maturation. Of note, Tle2 and Tle3 are co-expressed with Gro/TLE interaction domain containing transcription factors that are essential for endocrine pancreas development. We further demonstrate that Tle2 can interact with several of these factors and that Tle2 modulate Arx's repressive activity. Taken together our studies suggest that Gro/TLE proteins play a role in the repression of target genes during endocrine cell specification.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Pâncreas/embriologia , Pâncreas/metabolismo , Proteínas Repressoras/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Linhagem Celular , Proteínas Correpressoras , Feminino , Proteína Homeobox Nkx-2.2 , Masculino , Camundongos , Camundongos Endogâmicos ICR , Proteínas/genética , Proteínas/metabolismo , Ratos , Proteínas Repressoras/biossíntese
12.
Mol Cell Biol ; 25(24): 10916-29, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16314515

RESUMO

Groucho (Gro)/TLE transcriptional corepressors are involved in a variety of developmental mechanisms, including neuronal differentiation. They contain a conserved C-terminal WD40 repeat domain that mediates interactions with several DNA-binding proteins. In particular, Gro/TLE1 interacts with forkhead transcription factor brain factor 1 (BF-1; also termed FoxG1). BF-1 is an essential regulator of neuronal differentiation during cerebral cortex development and represses transcription together with Gro/TLE1. Gro/TLE-related gene product 6 (Grg6) shares with Gro/TLEs a conserved WD40 repeat domain but is more distantly related at its N-terminal half. We demonstrate that Grg6 is expressed in cortical neural progenitor cells and interacts with BF-1. In contrast to Gro/TLE1, however, Grg6 does not promote, but rather suppresses, BF-1-mediated transcriptional repression. Consistent with these observations, Grg6 interferes with the binding of Gro/TLE1 to BF-1 and does not repress transcription when targeted to DNA. Moreover, coexpression of Grg6 and BF-1 in cortical progenitor cells leads to a decrease in the number of proliferating cells and increased neuronal differentiation. Conversely, Grg6 knockdown by RNA interference causes decreased neurogenesis. These results identify a new role for Grg6 in cortical neuron development and establish a functional link between Grg6 and BF-1.


Assuntos
Córtex Cerebral/citologia , Fatores de Transcrição Forkhead/antagonistas & inibidores , Proteínas do Tecido Nervoso/antagonistas & inibidores , Neurônios/citologia , Proteínas Repressoras/metabolismo , Sequência de Aminoácidos , Animais , Diferenciação Celular/genética , Proteínas Correpressoras , Regulação para Baixo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Dados de Sequência Molecular , Neurônios/metabolismo , Interferência de RNA , Proteínas Repressoras/genética , Transcrição Gênica
13.
Exp Hematol ; 35(4): 627-39, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17379073

RESUMO

OBJECTIVE: Dendritic cells (DC) play a critical role in establishment and maintenance of central and peripheral tolerance. Despite intensive research, our knowledge of the molecular mechanisms regulating DC development and function is limited, thus hindering our ability to generate appropriate DC populations for manipulating immune tolerance. We utilized mice deficient in the SH2-containing inositol-5-phosphatase (SHIP) to examine the role of cytokine signaling in DC development and function. METHODS: We analyzed the phenotype of both primary and bone marrow (BM)-derived DC (BMDC) using flow cytometry. In addition, cytokine production was measured using cytometric bead arrays and the ability of DC to induce allogeneic T-cell proliferation was assessed using thymidine incorporation assays. RESULTS: We demonstrated that spleen DC isolated from SHIP-deficient mice are increased in number and have an altered phenotype. In vitro analyses revealed that SHIP-deficient BM cells give rise to a higher frequency of myeloid, but not plasmacytoid, DC due to both an increased progenitor frequency and enhanced cytokine sensitivity. The BMDC exhibit an altered phenotype that correlates with a reduced capacity to induce allogeneic T-cell proliferation. Addition of interleukin-6 to WT BM cultures during DC differentiation partially induces a KO phenotype. CONCLUSION: These studies suggest that myeloid and plasmacytoid DC progenitors are differentially sensitive to signaling pathways in which SHIP is involved. Moreover, they suggest that interleukin-6 may have an important role in regulating the phenotype and function of myeloid DC.


Assuntos
Células Dendríticas/imunologia , Monoéster Fosfórico Hidrolases/imunologia , Animais , Citometria de Fluxo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Inositol Polifosfato 5-Fosfatases , Interleucina-4/administração & dosagem , Camundongos , Camundongos Knockout
14.
Exp Hematol ; 35(8): 1293-302, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17562354

RESUMO

OBJECTIVE: We previously demonstrated that c-kit expression decreases during murine embryonic stem cell (ESC) differentiation induced by leukemia inhibitory factor removal. In this study, we addressed the possibility that c-kit is a marker of undifferentiated murine ESC and, moreover, that it plays a role in maintaining the undifferentiated state of these cells. MATERIALS AND METHODS: c-kit expression was analyzed under various differentiation conditions by flow cytometry and quantitative reverse transcription polymerase chain reaction. ESC were then sorted on the basis of c-kit expression and functionality was investigated using embryoid body and colony-forming cell assays. Imatinib (Gleevec) and ACK2 were used to block, and stem cell factor was used to stimulate, c-kit activity. RESULTS: c-kit expression decreased in two murine ESC lines under various differentiation conditions. Sorting of ESC populations on the basis of c-kit expression revealed significant differences in the functional capacities and gene expression profiles of the sorted populations. The inhibition studies revealed an important role for tyrosine kinase activity in maintaining ESC viability and differentiation capacity, at least in part by preventing apoptosis and enhancing cell cycle progression. However, activation of c-kit alone is not sufficient for maintaining undifferentiated ESC. CONCLUSION: The results suggest that c-kit may represent a useful marker for monitoring ESC functionality. Moreover, tyrosine kinase signaling plays an important role in maintaining undifferentiated ESC. This work provides valuable insights into the complex signaling pathways that synergize to maintain the undifferentiated state of murine ESC.


Assuntos
Células-Tronco Embrionárias/fisiologia , Proteínas Tirosina Quinases/fisiologia , Animais , Técnicas de Cultura de Células , Ciclo Celular/fisiologia , Diferenciação Celular , Ensaio de Unidades Formadoras de Colônias , Primers do DNA , Células-Tronco Embrionárias/citologia , Citometria de Fluxo , Camundongos , Proteínas Proto-Oncogênicas c-kit/genética , Transdução de Sinais , Transcrição Gênica
15.
PLoS Comput Biol ; 2(11): e158, 2006 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-17121458

RESUMO

Stem cell differentiation involves critical changes in gene expression. Identification of these should provide endpoints useful for optimizing stem cell propagation as well as potential clues about mechanisms governing stem cell maintenance. Here we describe the results of a new meta-analysis methodology applied to multiple gene expression datasets from three mouse embryonic stem cell (ESC) lines obtained at specific time points during the course of their differentiation into various lineages. We developed methods to identify genes with expression changes that correlated with the altered frequency of functionally defined, undifferentiated ESC in culture. In each dataset, we computed a novel statistical confidence measure for every gene which captured the certainty that a particular gene exhibited an expression pattern of interest within that dataset. This permitted a joint analysis of the datasets, despite the different experimental designs. Using a ranking scheme that favored genes exhibiting patterns of interest, we focused on the top 88 genes whose expression was consistently changed when ESC were induced to differentiate. Seven of these (103728_at, 8430410A17Rik, Klf2, Nr0b1, Sox2, Tcl1, and Zfp42) showed a rapid decrease in expression concurrent with a decrease in frequency of undifferentiated cells and remained predictive when evaluated in additional maintenance and differentiating protocols. Through a novel meta-analysis, this study identifies a small set of genes whose expression is useful for identifying changes in stem cell frequencies in cultures of mouse ESC. The methods and findings have broader applicability to understanding the regulation of self-renewal of other stem cell types.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteoma/metabolismo , Adaptação Fisiológica/fisiologia , Animais , Cinética , Camundongos
16.
Cancer Res ; 65(7): 2947-55, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15805298

RESUMO

Increasing evidence suggests that altered immune function accompanies, and indeed may facilitate, cancer progression. In this study, we sought to determine the nature of, and cellular mechanisms underlying, changes in immune status during disease progression in a transgenic mouse model of prostate dysplasia. Immune cells in the tumor microenvironment, as well as in the secondary lymphoid tissues, displayed altered phenotypes. Although evidence of antitumor immunity was detected, there was a paradoxical decrease in the ability of T cells to proliferate in vitro at later stages of disease progression. Detailed analysis of the draining lumbar lymph nodes revealed an increased frequency and number of CD4(+)CD25(+) T cells and an enhanced production of inhibitory cytokines, which correlated with impaired T-cell function. Functional studies confirmed a role for CD4(+)CD25(+) regulatory T cells in suppressing T-cell proliferation as well as regulating the growth of transplanted prostate tumor cells. In addition, our studies show for the first time that anti-CD25 antibody treatment reduces, but does not prevent, tumor growth in a transgenic mouse model of prostate dysplasia. Taken together, this work provides compelling evidence that prostate tumor progression is accompanied by altered immune function and, moreover, that regulatory T cells play an important role in this process. These studies thus provide the impetus for development of specific and effective strategies to deplete regulatory T cells, or suppress their function, as an alternative or adjunct strategy for reducing tumor growth.


Assuntos
Próstata/imunologia , Próstata/patologia , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Modelos Animais de Doenças , Progressão da Doença , Feminino , Tecido Linfoide/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Interleucina-2/imunologia , Linfócitos T/imunologia
17.
Gene Expr Patterns ; 6(3): 310-24, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16378759

RESUMO

To identify genes and pathways not previously implicated in the mesenchymal-epithelial (M/E) interactions that are critical for normal mouse prostate development, we constructed six serial analysis of gene expression (SAGE) libraries. Bioinformatic analyses revealed expression of various members of numerous signalling pathways and the differential expression of several members of the wingless-related MMTV integration site (Wnt) signalling pathway. This pathway has not been previously implicated in prostate development thus expression of selected Wnt pathway members in the developing prostate was confirmed by RT-qPCR. Of particular interest, an antagonist of the Wnt pathway, secreted frizzled related protein 2 (Sfrp2), was highly expressed in the early prostate libraries and down regulated at later developmental stages. The expression levels of four Wnt ligands reported to interact with Sfrp2 were, therefore, examined by RT-qPCR. We found that only Wnt4 transcripts were detectable in the developing prostate. Expression of Sfrp2 was validated using RT-qPCR and localization of Sfrp2 transcripts and protein was carried out using in situ hybridization and immunofluorescence, respectively. These studies provide the first evidence that Wnt pathway members are expressed in the developing prostate. Functional analyses are now required to establish the biological significance of this observation.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Biblioteca Gênica , Próstata/embriologia , Transdução de Sinais , Proteínas Wnt/metabolismo , Animais , Biologia Computacional , Imunofluorescência , Hibridização In Situ , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Próstata/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Front Biosci (Landmark Ed) ; 21(2): 430-46, 2016 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-26709784

RESUMO

Biomarkers are molecules or features which can provide clinically-relevant information about a particular disease state, thus providing useful tools for oncologists. Recently, a number of studies have demonstrated that DNA methylation holds great promise as a novel source of cancer biomarkers. Although promoter regions have been the focus of most investigations thus far, mounting evidence demonstrates that enhancer sequences also undergo extensive differential methylation in cancer cells. Moreover, enhancer methylation correlates with target gene expression better than promoter methylation, providing unexplored strategies for biomarker development. Here, we review important considerations associated with the clinical analysis of DNA methylation at distal regulatory regions. Notably, we highlight emerging literature addressing the methylation status of enhancers in development and cancer, and subsequently discuss how enhancer methylation can be exploited to guide disease management. While acknowledging current limitations, we propose that the methylation state of enhancer regions has the potential to headline the next generation of epigenetic biomarkers.


Assuntos
Biomarcadores Tumorais , Metilação de DNA , Elementos Facilitadores Genéticos , Epigênese Genética , Humanos , Neoplasias/diagnóstico , Neoplasias/terapia
19.
Clin Epigenetics ; 8: 16, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26877821

RESUMO

BACKGROUND: While localized prostate cancer (PCa) can be effectively cured, metastatic disease inevitably progresses to a lethal state called castration-resistant prostate cancer (CRPC). Emerging evidence suggests that aberrant epigenetic repression by the polycomb group (PcG) complexes fuels PCa progression, providing novel therapeutic opportunities. RESULTS: In the search for potential epigenetic drivers of CRPC, we analyzed the molecular profile of PcG members in patient-derived xenografts and clinical samples. Overall, our results identify the PcG protein and methyl-lysine reader CBX2 as a potential therapeutic target in advanced PCa. We report that CBX2 was recurrently up-regulated in metastatic CRPC and that elevated CBX2 expression was correlated with poor clinical outcome in PCa cohorts. Furthermore, CBX2 depletion abrogated cell viability and induced caspase 3-mediated apoptosis in metastatic PCa cell lines. Mechanistically explaining this phenotype, microarray analysis in CBX2-depleted cells revealed that CBX2 controls the expression of many key regulators of cell proliferation and metastasis. CONCLUSIONS: Taken together, this study provides the first evidence that CBX2 inhibition induces cancer cell death, positioning CBX2 as an attractive drug target in lethal CRPC.


Assuntos
Complexo Repressor Polycomb 1/efeitos dos fármacos , Neoplasias da Próstata/tratamento farmacológico , Apoptose , Caspase 3/metabolismo , Epigênese Genética/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Complexo Repressor Polycomb 1/fisiologia , Neoplasias da Próstata/genética , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/genética , Células Tumorais Cultivadas , Regulação para Cima
20.
Mol Oncol ; 10(5): 693-703, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26809501

RESUMO

Metastasis is the primary cause of death in prostate cancer (PCa) patients. Small nucleolar RNAs (snoRNAs) have long been considered "housekeeping" genes with no relevance for cancer biology. Emerging evidence has challenged this assumption, suggesting that snoRNA expression is frequently modulated during cancer progression. Despite this, no study has systematically addressed the prognostic and functional significance of snoRNAs in PCa. We performed RNA Sequencing on paired metastatic/non-metastatic PCa xenografts derived from clinical specimens. The clinical significance of differentially expressed snoRNAs was further investigated in two independent primary PCa cohorts (131 and 43 patients, respectively). The snoRNA demonstrating the strongest association with clinical outcome was quantified in PCa patient-derived serum samples and its functional relevance was investigated in PCa cells via gene expression profiling, pathway analysis and gene silencing. Our comparison revealed 21 differentially expressed snoRNAs in the metastatic vs. non-metastatic xenografts. Of those, 12 were represented in clinical databases and were further analyzed. SNORA55 emerged as a predictor of shorter relapse-free survival (results confirmed in two independent databases). SNORA55 was reproducibly detectable in serum samples from PCa patients. SNORA55 silencing in PCa cell lines significantly inhibited cell proliferation and migration. Pathway analysis revealed that SNORA55 expression is significantly associated with growth factor signaling and pro-inflammatory cytokine expression in PCa. Our results demonstrate that SNORA55 up-regulation predicts PCa progression and that silencing this non-coding gene affects PCa cell proliferation and metastatic potential, thus positioning it as both a novel biomarker and therapeutic target.


Assuntos
Regulação Neoplásica da Expressão Gênica , Próstata/patologia , Neoplasias da Próstata/genética , RNA Nucleolar Pequeno/genética , Transcriptoma , Idoso , Linhagem Celular Tumoral , Progressão da Doença , Perfilação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Próstata/metabolismo , Neoplasias da Próstata/sangue , Neoplasias da Próstata/patologia , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa