Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cancer Sci ; 112(12): 4957-4967, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34689384

RESUMO

Laminins are heterotrimeric ECM proteins composed of α, ß, and γ chains. The γ2 chain (Lm-γ2) is a frequently expressed monomer and its expression is closely associated with cancer progression. Laminin-γ2 contains an epidermal growth factor (EGF)-like domain in its domain III (DIII or LEb). Matrix metalloproteinases can cleave off the DIII region of Lm-γ2 that retains the ligand activity for EGF receptor (EGFR). Herein, we show that a novel short form of Lm-γ2 (Lm-γ2F) containing DIII is generated without requiring MMPs and chromosomal translocation between LAMC2 on chromosome 1 and NR6A1 gene locus on chromosome 9 in human ovarian cancer SKOV3 cells. Laminin-γ2F is expressed as a truncated form lacking domains I and II, which are essential for its association with Lm-α3 and -ß3 chains of Lm-332. Secreted Lm-γ2F can act as an EGFR ligand activating the EGFR/AKT pathways more effectively than does the Lm-γ2 chain, which in turn promotes proliferation, survival, and motility of ovarian cancer cells. LAMC2-NR6A1 translocation was detected using in situ hybridization, and fusion transcripts were expressed in ovarian cancer cell tissues. Overexpression and suppression of fusion transcripts significantly increased and decreased the tumorigenic growth of cells in mouse models, respectively. To the best of our knowledge, this is the first report regarding a fusion gene of ECM showing that translocation of LAMC2 plays a crucial role in the malignant growth and progression of ovarian cancer cells and that the consequent product is a promising therapeutic target against ovarian cancers.


Assuntos
Cocarcinogênese/genética , Regulação Neoplásica da Expressão Gênica , Laminina/genética , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/genética , Proteínas de Fusão Oncogênica/genética , Neoplasias Ovarianas/genética , Animais , Linhagem Celular Tumoral , Cocarcinogênese/metabolismo , Feminino , Humanos , Laminina/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Interferência de RNA , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
2.
Genes Dev ; 27(8): 941-54, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23630078

RESUMO

MicroRNAs (miRNAs) are critical to proliferation, differentiation, and development. Here, we characterize gene expression in murine Dicer-null adult mesenchymal stem cell lines, a fibroblast cell type. Loss of Dicer leads to derepression of let-7 targets at levels that exceed 10-fold to 100-fold with increases in transcription. Direct and indirect targets of this miRNA belong to a mid-gestation embryonic program that encompasses known oncofetal genes as well as oncogenes not previously associated with an embryonic state. Surprisingly, this mid-gestation program represents a distinct period that occurs between the pluripotent state of the inner cell mass at embryonic day 3.5 (E3.5) and the induction of let-7 upon differentiation at E10.5. Within this mid-gestation program, we characterize the let-7 target Nr6a1, an embryonic transcriptional repressor that regulates gene expression in adult fibroblasts following miRNA loss. In total, let-7 is required for the continual suppression of embryonic gene expression in adult cells, a mechanism that may underlie its tumor-suppressive function.


Assuntos
Fibroblastos/citologia , Regulação da Expressão Gênica , MicroRNAs/genética , MicroRNAs/metabolismo , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/genética , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Animais , Antígenos de Neoplasias/metabolismo , Linhagem Celular , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Fibroblastos/metabolismo , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Ligação Proteica , Ribonuclease III/genética , Ribonuclease III/metabolismo
3.
Cell Commun Signal ; 17(1): 77, 2019 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-31315616

RESUMO

BACKGROUND: Lipogenesis is required for the optimal growth of many types of cancer cells, it is shown to control the biosynthesis of the lipid bilayer membrane during rapid proliferation and metastasis, provides cancer cells with signaling lipid molecules to support cancer development and make cancer cells more resistant to oxidative stress-induced cell death. Though multiple lipogenic enzymes have been identified to mediate this metabolic change, how the expression of these lipogenic enzymes are transcriptionally regulated remains unclear. METHODS: Gain- and loss-of-function experiments were conducted to assess the role of transcriptional repressor, nuclear receptor sub-family 6, group A, member 1 (NR6A1) in HepG2 cells. RT-qPCR method was performed to investigate target gene of NR6A1. Western blot was employed to determine the mechanisms by which NR6A1 regulates lipid accumulation in HepG2 cells. RESULTS: We provide evidence that NR6A1 is a novel regulator of lipid metabolism in HepG2 cells. NR6A1 knockdown can increase lipid accumulation as well as insulin-induced proliferation and migration of HepG2 cells. The lipogenic effect correlated well with the expression of lipogenic genes, including fatty acid synthase (FAS), diglyceride acyltransferase-2 (DGAT2), malic enzyme 1 (ME1), microsomal triglyceride transfer protein (MTTP) and phosphoenolpyruvate carboxykinase (PEPCK). NR6A1 knockdown also increased the expression of carnitine palmitoyltransferase 1A (CPT1a), the rate-limiting enzyme in fatty acid oxidation. Furthermore, NR6A1 knockdown induced lipid accumulation through mammalian target of rapamycin complex 1 (mTORC1), but not mTORC2. Moreover, siRNA-mediated knockdown of NR6A1 increased expression of insulin receptor (INSR) and potentitated insulin-induced phosphorylation of mTOR and AKT partly via miR-205-5p in HepG2 cells. CONCLUSIONS: These findings provide important new insights into the role of NR6A1 in the lipogenesis in HepG2 cells. .


Assuntos
Metabolismo dos Lipídeos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Animais , Movimento Celular , Proliferação de Células , Regulação da Expressão Gênica , Inativação Gênica , Células HEK293 , Células Hep G2 , Humanos , Insulina/metabolismo , Lipogênese , MicroRNAs/genética , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/deficiência , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/genética , Receptor de Insulina/genética , Transdução de Sinais
4.
Anim Genet ; 50(2): 166-171, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30741434

RESUMO

Autochthonous pig breeds are usually reared in extensive or semi-extensive production systems that might facilitate contact with wild boars and, thus, reciprocal genetic exchanges. In this study, we analysed variants in the melanocortin 1 receptor (MC1R) gene (which cause different coat colour phenotypes) and in the nuclear receptor subfamily 6 group A member 1 (NR6A1) gene (associated with increased vertebral number) in 712 pigs of 12 local pig breeds raised in Italy (Apulo-Calabrese, Casertana, Cinta Senese, Mora Romagnola, Nero Siciliano and Sarda) and south-eastern European countries (Krskopolje from Slovenia, Black Slavonian and Turopolje from Croatia, Mangalitsa and Moravka from Serbia and East Balkan Swine from Bulgaria) and compared the data with the genetic variability at these loci investigated in 229 wild boars from populations spread in the same macro-geographic areas. None of the autochthonous pig breeds or wild boar populations were fixed for one allele at both loci. Domestic and wild-type alleles at these two genes were present in both domestic and wild populations. Findings of the distribution of MC1R alleles might be useful for tracing back the complex genetic history of autochthonous breeds. Altogether, these results indirectly demonstrate that bidirectional introgression of wild and domestic alleles is derived and affected by the human and naturally driven evolutionary forces that are shaping the Sus scrofa genome: autochthonous breeds are experiencing a sort of 'de-domestication' process, and wild resources are challenged by a 'domestication' drift. Both need to be further investigated and managed.


Assuntos
Domesticação , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/genética , Receptor Tipo 1 de Melanocortina/genética , Sus scrofa/genética , Alelos , Animais , Cruzamento , Europa Oriental , Itália , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Receptor Tipo 1 de Melanocortina/metabolismo
5.
J Cell Biochem ; 119(3): 2719-2730, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29057499

RESUMO

We demonstrate that OCT4 expression is regulated by germ cell nuclear factor (GCNF) via its interactions with three nuclear receptor (NR) binding sites within OCT4 promoter conserved regions (CRs) in human embryonic carcinoma (EC) NCCIT cells. OCT4 expression is gradually reduced during the retinoic acid-induced differentiation, while GCNF temporarily increased after 2 days and then significantly decreased. In addition, OCT4 expression is significantly reduced by overexpression of exogenous GCNF, but increased by GCNF shRNA-mediated knockdown. The transcriptional activity of OCT4 is significantly inhibited by dose-dependent overexpression of GCNF. While mutants at each of the NR binding sites retain the repressive effects of GCNF on OCT4 promoter activity, the repressive effect was completely eliminated in the reporter construct with all binding sites mutated even in the presence of GCNF. Furthermore, the transcriptional activity of native minimal promoter (CR1-Luc) containing the first NR binding site was significantly reduced by GCNF overexpression, while the mutant retained basal activity to some extent. Next, an exogenous minimal ti promoter-inserted CR2 reporter construct containing the second and third NR binding sites (CR2-ti-Luc) was co-transfected with GCNF expression vector. The transcriptional activity of CR2-ti-Luc was significantly decreased by GCNF overexpression, while mutation of both binding sites retained the transcriptional activity of the reporter construct. Binding assays confirmed the direct interaction of GCNF with all three NR binding sites cooperatively. Taken together, GCNF acts as a transcriptional repressor in the regulation of OCT4 gene expression through cooperative interaction with three NR binding elements in pluripotent NCCIT cells.


Assuntos
Carcinoma Embrionário/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/metabolismo , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Fator 3 de Transcrição de Octâmero/biossíntese , Elementos de Resposta , Carcinoma Embrionário/genética , Carcinoma Embrionário/patologia , Linhagem Celular Tumoral , Humanos , Proteínas de Neoplasias/genética , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/genética , Fator 3 de Transcrição de Octâmero/genética
6.
J Biol Chem ; 291(16): 8644-52, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26769970

RESUMO

Oct4 is considered a key transcription factor for pluripotent stem cell self-renewal. It binds to specific regions within target genes to regulate their expression and is downregulated upon induction of differentiation of pluripotent stem cells; however, the mechanisms that regulate the levels of human Oct4 expression remain poorly understood. Here we show that expression of human Oct4 is directly repressed by germ cell nuclear factor (GCNF), an orphan nuclear receptor, in hES cells. Knockdown of GCNF by siRNA resulted in maintenance of Oct4 expression during RA-induced hES cell differentiation. While overexpression of GCNF promoted repression of Oct4 expression in both undifferentiated and differentiated hES cells. The level of Oct4 repression was dependent on the level of GCNF expression in a dose-dependent manner. mRNA microarray analysis demonstrated that overexpression of GCNF globally regulates gene expression in undifferentiated and differentiated hES cells. Within the group of altered genes, GCNF down-regulated 36% of the genes, and up-regulated 64% in undifferentiated hES cells. In addition, GCNF also showed a regulatory gene pattern that is different from RA treatment during hES cell differentiation. These findings increase our understanding of the mechanisms that maintain hES cell pluripotency and regulate gene expression during the differentiation process.


Assuntos
Diferenciação Celular/fisiologia , Regulação da Expressão Gênica/fisiologia , Células-Tronco Embrionárias Humanas/metabolismo , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Fator 3 de Transcrição de Octâmero/biossíntese , Proteínas Repressoras/metabolismo , Linhagem Celular , Células-Tronco Embrionárias Humanas/citologia , Humanos , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/genética , Fator 3 de Transcrição de Octâmero/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas Repressoras/genética
7.
Nanomedicine ; 13(4): 1363-1375, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28219741

RESUMO

The human glial-cell derived neurotrophic factor (hGDNF) gene transfer by neurotensin (NTS)-polyplex nanoparticles functionally restores the dopamine nigrostriatal system in experimental Parkinson's disease models. However, high levels of sustained expression of GDNF eventually can cause harmful effects. Herein, we report an improved NTS-polyplex nanoparticle system that enables regulation of hGDNF expression within dopaminergic neurons. We constructed NTS-polyplex nanoparticles containing a single bifunctional plasmid that codes for the reverse tetracycline-controlled transactivator advanced (rtTA-Adv) under the control of NBRE3x promoter, and for hGDNF under the control of tetracycline-response element (TRE). Another bifunctional plasmid contained the enhanced green fluorescent protein (GFP) gene. Transient transfection experiments in N1E-115-Nurr1 cells showed that doxycycline (100 ng/mL) activates hGDNF and GFP expression. Doxycycline (5 mg/kg, i.p.) administration in rats activated hGDNF expression only in transfected dopaminergic neurons, whereas doxycycline withdrawal silenced transgene expression. Our results offer a specific doxycycline-regulated system suitable for nanomedicine-based treatment of Parkinson's disease.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Doxiciclina/farmacologia , Regulação da Expressão Gênica , Nanopartículas/química , Neurotensina/química , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/genética , Animais , Linhagem Celular Tumoral , Vetores Genéticos , Humanos , Masculino , Camundongos , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Doença de Parkinson/tratamento farmacológico , Plasmídeos , Regiões Promotoras Genéticas , Ratos , Ratos Wistar , Elementos de Resposta , Transfecção , Transgenes
8.
Semin Cell Dev Biol ; 24(10-12): 679-86, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24029702

RESUMO

GCNF (NR6A1) is essential for embryonic development. GCNF belongs to the nuclear receptor (NR) gene family, it is distantly related to other NRs and is the only member of subfamily 6. As the ligand for GCNF has not been identified, GCNF is designated an orphan nuclear receptor. GCNF has been found to be a transcriptional repressor, through specific binding to DR0 response elements, which is found in the Oct4 proximal promoter for example. GCNF is expressed widely in early mouse embryos, and later in the developing nervous system. GCNF knockout mouse embryos die around E10.5. GCNF is required for the restriction of Oct4 expression to primordial germ cells after gastrulation. GCNF is expressed in ES/EC cells and during their differentiation, and has been reported to be required for pluripotency gene repression during retinoic acid (RA)-induced mES cell differentiation. GCNF can interact with DNA methylation proteins, and is suggested to recruit DNA methylation complexes to repress and silence Oct4 expression. Nuclear receptor regulation in embryonic development is a complex process, as different nuclear receptors have overlapping and distinct functions. In-depth exploration of GCNF function and mechanism of action will help to comprehensively understand the nuclear receptor regulation in embryonic development.


Assuntos
Desenvolvimento Embrionário , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Sequência de Aminoácidos , Animais , DNA/metabolismo , Humanos , Dados de Sequência Molecular , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/química , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/genética , Multimerização Proteica , Proteínas Repressoras/metabolismo , Elementos de Resposta/genética
9.
Stem Cells ; 32(6): 1527-37, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24578347

RESUMO

Cyclin D1 plays an important role in the regulation of cellular proliferation and its expression is activated during gastrulation in the mouse; however, it remains unknown how cyclin D1 expression is regulated during early embryonic development. Here, we define the role of germ cell nuclear factor (GCNF) in the activation of cyclin D1 expression during embryonic stem cell (ESC) differentiation as a model of early development. During our study of GCNF knockout (GCNF(-) (/) (-) ) ESC, we discovered that loss of GCNF leads to the repression of cyclin D1 activation during ESC differentiation. This was determined to be an indirect effect of deregulation Mir302a, which is a cyclin D1 suppressor via binding to the 3'UTR of cyclin D1 mRNA. Moreover, we showed that Mir302 is a target gene of GCNF that inhibits Mir302 expression by binding to a DR0 element within its promoter. Inhibition of Mir302a using Mir302 inhibitor during differentiation of GCNF(-) (/) (-) ESCs restored cyclin D1 expression. Similarly over-expression of GCNF during differentiation of GCNF(-) (/) (-) ESCs rescued the inhibition of Mir302a expression and the activation of cyclin D1. These results reveal that GCNF plays a key role in regulating activation of cyclin D1 expression via inhibition of Mir302a.


Assuntos
Diferenciação Celular/genética , Ciclina D1/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Animais , Proliferação de Células , Forma Celular , Ensaio de Unidades Formadoras de Colônias , Ciclina D1/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Modelos Biológicos , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/deficiência , Regiões Promotoras Genéticas/genética , Ligação Proteica/genética
10.
J Cell Physiol ; 228(6): 1174-88, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23129342

RESUMO

Human Cripto-1 (CR-1) plays an important role in regulating embryonic development while also regulating various stages of tumor progression. However, mechanisms that regulate CR-1 expression during embryogenesis and tumorigenesis are still not well defined. In the present study, we investigated the effects of two nuclear receptors, liver receptor homolog (LRH)-1 and germ cell nuclear factor receptor (GCNF) and epigenetic modifications on CR-1 gene expression in NTERA-2 human embryonal carcinoma cells and in breast cancer cells. CR-1 expression in NTERA-2 cells was positively regulated by LRH-1 through direct binding to a DR0 element within the CR-1 promoter, while GCNF strongly suppressed CR-1 expression in these cells. In addition, the CR-1 promoter was unmethylated in NTERA-2 cells, while T47D, ZR75-1, and MCF7 breast cancer cells showed high levels of CR-1 promoter methylation and low CR-1 mRNA and protein expression. Treatment of breast cancer cells with a demethylating agent and histone deacetylase inhibitors reduced methylation of the CR-1 promoter and reactivated CR-1 mRNA and protein expression in these cells, promoting migration and invasion of breast cancer cells. Analysis of a breast cancer tissue array revealed that CR-1 was highly expressed in the majority of human breast tumors, suggesting that CR-1 expression in breast cancer cell lines might not be representative of in vivo expression. Collectively, these findings offer some insight into the transcriptional regulation of CR-1 gene expression and its critical role in the pathogenesis of human cancer.


Assuntos
Neoplasias da Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Carcinoma Embrionário/metabolismo , Metilação de DNA , Proteínas Ligadas por GPI/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Neoplasias/metabolismo , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Regiões Promotoras Genéticas , Receptores Citoplasmáticos e Nucleares/metabolismo , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Sítios de Ligação , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/patologia , Carcinoma Embrionário/genética , Carcinoma Embrionário/patologia , Movimento Celular , Metilação de DNA/efeitos dos fármacos , Metilases de Modificação do DNA/antagonistas & inibidores , Metilases de Modificação do DNA/metabolismo , Decitabina , Relação Dose-Resposta a Droga , Células-Tronco de Carcinoma Embrionário/metabolismo , Células-Tronco de Carcinoma Embrionário/patologia , Feminino , Proteínas Ligadas por GPI/genética , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Inibidores de Histona Desacetilases/farmacologia , Humanos , Ácidos Hidroxâmicos/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Luciferases/biossíntese , Luciferases/genética , Células MCF-7 , Invasividade Neoplásica , Proteínas de Neoplasias/genética , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/genética , Interferência de RNA , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Tempo , Análise Serial de Tecidos , Transcrição Gênica , Transfecção , Tretinoína/farmacologia , Ácido Valproico/farmacologia
11.
Prostate ; 73(10): 1103-14, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23532770

RESUMO

BACKGROUND: Cancer/Testis (CT) genes are expressed in male gonads, repressed in most healthy somatic tissues and de-repressed in various somatic malignancies including prostate cancers (PCa). Because of their specific expression signature and their associations with tumor aggressiveness and poor outcomes, CT genes are considered to be useful biomarkers and they are also targets for the development of new anti-cancer immunotherapies. The aim of this study was to identify novel CT genes associated with hormone-sensitive prostate cancer (HSPC), and castration-resistant prostate cancer (CRPC). METHODS: To identify novel CT genes we screened genes for which transcripts were detected by RNA profiling specifically in normal testis and in either HSPC or CRPC as compared to normal prostate and 44 other healthy tissues using GeneChips. The expression and clinicopathological significance of a promising candidate--NR6A1--was examined in HSPC, CRPC, and metastatic site samples using tissue microarrays. RESULTS: We report the identification of 98 genes detected in CRPC, HSPC and testicular samples but not in the normal controls. Among them, cellular levels of NR6A1 were found to be higher in HSPC compared to normal prostate and further increased in metastatic lesions and CRPC. Furthermore, increased NR6A1 immunoreactivity was significantly associated with a high Gleason score, advanced pT stage and cancer cell proliferation. CONCLUSIONS: Our results show that cellular levels of NR6A1 are correlated with disease progression in PCa. We suggest that this essential orphan nuclear receptor is a potential therapeutic target as well as a biomarker of PCa aggressiveness.


Assuntos
Biomarcadores Tumorais/genética , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/genética , Próstata/metabolismo , Neoplasias da Próstata/genética , Testículo/metabolismo , Idoso , Biomarcadores Tumorais/metabolismo , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Orquiectomia , Próstata/patologia , Próstata/cirurgia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Neoplasias da Próstata/cirurgia , Testículo/patologia , Testículo/cirurgia , Análise Serial de Tecidos
12.
Stem Cells ; 29(7): 1041-51, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21608077

RESUMO

The pluripotency gene Oct4 encodes a key transcription factor that maintains self-renewal of embryonic stem cell (ESC) and is downregulated upon differentiation of ESCs and silenced in somatic cells. A combination of cis elements, transcription factors, and epigenetic modifications, such as DNA methylation, mediates Oct4 gene expression. Here, we show that the orphan nuclear receptor germ cell nuclear factor (GCNF) initiates Oct4 repression and DNA methylation by the differential recruitment of methyl-CpG binding domain (MBD) and DNA methyltransferases (Dnmts) to the Oct4 promoter. When compared with wild-type ESCs and gastrulating embryos, Oct4 repression is lost and its proximal promoter is significantly hypomethylated in retinoic acid (RA)-differentiated GCNF(-/-) ESCs and GCNF(-/-) embryos. Efforts to characterize mediators of GCNF's repressive function and DNA methylation of the Oct4 promoter identified MBD3, MBD2, and de novo Dnmts as GCNF interacting factors. Upon differentiation, endogenous GCNF binds to the Oct4 proximal promoter and differentially recruits MBD3 and MBD2 as well as Dnmt3A. In differentiated GCNF(-/-) ESCs, recruitment of MBD3 and MBD2 as well as Dnmt3A to Oct4 promoter is lost and subsequently Oct4 repression and DNA methylation failed to occur. Hypomethylation of the Oct4 promoter is also observed in RA-differentiated MBD3(-/-) and Dnmt3A(-/-) ESCs, but not in MBD2(-/-) and Dnmt3B(-/-) ESCs. Thus, recruitment of MBD3, MBD2, and Dnmt3A by GCNF links two events: gene-specific repression and DNA methylation, which occur differentially at the Oct4 promoter. GCNF initiates the repression and epigenetic modification of Oct4 gene during ESC differentiation.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/fisiologia , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , Proteínas de Ligação a DNA/genética , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Camundongos , Dados de Sequência Molecular , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/genética , Fator 3 de Transcrição de Octâmero/biossíntese , Regiões Promotoras Genéticas , Ligação Proteica , Fatores de Transcrição/genética
13.
Nucleic Acids Res ; 38(7): 2268-78, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20071744

RESUMO

Cellular differentiation and development of germ cells critically depend on a coordinated activation and repression of specific genes. The underlying regulation mechanisms, however, still lack a lot of understanding. Here, we describe that both the testis-specific transcriptional activator CREMtau (cAMP response element modulator tau) and the repressor GCNF (germ cell nuclear factor) have an overlapping binding site which alone is sufficient to direct cell type-specific expression in vivo in a heterologous promoter context. Expression of the transgene driven by the CREM/GCNF site is detectable in spermatids, but not in any somatic tissue or at any other stages during germ cell differentiation. CREMtau acts as an activator of gene transcription whereas GCNF suppresses this activity. Both factors compete for binding to the same DNA response element. Effective binding of CREM and GCNF highly depends on composition and epigenetic modification of the binding site. We also discovered that CREM and GCNF bind to each other via their DNA binding domains, indicating a complex interaction between the two factors. There are several testis-specific target genes that are regulated by CREM and GCNF in a reciprocal manner, showing a similar activation pattern as during spermatogenesis. Our data indicate that a single common binding site for CREM and GCNF is sufficient to specifically direct gene transcription in a tissue-, cell type- and differentiation-specific manner.


Assuntos
Modulador de Elemento de Resposta do AMP Cíclico/metabolismo , Regulação da Expressão Gênica , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Testículo/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Cromatina/química , DNA/química , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Elementos de Resposta , Espermátides/metabolismo
14.
Neuropharmacology ; 209: 108990, 2022 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-35183538

RESUMO

Chronic stress exposure is a risk factor that can induce the development of depression-like behaviors by impairing the hippocampal cyclic adenosine monophosphate-response element binding protein (CREB)-brain-derived neurotrophic factor (BDNF) signaling, but its underlying mechanisms remain largely unknown. We identified an orphan receptor that can suppress the activity of CREB, nuclear receptor sub-family 6, group A, member 1 (NR6A1), in mouse brain neurons. Given the critical role of the impaired CREB-BDNF signaling in depression, we speculate that the neuronal NR6A1 may mediate the pathogenesis of depression. Results showed that chronic unpredictable stress (CUS) markedly increased the expression levels of hippocampal NR6A1 protein, which reduced hippocampal CREB phosphorylation and BDNF protein expression. Overexpression of hippocampal NR6A1 in stress-naïve mice simulated chronic stress, inducing depression-like behaviors in the tail suspension test, forced swimming test, and sucrose preference test, and impairing the hippocampal CREB-BDNF signaling cascade. Genetic knockdown of hippocampal NR6A1 did not affect mouse behaviors but prevented the CUS-induced depression-like behaviors in mice and impairment in hippocampal CREB-BDNF signaling. Furthermore, genetic knockdown of hippocampal CREB or BDNF abrogated the preventive effect of hippocampal NR6A1 down-regulation on CUS-induced depression-like behaviors in mice. Collectively, these results for the first time identified a nuclear expression of NR6A1 in mouse brain neurons, and showed that the abnormally increased NR6A1 protein in the hippocampus in mice treated with or without chronic stress can impair the CREB-BDNF signaling cascade and lead to the development of depression-like behaviors. Hippocampal NR6A1 could be a novel target for the development of antidepressants.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Depressão , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Depressão/metabolismo , Modelos Animais de Doenças , Hipocampo , Camundongos , Estresse Psicológico/complicações , Estresse Psicológico/metabolismo
15.
Biochem Biophys Res Commun ; 403(2): 178-83, 2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-21056546

RESUMO

The hypocretin (also known as orexin) neuropeptide system coordinates the regulation of various physiological processes. A reduction in Nr6a1 expression was observed in hypocretin neuron-ablated transgenic mice. To show that prepro-hypocretin transcription is functionally modulated by NR6A1, we performed chromatin immunoprecipitation (ChIP) analysis, double-immunostaining, a luciferase reporter assay, and an in utero electroporation study. ChIP analysis showed that endogenous NR6A1 binds to a putative NR6A1-binding site. Double-immunostaining indicated almost all hypocretin neurons were positive for NR6A1 immunoreactivity. NR6A1 overexpression in SH-SY5Y cells modulated hypocretin promoter activity, an effect that was countered by lacking a putative NR6A1-binding site. Electroporation with Nr6a1 in the foetal hypothalamus promoted hypocretin transcription as compared to GFP-electroporation. These experiments confirmed that NR6A1 works as a regulator for hypocretin transcription.


Assuntos
Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neuropeptídeos/genética , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Análise Mutacional de DNA , Humanos , Hipotálamo/metabolismo , Imunoprecipitação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Orexinas , Regiões Promotoras Genéticas , Deleção de Sequência , Transcrição Gênica
16.
Mol Reprod Dev ; 77(3): 273-84, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20020523

RESUMO

Nuclear receptor subfamily 6, group A, member 1 (NR6A1), also known as germ cell nuclear factor/retinoid receptor-related testis-associated receptor and neuronal cell nuclear factor, is a member of the nuclear orphan receptor superfamily. NR6A1 has been cloned in various species including humans and mice, but it has been scarcely investigated in avian species. In the present study, we cloned the chicken NR6A1 (cNR6A1) from a testis cDNA library. The cloned cNR6A1 sequence was mapped to chromosome 17 and contained an open reading frame of 1.4 kb encoding 445 amino acids. Multiple alignment analysis of the cNR6A1 protein-coding sequence with NR6A1s from humans, mice, boars, rats, zebrafish, and Xenopus showed high degrees of homology, 89%, 90%, 89%, 88%, 83%, and 87%, respectively. Using RNA interference, changes in the expression of pluripotency-, germ cell-, and differentiation-related key genes by silencing of cNR6A1 were validated in chicken blastoderm-derived embryonic stem cells. Among those genes, the relative expression levels of POU5F1, CRIPTO, DAZL, DDX4, BMP15, GSC, and SOX7 changed significantly compared to the control group. We also confirmed that the activity of alkaline phosphatase, known as a pluripotency marker, was maintained by cNR6A1 gene silencing in chicken blastodermal cells. Collectively, our data suggest that cNR6A1 may play an important role during chicken embryonic development and differentiation.


Assuntos
Galinhas/genética , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/genética , Interferência de RNA/fisiologia , Fosfatase Alcalina/metabolismo , Sequência de Aminoácidos , Animais , Blastoderma/metabolismo , Diferenciação Celular/genética , Mapeamento Cromossômico/métodos , Clonagem Molecular/métodos , Cães , Células-Tronco Embrionárias/metabolismo , Perfilação da Expressão Gênica , Humanos , Dados de Sequência Molecular , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Filogenia , Células-Tronco Pluripotentes/metabolismo , Reação em Cadeia da Polimerase , Ratos , Reprodutibilidade dos Testes , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
17.
Cancer Med ; 9(23): 9107-9122, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33034957

RESUMO

Testicular germ cell tumors (TGCTs) are a diverse group of neoplasms that are derived from dysfunctional fetal germ cells and can also present in extragonadal sites. The genetic drivers underlying malignant transformation of TGCTs have not been fully elucidated so far. The aim of the present study is to clarify the functional role and regulatory mechanism of miR-196a-5p in TGCTs. We demonstrated that miR-196a-5p was downregulated in TGCTs. It can inhibit the proliferation, migration, and invasion of testicular tumor cell lines including NT-2 and NCCIT through targeting the NR6A1 gene, which we proved its role in promotion of cell proliferation and repression of cellular junction and aggregation. Mechanistically, NR6A1 inhibited E-cadherin through binding with DR0 sites in the CDH1 gene promoter and recruiting methyltransferases Dnmt1. Further, NR6A1 promoted neuronal marker protein MAP2 expression in RA-induced neurodifferentiation of NT-2 cells and testicular tumor xenografts. Clinical histopathologically, NR6A1 was positively correlated with MAP2, and negatively correlated with E-cadherin in TGCTs. These findings revealed that the miR-196a-5p represses cell proliferation, migration, invasion, and tumor neurogenesis by inhibition of NR6A1/E-cadherin signaling axis, which may be a potential target for diagnosis and therapy of TGCTs.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , MicroRNAs/metabolismo , Neoplasias Embrionárias de Células Germinativas/metabolismo , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Neoplasias Testiculares/metabolismo , Animais , Antígenos CD/genética , Caderinas/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , DNA (Citosina-5-)-Metiltransferase 1/genética , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , Invasividade Neoplásica , Neoplasias Embrionárias de Células Germinativas/genética , Neoplasias Embrionárias de Células Germinativas/patologia , Neurogênese , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/genética , Transdução de Sinais , Neoplasias Testiculares/genética , Neoplasias Testiculares/patologia
18.
Andrology ; 7(3): 319-328, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30786176

RESUMO

BACKGROUND: Golgi-associated PDZ and coiled-coil motif-containing protein (GOPC) is a Golgi protein that plays a role in vesicular transport and intracellular protein trafficking and degradation. Mice deficient in GOPC protein have globozoospermia and are infertile. The germ cell nuclear factor (GCNF) is a member of the nuclear receptor superfamily which is expressed in male germ cells, from spermatocytes and spermatids, both in the nucleus and the acrosomal region. It is not known if its expression could be altered in Gopc-/- mice with defective acrosomes. OBJECTIVES: The aim of the present work was to analyze the distribution of GCNF protein in spermatids of Gopc-/- knockout mice. MATERIALS AND METHODS: We have analyzed the expression and distribution during spermatogenesis of GCNF and its deregulation in Gopc-/- mutant mice by RT-qPCR, Western blot, immunohistochemistry and immunogold. RESULTS: Germ cell nuclear factor was localized in the nucleus of all the cell types in the seminiferous tubules. Despite being a nuclear protein, it was also located in the acrosome and in the manchette of elongating spermatids. We have found that in the absence of GOPC, the expression of GCNF was increased in the nucleus of spermatocytes, mainly in leptotene, and in the nucleus and the manchette during the spermatid elongation. DISCUSSION AND CONCLUSION: Gopc-/- mice have defective acrosome and manchette. The manchette is involved in the transport of proteins through the cytoplasm and the nucleus. Considering that the GCNF protein is normally transported to the acrosome and the nucleus, it can be thought that transport deficiencies in Gopc-/- mice are responsible for the increased expression of this protein.


Assuntos
Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Espermátides/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Western Blotting , Núcleo Celular/metabolismo , Proteínas da Matriz do Complexo de Golgi/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Espermátides/ultraestrutura , Espermatogênese , Testículo/metabolismo
19.
PLoS One ; 12(2): e0170760, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28151980

RESUMO

Lhx8 is an important transcription factor that is preferentially expressed in germ cells. Lhx8 null mice are infertile due to lack of oocytes and impairment of the transition from primordial follicles to primary follicles. Lhx8 deficiency also affects the expression of many important oocyte-specific genes. In this study, we report the characterization of rainbow trout lhx8 genes and identification of a novel germ cell-specific nuclear factor that interacts with Lhx8. Two lhx8 genes, lhx8a and lhx8b, were identified, encoding proteins of 344 and 361 amino acids, respectively. The two proteins share 83% sequence identity and both transcripts are specifically expressed in the ovary. Quantitative real time PCR analysis demonstrated that both genes are expressed highly in pre-vitellogenic ovaries as well as in early stage embryos. Using a yeast two-hybrid screening system, a novel protein (Borealin-2) interacting with Lhx8 was identified. The interaction between either Lhx8a or Lhx8b and Borealin-2 was further confirmed by a bimolecular fluorescence complementation (BiFC) assay. Borealin-2 is a protein of 255 amino acids containing an Nbl1 domain, and its mRNA expression is restricted to the ovary and testis. A GFP reporter assay revealed that Borealin-2 is a nuclear protein. Collectively, results indicate that both Lhx8a and Lhx8b function through interaction with Borealin-2, which may play an important role during oogenesis and early embryogenesis in rainbow trout.


Assuntos
Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Proteínas com Homeodomínio LIM/genética , Proteínas com Homeodomínio LIM/metabolismo , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/genética , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Oncorhynchus mykiss/genética , Oncorhynchus mykiss/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Feminino , Proteínas de Peixes/química , Proteínas com Homeodomínio LIM/química , Masculino , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/química , Oncorhynchus mykiss/embriologia , Oogênese/genética , Domínios e Motivos de Interação entre Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição/química , Transcriptoma , Técnicas do Sistema de Duplo-Híbrido
20.
J Mol Biol ; 428(24 Pt B): 4981-4992, 2016 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-27984042

RESUMO

Oct4 is a transcription factor required for maintaining pluripotency and self-renewal in stem cells. Prior to differentiation, Oct4 must be silenced to allow for the development of the three germ layers in the developing embryo. This fine-tuning is controlled by the nuclear receptors (NRs), liver receptor homolog-1 (LRH-1) and germ cell nuclear factor (GCNF). Liver receptor homolog-1 is responsible for driving the expression of Oct4 where GCNF represses its expression upon differentiation. Both receptors bind to a DR0 motif located within the Oct4 promoter. Here, we present the first structure of mouse GCNF DNA-binding domain in complex with the Oct4 DR0. The overall structure revealed two molecules bound in a head-to-tail fashion on opposite sides of the DNA. Additionally, we solved the structure of the human LRH-1 DNA-binding domain bound to the same element. We explore the structural elements that govern Oct4 recognition by these two NRs.


Assuntos
Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/química , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Fator 3 de Transcrição de Octâmero/biossíntese , Fator 3 de Transcrição de Octâmero/química , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Cristalografia por Raios X , DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Modelos Moleculares , Regiões Promotoras Genéticas , Ligação Proteica , Conformação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA