Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 12(1): 7308, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34911936

RESUMO

Androgen receptor (AR) in prostate cancer (PCa) can drive transcriptional repression of multiple genes including MYC, and supraphysiological androgen is effective in some patients. Here, we show that this repression is independent of AR chromatin binding and driven by coactivator redistribution, and through chromatin conformation capture methods show disruption of the interaction between the MYC super-enhancer within the PCAT1 gene and the MYC promoter. Conversely, androgen deprivation in vitro and in vivo increases MYC expression. In parallel, global AR activity is suppressed by MYC overexpression, consistent with coactivator redistribution. These suppressive effects of AR and MYC are mitigated at shared AR/MYC binding sites, which also have markedly higher levels of H3K27 acetylation, indicating enrichment for functional enhancers. These findings demonstrate an intricate balance between AR and MYC, and indicate that increased MYC in response to androgen deprivation contributes to castration-resistant PCa, while decreased MYC may contribute to responses to supraphysiological androgen therapy.


Assuntos
Elementos Facilitadores Genéticos , Proteína Oncogênica p55(v-myc)/genética , Receptores Androgênicos/genética , Androgênios/metabolismo , Linhagem Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Proteína Oncogênica p55(v-myc)/metabolismo , Regiões Promotoras Genéticas , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/metabolismo , Transdução de Sinais
2.
Nat Rev Cancer ; 21(9): 579-591, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34188192

RESUMO

The transcription factor and oncoprotein MYC is a potent driver of many human cancers and can regulate numerous biological activities that contribute to tumorigenesis. How a single transcription factor can regulate such a diverse set of biological programmes is central to the understanding of MYC function in cancer. In this Perspective, we highlight how multiple proteins that interact with MYC enable MYC to regulate several central control points of gene transcription. These include promoter binding, epigenetic modifications, initiation, elongation and post-transcriptional processes. Evidence shows that a combination of multiple protein interactions enables MYC to function as a potent oncoprotein, working together in a 'coalition model', as presented here. Moreover, as MYC depends on its protein interactome for function, we discuss recent research that emphasizes an unprecedented opportunity to target protein interactors to directly impede MYC oncogenesis.


Assuntos
Neoplasias/metabolismo , Proteína Oncogênica p55(v-myc)/metabolismo , Transcrição Gênica , Animais , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/genética , Proteína Oncogênica p55(v-myc)/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
3.
Nat Commun ; 10(1): 5494, 2019 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-31792211

RESUMO

LRIG1 has been reported to be a tumor suppressor in gastrointestinal tract and epidermis. However, little is known about the expression, regulation and biological functions of LRIG1 in prostate cancer (PCa). We find that LRIG1 is overexpressed in PCa, but its expression correlates with better patient survival. Functional studies reveal strong tumor-suppressive functions of LRIG1 in both AR+ and AR- xenograft models, and transgenic expression of LRIG1 inhibits tumor development in Hi-Myc and TRAMP models. LRIG1 also inhibits castration-resistant PCa and exhibits therapeutic efficacy in pre-established tumors. We further show that 1) AR directly transactivates LRIG1 through binding to several AR-binding sites in LRIG1 locus, and 2) LRIG1 dampens ERBB expression in a cell type-dependent manner and inhibits ERBB2-driven tumor growth. Collectively, our study indicates that LRIG1 represents a pleiotropic AR-regulated feedback tumor suppressor that functions to restrict oncogenic signaling from AR, Myc, ERBBs, and, likely, other oncogenic drivers.


Assuntos
Glicoproteínas de Membrana/metabolismo , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Glicoproteínas de Membrana/genética , Camundongos Endogâmicos NOD , Camundongos SCID , Proteína Oncogênica p55(v-myc)/genética , Proteína Oncogênica p55(v-myc)/metabolismo , Neoplasias da Próstata/genética , Ligação Proteica , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptores Androgênicos/genética , Transdução de Sinais , Proteínas Supressoras de Tumor/genética
4.
Nat Commun ; 10(1): 5498, 2019 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-31792212

RESUMO

Interactions between thymic epithelial cells (TEC) and developing thymocytes are essential for T cell development, but molecular insights on TEC and thymus homeostasis are still lacking. Here we identify distinct transcriptional programs of TEC that account for their age-specific properties, including proliferation rates, engraftability and function. Further analyses identify Myc as a regulator of fetal thymus development to support the rapid increase of thymus size during fetal life. Enforced Myc expression in TEC induces the prolonged maintenance of a fetal-specific transcriptional program, which in turn extends the growth phase of the thymus and enhances thymic output; meanwhile, inducible expression of Myc in adult TEC similarly promotes thymic growth. Mechanistically, this Myc function is associated with enhanced ribosomal biogenesis in TEC. Our study thus identifies age-specific transcriptional programs in TEC, and establishes that Myc controls thymus size.


Assuntos
Células Epiteliais/metabolismo , Proteína Oncogênica p55(v-myc)/metabolismo , Timo/embriologia , Transcrição Gênica , Animais , Células Epiteliais/citologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Proteína Oncogênica p55(v-myc)/genética , Tamanho do Órgão , Organogênese , Timo/metabolismo
5.
Clin Sci (Lond) ; 133(3): 409-423, 2019 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-29122967

RESUMO

Progression of non-alcoholic fatty liver disease (NAFLD) in the context of metabolic syndrome (MetS) is only partially explored due to the lack of preclinical models. In order to study the alterations in hepatic metabolism that accompany this condition, we developed a model of MetS accompanied by the onset of steatohepatitis (NASH) by challenging golden hamsters with a high-fat diet low in vitamin E and selenium (HFD), since combined deficiency results in hepatic necroinflammation in rodents. Metabolomics and transcriptomics integrated analyses of livers revealed an unexpected accumulation of hepatic S-Adenosylmethionine (SAM) when compared with healthy livers likely due to diminished methylation reactions and repression of GNMT. SAM plays a key role in the maintenance of cellular homeostasis and cell cycle control. In agreement, analysis of over-represented transcription factors revealed a central role of c-myc and c-Jun pathways accompanied by negative correlations between SAM concentration, MYC expression and AMPK phosphorylation. These findings point to a drift of cell cycle control toward senescence in livers of HFD animals, which could explain the onset of NASH in this model. In contrast, hamsters with NAFLD induced by a conventional high-fat diet did not show SAM accumulation, suggesting a key role of selenium and vitamin E in SAM homeostasis. In conclusion, our results suggest that progression of NAFLD in the context of MetS can take place even in a situation of hepatic SAM excess and that selenium and vitamin E status might be considered in current therapies against NASH based on SAM supplementation.


Assuntos
Fígado/metabolismo , Síndrome Metabólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , S-Adenosilmetionina/metabolismo , Selênio/deficiência , Vitamina E/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Animais , Cricetinae , Dieta Hiperlipídica/efeitos adversos , Progressão da Doença , Humanos , Masculino , Mesocricetus , Síndrome Metabólica/genética , Hepatopatia Gordurosa não Alcoólica/genética , Proteína Oncogênica p55(v-myc)/genética , Proteína Oncogênica p55(v-myc)/metabolismo , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Selênio/análise , Vitamina E/análise
6.
J Biol Chem ; 293(48): 18757-18769, 2018 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-30404920

RESUMO

This paper is in recognition of the 100th birthday of Dr. Herbert Tabor, a true pioneer in the polyamine field for over 70 years, who served as the editor-in-chief of the Journal of Biological Chemistry from 1971 to 2010. We review current knowledge of MYC proteins (c-MYC, MYCN, and MYCL) and focus on ornithine decarboxylase 1 (ODC1), an important bona fide gene target of MYC, which encodes the sentinel, rate-limiting enzyme in polyamine biosynthesis. Although notable advances have been made in designing inhibitors against the "undruggable" MYCs, their downstream targets and pathways are currently the main avenue for therapeutic anticancer interventions. To this end, the MYC-ODC axis presents an attractive target for managing cancers such as neuroblastoma, a pediatric malignancy in which MYCN gene amplification correlates with poor prognosis and high-risk disease. ODC and polyamine levels are often up-regulated and contribute to tumor hyperproliferation, especially of MYC-driven cancers. We therefore had proposed to repurpose α-difluoromethylornithine (DFMO), an FDA-approved, orally available ODC inhibitor, for management of neuroblastoma, and this intervention is now being pursued in several clinical trials. We discuss the regulation of ODC and polyamines, which besides their well-known interactions with DNA and tRNA/rRNA, are involved in regulating RNA transcription and translation, ribosome function, proteasomal degradation, the circadian clock, and immunity, events that are also controlled by MYC proteins.


Assuntos
Proteína Oncogênica p55(v-myc)/metabolismo , Ornitina Descarboxilase/metabolismo , Poliaminas/metabolismo , Animais , Humanos , Proteína Oncogênica p55(v-myc)/genética , Ornitina Descarboxilase/genética
7.
Cell Death Dis ; 9(3): 315, 2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29472532

RESUMO

The bromodomain and extra-terminal domain inhibitors (BETi) are promising epigenetic drugs for the treatment of various cancers through suppression of oncogenic transcription factors. However, only a subset of colorectal cancer (CRC) cells response to BETi. We investigate additional agents that could be combined with BETi to overcome this obstacle. JQ1-resistant CRC cells were used for screening of the effective combination therapies with JQ1. RNA-seq was performed to explore the mechanism of synergistic effect. The efficacy of combinational treatment was tested in the CRC cell line- and patient-derived xenograft (PDX) models. In BETi-sensitive CRC cells, JQ1 also impaired tumor angiogenesis through the c-myc/miR-17-92/CTGF+THBS1 axis. CTGF knockdown moderately counteracted anti-angiogenic effect of JQ1 and led to partially attenuated tumor regression. JQ1 decreased c-myc expression and NF-κB activity in BETi-sensitive CRC cells but not in resistant cells. Bortezomib synergistically sensitized BETi-resistant cells to the JQ1 treatment, and JQ1+Bortezomib induced G2/M arrest in CRC cells. Mechanistically, inhibition of NF-κB by Bortezomib or NF-κB inhibitor or IKK1/2 siRNA all rendered BETi-resistant cells more sensitive to BETi by synergistic repression of c-myc, which in turn induces GADD45s' expression, and by synergistic repression of FOXM1 which in turn inhibit G2/M checkpoint genes' expression. Activation of NF-κB by IκBα siRNA induced resistance to JQ1 in BETi-sensitive CRC cells. Last, JQ1+Bortezomib inhibited tumor growth and angiogenesis in CRC cell line xenograft model and four PDX models. Our results indicate that anti-angiogenic effect of JQ1 plays a vital role in therapeutic effect of JQ1 in CRC, and provide a rationale for combined inhibition of BET proteins and NF-κB as a potential therapy for CRC.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias Colorretais/tratamento farmacológico , Proteína Forkhead Box M1/genética , NF-kappa B/metabolismo , Proteínas Nucleares/antagonistas & inibidores , Proteína Oncogênica p55(v-myc)/genética , Proteínas Proto-Oncogênicas c-myc/genética , Animais , Azepinas/administração & dosagem , Bortezomib/administração & dosagem , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/fisiopatologia , Sinergismo Farmacológico , Feminino , Proteína Forkhead Box M1/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Nus , NF-kappa B/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína Oncogênica p55(v-myc)/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Triazóis/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
8.
J Agric Food Chem ; 63(9): 2432-41, 2015 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-25686711

RESUMO

Breast cancer stem cells (BCSCs) constitute a small fraction of the primary tumor that can self-renew and become a drug-resistant cell population, thus limiting the treatment effects of chemotherapeutic drugs. The present study evaluated the cytotoxic effects of five phytochemicals including 6-gingerol (6-G), 6-shogaol (6-S), 5-hydroxy-3,6,7,8,3',4'-hexamethoxyflavone (5-HF), nobiletin (NOL), and pterostilbene (PTE) on MCF-7 breast cancer cells and BCSCs. The results showed that 6-G, 6-S, and PTE selectively killed BCSCs and had high sensitivity for BCSCs isolated from MCF-7 cells that expressed the surface antigen CD44(+)/CD24(-). 6-S and PTE induced cell necrosis phenomena such as membrane injury and bleb formation in BCSCs and inhibited mammosphere formation. In addition, 6-S and PTE increased the sensitivity of isolated BCSCs to chemotherapeutic drugs and significantly increased the anticancer activity of paclitaxel. Analysis of the underlying mechanism showed that 6-S and PTE decreased the expression of the surface antigen CD44 on BCSCs and promoted ß-catenin phosphorylation through the inhibition of hedgehog/Akt/GSK3ß signaling, thus decreasing the protein expression of downstream c-Myc and cyclin D1 and reducing BCSC stemness.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Catecóis/farmacologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Estilbenos/farmacologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/fisiopatologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Feminino , Humanos , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Proteína Oncogênica p55(v-myc)/genética , Proteína Oncogênica p55(v-myc)/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
Virology ; 476: 72-84, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25528440

RESUMO

Marek's disease virus (MDV) is a representative alpha herpes virus able to induce rapid-onset T-cell lymphoma in its natural host and regarded as an ideal model for the study of virus-induced tumorigenesis. Recent studies have shown that the mdv1-miR-M4-5p, a viral analog of cellular miR-155, is critical for MDV׳s oncogenicity. However, the precise mechanism whereby it was involved in MD lymphomagenesis remained unknown. We have presently identified the host mRNA targets of mdv1-miR-M4-5 and identified the latent TGF-ß binding protein 1 (LTBP1) as a critical target for it. We found that during MDV infection, down-regulation of LTBP1 expression by mdv1-miR-M4-5p led to a significant decrease of the secretion and activation of TGF-ß1, with suppression of TGF-ß signaling and a significant activation of expression of c-Myc, a well-known oncogene which is critical for virus-induced tumorigenesis. Our findings reveal a novel and important mechanism of how mdv1-miR-M4-5p potentially contributes to MDV-induced tumorigenesis.


Assuntos
Proteínas de Ligação a TGF-beta Latente/metabolismo , Doença de Marek/metabolismo , MicroRNAs/metabolismo , Proteína Oncogênica p55(v-myc)/metabolismo , RNA Viral/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Sequência de Bases , Galinhas , Regulação para Baixo , Regulação Viral da Expressão Gênica , Interações Hospedeiro-Patógeno , Proteínas de Ligação a TGF-beta Latente/genética , Doença de Marek/genética , Doença de Marek/virologia , MicroRNAs/genética , Dados de Sequência Molecular , Proteína Oncogênica p55(v-myc)/genética , RNA Viral/genética
10.
Nat Commun ; 5: 5057, 2014 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-25266931

RESUMO

The capacity of dendritic cells (DCs) to regulate tumour-specific adaptive immune responses depends on their proper differentiation and homing status. Whereas DC-associated tumour-promoting functions are linked to T-cell tolerance and formation of an inflammatory milieu, DC-mediated direct effects on tumour growth have remained unexplored. Here we show that deletion of DCs substantially delays progression of Myc-driven lymphomas. Lymphoma-exposed DCs upregulate immunomodulatory cytokines, growth factors and the CCAAT/enhancer-binding protein ß (C/EBPß). Moreover, Eµ-Myc lymphomas induce the preferential translation of the LAP/LAP* isoforms of C/EBPß. C/EBPß(-/-) DCs are unresponsive to lymphoma-associated cytokine changes and in contrast to wild-type DCs, they are unable to mediate enhanced Eµ-Myc lymphoma cell survival. Antigen-specific T-cell proliferation in lymphoma-bearing mice is impaired; however, this immune suppression is reverted by the DC-restricted deletion of C/EBPß. Thus, we show that C/EBPß-controlled DC functions are critical steps for the creation of a lymphoma growth-promoting and -immunosuppressive niche.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/imunologia , Células Dendríticas/imunologia , Linfoma de Células B/imunologia , Proteína Oncogênica p55(v-myc)/imunologia , Animais , Proteína beta Intensificadora de Ligação a CCAAT/genética , Diferenciação Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Células Dendríticas/citologia , Humanos , Linfoma de Células B/genética , Linfoma de Células B/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Proteína Oncogênica p55(v-myc)/genética
11.
EMBO Mol Med ; 6(12): 1525-41, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25253726

RESUMO

Deregulated expression of MYC is a driver of colorectal carcinogenesis, necessitating novel strategies to inhibit MYC function. The ubiquitin ligase HUWE1 (HECTH9, ARF-BP1, MULE) associates with both MYC and the MYC-associated protein MIZ1. We show here that HUWE1 is required for growth of colorectal cancer cells in culture and in orthotopic xenograft models. Using high-throughput screening, we identify small molecule inhibitors of HUWE1, which inhibit MYC-dependent transactivation in colorectal cancer cells, but not in stem and normal colon epithelial cells. Inhibition of HUWE1 stabilizes MIZ1. MIZ1 globally accumulates on MYC target genes and contributes to repression of MYC-activated target genes upon HUWE1 inhibition. Our data show that transcriptional activation by MYC in colon cancer cells requires the continuous degradation of MIZ1 and identify a novel principle that allows for inhibition of MYC function in tumor cells.


Assuntos
Neoplasias Colorretais/enzimologia , Proteína Oncogênica p55(v-myc)/antagonistas & inibidores , Proteína Oncogênica p55(v-myc)/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/fisiopatologia , Regulação Neoplásica da Expressão Gênica , Humanos , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos SCID , Proteína Oncogênica p55(v-myc)/genética , Ligação Proteica , Bibliotecas de Moléculas Pequenas/administração & dosagem , Ativação Transcricional , Proteínas Supressoras de Tumor , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
12.
Cell Res ; 24(11): 1284-5, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25104732

RESUMO

New findings bring to light a previously unappreciated mechanism involved in the regulation of the oncoprotein MYC. Interesting observations find that the long noncoding RNA (lncRNA) PVT1 is active in controlling levels of MYC through regulation of MYC protein stability.


Assuntos
Variações do Número de Cópias de DNA/genética , Amplificação de Genes/genética , Dosagem de Genes/genética , Genes myc/genética , Proteína Oncogênica p55(v-myc)/genética , RNA Longo não Codificante/genética , Animais , Humanos
13.
Nature ; 512(7512): 82-6, 2014 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-25043044

RESUMO

'Gain' of supernumerary copies of the 8q24.21 chromosomal region has been shown to be common in many human cancers and is associated with poor prognosis. The well-characterized myelocytomatosis (MYC) oncogene resides in the 8q24.21 region and is consistently co-gained with an adjacent 'gene desert' of approximately 2 megabases that contains the long non-coding RNA gene PVT1, the CCDC26 gene candidate and the GSDMC gene. Whether low copy-number gain of one or more of these genes drives neoplasia is not known. Here we use chromosome engineering in mice to show that a single extra copy of either the Myc gene or the region encompassing Pvt1, Ccdc26 and Gsdmc fails to advance cancer measurably, whereas a single supernumerary segment encompassing all four genes successfully promotes cancer. Gain of PVT1 long non-coding RNA expression was required for high MYC protein levels in 8q24-amplified human cancer cells. PVT1 RNA and MYC protein expression correlated in primary human tumours, and copy number of PVT1 was co-increased in more than 98% of MYC-copy-increase cancers. Ablation of PVT1 from MYC-driven colon cancer line HCT116 diminished its tumorigenic potency. As MYC protein has been refractory to small-molecule inhibition, the dependence of high MYC protein levels on PVT1 long non-coding RNA provides a much needed therapeutic target.


Assuntos
Variações do Número de Cópias de DNA/genética , Amplificação de Genes/genética , Dosagem de Genes/genética , Genes myc/genética , Proteína Oncogênica p55(v-myc)/genética , RNA Longo não Codificante/genética , Animais , Transformação Celular Neoplásica , Cromossomos Humanos Par 8/genética , Modelos Animais de Doenças , Células HCT116 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteína Oncogênica p55(v-myc)/metabolismo , Fenótipo
14.
Med Oncol ; 30(1): 465, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23377984

RESUMO

The copy number gain of genes in chromosomal region 8q21-24 has been demonstrated to be associated with genesis and progression of prostate cancer (PCa). The aim of this study was to identify novel and effective molecular markers in this chromosomal region for PCa. The differentially expressed genes in PCa specimens were screened by gene microarray analysis, which was validated by RT-QPCR analysis. Then, the DNA qPCR analysis was carried out to detect the copy number changes of these differentially expressed genes. Moreover, the clinical significance of candidate markers (MYC and E2F5) in PCa were further determined. E2F5 and MYC were identified as candidate markers in PCa tissues and PCa cell lines. The DNA qPCR revealed the significant copy number gains of E2F5 and MYC in PCa tissues but not in PCa cell lines. In addition, Western blot analysis and immunohistochemical staining both found the significant higher expression of E2F5 and MYC proteins in PCa tissues than those in adjacent benign specimens (all P < 0.01). Moreover, the overexpression of E2F5 protein was significantly associated with a high Gleason score (P < 0.01), an advanced clinical stage (P = 0.01), a positive metastasis (P < 0.01) and PSA Failure (P < 0.01). The overexpression of MYC was more frequently found in PCa tissues with positive metastasis (P = 0.02) and PSA failure (P = 0.02). Interestingly, there was a close correlation in the expression level of MYC in PCa tissues with that of E2F5 (r ( s ) = 0.5, P ≤ 0.001). Our data offers the convincing evidence that the copy number gains of E2F5 and MYC may play an important role in genesis and progression of PCa. Especially, E2F5 may be a novel potential candidate marker for malignant PCa.


Assuntos
Biomarcadores Tumorais/genética , Cromossomos Humanos Par 8/genética , Fator de Transcrição E2F5/genética , Dosagem de Genes , Proteínas Oncogênicas/genética , Neoplasias da Próstata/genética , Idoso , Idoso de 80 Anos ou mais , Western Blotting , Linhagem Celular Tumoral , Aberrações Cromossômicas , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Proteína Oncogênica p55(v-myc)/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Biochim Biophys Acta ; 1819(9-10): 1075-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22766037

RESUMO

Eukaryotes control nearly every cellular process in part by modulating the transcription of genes encoded by their nuclear genome. However, these cells are faced with the added complexity of possessing a second genome, within the mitochondria, which encodes critical components of several essential processes, including energy metabolism and macromolecule biosynthesis. As these cellular processes require gene products encoded by both genomes, cells have adopted strategies for linking mitochondrial gene expression to nuclear gene expression and other dynamic cellular events. Here we discuss examples of several mechanisms that have been identified, by which eukaryotic cells link extramitochondrial signals to dynamic alterations in mitochondrial transcription. This article is part of a Special Issue entitled: Mitochondrial Gene Expression.


Assuntos
Adaptação Biológica/genética , Mitocôndrias , Proteínas Nucleares/metabolismo , Transcrição Gênica , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , RNA Polimerases Dirigidas por DNA/genética , RNA Polimerases Dirigidas por DNA/metabolismo , Regulação da Expressão Gênica , Humanos , Mitocôndrias/genética , Mitocôndrias/fisiologia , Proteínas Nucleares/genética , Proteína Oncogênica p55(v-myc)/genética , Proteína Oncogênica p55(v-myc)/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Esteroides/metabolismo
16.
Nature ; 483(7391): 608-12, 2012 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-22460906

RESUMO

Deregulated expression of the MYC oncoprotein contributes to the genesis of many human tumours, yet strategies to exploit this for a rational tumour therapy are scarce. MYC promotes cell growth and proliferation, and alters cellular metabolism to enhance the provision of precursors for phospholipids and cellular macromolecules. Here we show in human and murine cell lines that oncogenic levels of MYC establish a dependence on AMPK-related kinase 5 (ARK5; also known as NUAK1) for maintaining metabolic homeostasis and for cell survival. ARK5 is an upstream regulator of AMPK and limits protein synthesis via inhibition of the mammalian target of rapamycin 1 (mTORC1) signalling pathway. ARK5 also maintains expression of mitochondrial respiratory chain complexes and respiratory capacity, which is required for efficient glutamine metabolism. Inhibition of ARK5 leads to a collapse of cellular ATP levels in cells expressing deregulated MYC, inducing multiple pro-apoptotic responses as a secondary consequence. Depletion of ARK5 prolongs survival in MYC-driven mouse models of hepatocellular carcinoma, demonstrating that targeting cellular energy homeostasis is a valid therapeutic strategy to eliminate tumour cells that express deregulated MYC.


Assuntos
Regulação Neoplásica da Expressão Gênica , Genes myc/genética , Proteínas Quinases/metabolismo , Proteínas Repressoras/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Apoptose , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Respiração Celular , Sobrevivência Celular , Transformação Celular Neoplásica/genética , Modelos Animais de Doenças , Doxiciclina/farmacologia , Transporte de Elétrons , Glutamina/metabolismo , Homeostase , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Mitocôndrias/metabolismo , Complexos Multiproteicos , Proteína Oncogênica p55(v-myc)/genética , Proteína Oncogênica p55(v-myc)/metabolismo , Biossíntese de Proteínas , Proteínas Quinases/deficiência , Proteínas Quinases/genética , Proteínas/antagonistas & inibidores , Proteínas/metabolismo , Interferência de RNA , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
17.
PLoS One ; 7(12): e52714, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23300748

RESUMO

A major challenge for further development of drug screening procedures, cell replacement therapies and developmental studies is the identification of expandable human stem cells able to generate the cell types needed. We have previously reported the generation of an immortalized polyclonal neural stem cell (NSC) line derived from the human fetal ventral mesencephalon (hVM1). This line has been biochemically, genetically, immunocytochemically and electrophysiologically characterized to document its usefulness as a model system for the generation of A9 dopaminergic neurons (DAn). Long-term in vivo transplantation studies in parkinsonian rats showed that the grafts do not mature evenly. We reasoned that diverse clones in the hVM1 line might have different abilities to differentiate. In the present study, we have analyzed 9 hVM1 clones selected on the basis of their TH generation potential and, based on the number of v-myc copies, v-myc down-regulation after in vitro differentiation, in vivo cell cycle exit, TH⁺ neuron generation and expression of a neuronal mature marker (hNSE), we selected two clones for further in vivo PD cell replacement studies. The conclusion is that homogeneity and clonality of characterized NSCs allow transplantation of cells with controlled properties, which should help in the design of long-term in vivo experiments.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Células-Tronco Neurais/fisiologia , Doença de Parkinson Secundária/terapia , Animais , Pontos de Checagem do Ciclo Celular , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Neurônios Dopaminérgicos/transplante , Regulação para Baixo , Feminino , Dosagem de Genes , Expressão Gênica , Humanos , Proteínas de Filamentos Intermediários/genética , Proteínas de Filamentos Intermediários/metabolismo , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Mesencéfalo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Nestina , Células-Tronco Neurais/transplante , Proteína Oncogênica p55(v-myc)/genética , Proteína Oncogênica p55(v-myc)/metabolismo , Ratos , Ratos Sprague-Dawley , Pesquisa com Células-Tronco
18.
Proc Natl Acad Sci U S A ; 108(12): 4876-81, 2011 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-21378266

RESUMO

In the field of induced potency and fate reprogramming, it remains unclear what the best starting cell might be and to what extent a cell need be transported back to a more primitive state for translational purposes. Reprogramming a committed cell back to pluripotence to then instruct it toward a particular specialized cell type is demanding and may increase risks of neoplasia and undesired cell types. Precursor/progenitor cells from the organ of therapeutic concern typically lack only one critical attribute--the capacity for sustained self-renewal. We speculated that this could be induced in a regulatable manner such that cells proliferate only in vitro and differentiate in vivo without the need for promoting pluripotence or specifying lineage identity. As proof-of-concept, we generated and tested the efficiency, safety, engraftability, and therapeutic utility of "induced conditional self-renewing progenitor (ICSP) cells" derived from the human central nervous system (CNS); we conditionally induced self-renewal efficiently within neural progenitors solely by introducing v-myc tightly regulated by a tetracycline (Tet)-on gene expression system. Tet in the culture medium activated myc transcription and translation, allowing efficient expansion of homogeneous, clonal, karyotypically normal human CNS precursors ex vivo; in vivo, where Tet was absent, myc was not expressed, and self-renewal was entirely inactivated (as was tumorigenic potential). Cell proliferation ceased, and differentiation into electrophysiologically active neurons and other CNS cell types in vivo ensued upon transplantation into rats, both during development and after adult injury--with functional improvement and without neoplasia, overgrowth, deformation, emergence of non-neural cell types, phenotypic or genomic instability, or need for immunosuppression. This strategy of inducing self-renewal might be applied to progenitors from other organs and may prove to be a safe, effective, efficient, and practical method for optimizing insights gained from the ability to reprogram cells.


Assuntos
Lesões Encefálicas/terapia , Encéfalo/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Neurais/citologia , Transplante de Células-Tronco , Animais , Encéfalo/metabolismo , Linhagem Celular , Proliferação de Células , Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Células-Tronco Neurais/metabolismo , Proteína Oncogênica p55(v-myc)/genética , Proteína Oncogênica p55(v-myc)/metabolismo , Ratos , Ratos Sprague-Dawley , Transplante Heterólogo
19.
Oncogene ; 28(26): 2446-55, 2009 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-19448669

RESUMO

Myc, a key regulator of cellular proliferation, differentiation and apoptosis, exerts its biological functions by activating or suppressing the transcription of specific sets of target genes. C/EBP transcription factors play important roles during differentiation of various cell types and have been identified as critical targets for v-Myc- and c-Myc-dependent suppression of myeloid and fat cell differentiation. Here, we have addressed the mechanism by which v-Myc suppresses the activity of C/EBPbeta. We show that v-Myc is recruited to the aminoterminal domain of C/EBPbeta and interferes with the cooperation of C/EBPbeta and the co-activator p300 by preventing C/EBPbeta-induced phosphorylation of p300. We have identified the protein kinase responsible for C/EBPbeta-induced phosphorylation of p300 as homeo-domain interacting protein kinase 2 (HIPK2) and show that v-Myc displaces the kinase from the C/EBPbeta-p300 complex. Overall, our findings that the modulation of the C/EBPbeta-induced phosphorylation of p300 as a new mechanism of transcriptional suppression by v-Myc.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/antagonistas & inibidores , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Proteína p300 Associada a E1A/metabolismo , Inibidores Enzimáticos/metabolismo , Proteína Oncogênica p55(v-myc)/metabolismo , Sequência de Aminoácidos , Animais , Proteína beta Intensificadora de Ligação a CCAAT/química , Proteína beta Intensificadora de Ligação a CCAAT/genética , Linhagem Celular , Galinhas , Regulação da Expressão Gênica , Humanos , Camundongos , Dados de Sequência Molecular , Células Mieloides/metabolismo , Proteína Oncogênica p55(v-myc)/genética , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Codorniz
20.
Oncogene ; 28(17): 1892-903, 2009 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-19330026

RESUMO

Co-amplification at chromosomes 8p11-8p12 and 11q12-11q14 occurs often in breast tumors, suggesting possible cooperation between genes in these regions in oncogenesis. We used high-resolution array comparative genomic hybridization (array CGH) to map the minimal amplified regions. The 8p and 11q amplicons are complex and consist of at least four amplicon cores at each site. Candidate oncogenes mapping to these regions were identified by combining copy number and RNA and protein expression analyses. These studies also suggested that CCND1 at 11q13 induced expression of ZNF703 mapping at 8p12, which was subsequently shown to be mediated by the Rb/E2F pathway. Nine candidate oncogenes from 8p12 and four from 11q13 were further evaluated for oncogenic function. None of the genes individually promoted colony formation in soft agar or collaborated with each other functionally. On the other hand, FGFR1 and DDHD2 at 8p12 cooperated functionally with MYC, whereas CCND1 and ZNF703 cooperated with a dominant negative form of TP53. These observations highlight the complexity and functional consequences of the genomic rearrangements that occur in these breast cancer amplicons, including transcriptional cross-talk between genes in the 8p and 11q amplicons, as well as their cooperation with major pathways of tumorigenesis.


Assuntos
Neoplasias da Mama/genética , Cromossomos Humanos Par 11/genética , Cromossomos Humanos Par 8/genética , Amplificação de Genes , Oncogenes/genética , Neoplasias da Mama/patologia , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Hibridização Genômica Comparativa , Ciclina D1/genética , Epistasia Genética , Feminino , Dosagem de Genes , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Humanos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Proteína Oncogênica p55(v-myc)/genética , Fosfolipases/genética , RNA Interferente Pequeno/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Transfecção , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...