Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Anim Sci ; 1012023 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-37703114

RESUMEN

The high-fertility Meishan pig is currently categorized into medium sized (MMS) and small sized (SMS) based on body size. To identify causal genes responsible for the variation in body size within the two categories, we sequenced individuals representing the entire consanguinity of the existing Meishan pig. This enabled us to conduct genome selective signal analysis. Our findings revealed the genomes of MMS and SMS are stratified, with selective sweep regions formed by differential genomic intervals between the two categories enriched in multiple pig body size related quantitative trait loci (QTLs). Furthermore, the missense mutation c.575T > C of candidate causal gene NR6A1, accounting for the variation in lumbar vertebrae number in pigs, was positively selected in MMS only, leading to an increase in body length of MMS at 6 months of age. To precisely identify causal genes accounting for body size variation through multi-omics, we collected femoral cartilage and liver transcription data from MMS and SMS respectively, and re-sequencing data from pig breeds exhibiting varying body sizes. We found that two selected regions where the RSAD2-CMPK2 and COL3A1 genes are located, respectively, showed different haplotypes in pig breeds of varying body size, and was associated with body or carcass length in hybridized Suhuai pig. Additionally, the above three hub genes, were significantly greater expressed in SMS femoral cartilage and liver tissues compared to MMS. These three genes could strengthen the pathways related to bone resorption and metabolism in SMS, potentially hindering bone and skeletal development and resulting in a smaller body size in SMS. These findings provide valuable insights into the genetic mechanism of body size variation in Meishan pig population.


The existing well-known Meishan pig population has been categorized into medium sized (MMS), and small sized (SMS) based on body size, which is a result of artificial selection. MMS is relatively large in all body size traits, but otherwise have highly similar appearance and performance traits. To effectively identify the candidate selected genes that contribute to the body size variation in Meishan pigs, this study collected individuals from all lineages of MMS and SMS for re-sequencing. Additionally, femoral cartilage and liver transcription data were collected from MMS and SMS, respectively, and re-sequencing data from pig breeds exhibiting varying body sizes were also analyzed. Through multi-omics analysis, it was discovered that the missense mutation c.575T > C in the candidate causal gene NR6A1 was positively selected in MMS only, leading to an increase in the body length of MMS at 6 months of age. Moreover, the selected genes RSAD2-CMPK2 and COL3A1 were found to be significantly greater expressed in SMS femoral cartilage and liver tissues compared with MMS. These genes could potentially strengthen bone resorption and metabolism-related pathways in SMS. These findings contribute to a better understanding of the genetic mechanisms underlying body size variation in Meishan pigs and Chinese indigenous pigs.


Asunto(s)
Colágeno Tipo III , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares , Nucleósido-Fosfato Quinasa , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Sitios de Carácter Cuantitativo , Proteína Viperina , Animales , Secuencia de Bases , Tamaño Corporal/genética , Colágeno Tipo III/genética , Haplotipos , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/genética , Porcinos/genética , Proteína Viperina/genética , Sus scrofa , Nucleósido-Fosfato Quinasa/genética
2.
Cancer Sci ; 112(12): 4957-4967, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34689384

RESUMEN

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.


Asunto(s)
Cocarcinogénesis/genética , Regulación Neoplásica de la Expresión Génica , Laminina/genética , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/genética , Proteínas de Fusión Oncogénica/genética , Neoplasias Ováricas/genética , Animales , Línea Celular Tumoral , Cocarcinogénesis/metabolismo , Femenino , Humanos , Laminina/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Interferencia de ARN , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
3.
Exp Physiol ; 105(12): 2141-2153, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33006200

RESUMEN

NEW FINDINGS: What is the central question of this study? Does hsa_circ_001653 influence the development of gastric cancer (GC) and if so how? What is the main finding and its importance? Bioinformatics analysis revealed the presence of differentially expressed hsa_circ_001653 in GC and adjacent normal tissues, and this was strongly related to the pathology of patients with GC. Knockdown of hsa_circ_001653 suppressed the proliferation, invasion and migration of GC cells, while inducing cell apoptosis via miR-377-mediated NR6A1 inhibition. The effect of hsa_circ_001653 and miR-377 on tumour growth in GC was further confirmed in vivo. ABSTRACT: Gastric cancer (GC) is one of the leading causes of human mortality through malignant tumours. Circular RNAs (circRNAs) have been identified as binding to microRNAs (miRNAs) to modulate the progression of tumours. This study explores the role of hsa_circ_001653, a newly identified circRNA, in the development of GC. hsa_circ_001653 expression was measured in 86 paired normal and tumour tissues surgically resected from GC patients. Cross-talk between hsa_circ_001653 and microRNA-377 (miR-377)/nuclear receptor subfamily 6, group A, member 1 (NR6A1) was assessed using bioinformatics analysis, dual-luciferase reporter assay, Ago2 immunoprecipitation and western blot analysis. A series of functional experiments were carried out to elucidate the role of hsa_circ_001653 in GC cell proliferation, invasion, migration and apoptosis, and its underlying molecular mechanisms. Nude mice were inoculated with GC cells for in vivo analysis. hsa_circ_001653 was found to be an up-regulated circRNA in GC tissues and cells. Down-regulation of hsa_circ_001653 inhibited GC cell proliferation, migration and invasion, while stimulating cell apoptosis. hsa_circ_001653 was found to bind to miR-377, which targeted NR6A1 and repressed its expression. Inhibition of miR-377 and overexpression of NR6A1 restored the proliferation, migration and invasion in GC cells lacking hsa_circ_001653. Furthermore, inhibition of hsa_circ_001653 attenuated tumour growth in nude mice inoculated with GC cells. Collectively, the demonstration that hsa_circ_001653 exerts its anticancer effects by regulating the miR-377-NR6A1 axis increases our understanding of gastric cancer pathophysiology. The findings uncover new potential therapeutic targets for GC.


Asunto(s)
MicroARNs/genética , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/genética , ARN Circular/genética , Neoplasias Gástricas/genética , Regulación hacia Arriba/genética , Animales , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Progresión de la Enfermedad , Regulación hacia Abajo/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Estómago/patología , Neoplasias Gástricas/patología , Activación Transcripcional/genética
4.
Cancer Med ; 9(23): 9107-9122, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33034957

RESUMEN

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.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , MicroARNs/metabolismo , Neoplasias de Células Germinales y Embrionarias/metabolismo , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/metabolismo , Neoplasias Testiculares/metabolismo , Animales , Antígenos CD/genética , Cadherinas/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Invasividad Neoplásica , Neoplasias de Células Germinales y Embrionarias/genética , Neoplasias de Células Germinales y Embrionarias/patología , Neurogénesis , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/genética , Transducción de Señal , Neoplasias Testiculares/genética , Neoplasias Testiculares/patología
5.
Cell Commun Signal ; 17(1): 77, 2019 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-31315616

RESUMEN

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. .


Asunto(s)
Metabolismo de los Lípidos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/metabolismo , Animales , Movimiento Celular , Proliferación Celular , Regulación de la Expresión Génica , Silenciador del Gen , Células HEK293 , Células Hep G2 , Humanos , Insulina/metabolismo , Lipogénesis , MicroARNs/genética , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/deficiencia , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/genética , Receptor de Insulina/genética , Transducción de Señal
6.
Genes Genomics ; 41(10): 1165-1171, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31302865

RESUMEN

INTRODUCTION: The vertebral number is an economically significant trait, which is associated with body length and carcass traits. Nuclear Receptor Subfamily 6, Group A, Member 1 (NR6A1) is a member of the nuclear receptor superfamily and it plays an important role in the early development of embryos. OBJECTIVES: The NR6A1 gene was considered as an important candidate for influence vertebrae number, while the potential associations between this gene and the number of lumbar vertebrae traits of sheep have not been explored. METHODS: In this study, we detected the genetic variants of NR6A1 gene and analyzed the associations of the polymorphisms with lumbar number traits in 130 Kazakh sheep. We use single-strand conformation polymorphism (SSCP) technique to detect single nucleotide polymorphism (SNP) of NR6A1 gene, and the association of the genotype and lumbar number variation was analyzed by independent Chi-square test. RESULTS: We detect SNP of NR6A1 gene by PCR-SSCP technique, and polymorphisms were only found in the coding region of exon-6 and exon-8 of NR6A1 gene. In order to investigate the connection between the SNP locus and lumbar number traits in sheep, we conducted a Chi-square test for independence for exon-6 and exon-8 of NR6A1 gene, respectively. Association analysis revealed significant associations between the SNP (rs414302710: A >C) in the exon-8 of NR6A1 gene with the number of lumbar vertebrae (P < 0.01). CONCLUSION: Our study indicated that this SNP (rs414302710: A>C) locus of exon-8 of NR6A1 gene in sheep possible influence the number of lumbar vertebrae, which has the potential to be applied in selective breeding of sheep.


Asunto(s)
Vértebras Lumbares , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/genética , Fenotipo , Polimorfismo de Nucleótido Simple , Ovinos/genética , Animales , Secuencia de Bases , Cruzamiento , Exones/genética , Genotipo , Análisis de Secuencia
7.
Anim Genet ; 50(2): 166-171, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30741434

RESUMEN

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.


Asunto(s)
Domesticación , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/genética , Receptor de Melanocortina Tipo 1/genética , Sus scrofa/genética , Alelos , Animales , Cruzamiento , Europa Oriental , Italia , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/metabolismo , Receptor de Melanocortina Tipo 1/metabolismo
9.
J Cell Biochem ; 119(3): 2719-2730, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29057499

RESUMEN

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.


Asunto(s)
Carcinoma Embrionario/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/metabolismo , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Elementos de Respuesta , Carcinoma Embrionario/genética , Carcinoma Embrionario/patología , Línea Celular Tumoral , Humanos , Proteínas de Neoplasias/genética , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/genética , Factor 3 de Transcripción de Unión a Octámeros/genética
10.
Cancer ; 123(13): 2472-2481, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28222251

RESUMEN

BACKGROUND: Aberrant DNA methylation is known to occur in patients with acute myeloid leukemia (AML), whereas methylation signatures and prognostic markers have been proposed. The objective of the current study was to evaluate all CpG sites of the genome and identify prognostic methylation markers for overall survival in patients with AML with normal karyotype (AML-NK). METHODS: AML-NK samples from 7 SWOG trials were analyzed using a novel genome-wide approach called "CHARMcox" (comprehensive high-throughput array-based relative methylation analysis combined with the Cox proportional hazards model) controlling for known clinical covariates. CHARMcox was applied to a phase 1 discovery cohort (72 patients) to identify survival-associated methylation regions (SAMRs). Subsequently, using bisulfite pyrosequencing, SAMRs were studied in phase 2 model-building (65 patients) and phase 3 validation (65 patients) cohorts. An independent external cohort from The Cancer Genome Atlas (TCGA) AML study (LAML) was used for further validation (93 patients). RESULTS: Two SAMRs, located at the CpG island shores of leucine zipper tumor suppressor 2 (LZTS2) and nuclear receptor subfamily 6 group a member 1 (NR6A1), respectively, were identified. Multivariable analyses demonstrated that hypomethylation of either LZTS2 or NR6A1 was associated with worse overall survival in the SWOG cohort (P<.001). The prognosis was validated in patients with AML-NK from the TCGA-LAML cohort. Methylation values below the median at both markers predicted worse overall survival (SWOG: hazard ratio, 1.89 [P<.001]; and TCGA-LAML: hazard ratio, 2.08 [P=.006]). The C-statistic was 0.71 for both cohorts, and the impact was independent of the Fms-related tyrosine kinase 3 internal tandem duplication (FLT3-ITD) status. CONCLUSIONS: The 2 methylation markers, measurable by clinically applicable assays such as bisulfite pyrosequencing, are promising for risk stratification among patients with AML-NK. Cancer 2017;123:2472-81. © 2017 American Cancer Society.


Asunto(s)
Proteínas de Ciclo Celular/genética , Metilación de ADN/genética , Proteínas de Unión al ADN/genética , Leucemia Mieloide Aguda/genética , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/genética , Proteínas Supresoras de Tumor/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Causas de Muerte , Islas de CpG/genética , Femenino , Humanos , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/terapia , Masculino , Persona de Mediana Edad , Análisis Multivariante , Pronóstico , Modelos de Riesgos Proporcionales , Tasa de Supervivencia , Adulto Joven
11.
PLoS One ; 12(2): e0170760, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28151980

RESUMEN

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.


Asunto(s)
Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/genética , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/metabolismo , Oncorhynchus mykiss/genética , Oncorhynchus mykiss/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Femenino , Proteínas de Peces/química , Proteínas con Homeodominio LIM/química , Masculino , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/química , Oncorhynchus mykiss/embriología , Oogénesis/genética , Dominios y Motivos de Interacción de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción/química , Transcriptoma , Técnicas del Sistema de Dos Híbridos
12.
Nanomedicine ; 13(4): 1363-1375, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28219741

RESUMEN

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.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Doxiciclina/farmacología , Regulación de la Expresión Génica , Nanopartículas/química , Neurotensina/química , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/genética , Animales , Línea Celular Tumoral , Vectores Genéticos , Humanos , Masculino , Ratones , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Plásmidos , Regiones Promotoras Genéticas , Ratas , Ratas Wistar , Elementos de Respuesta , Transfección , Transgenes
13.
J Biol Chem ; 291(16): 8644-52, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26769970

RESUMEN

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.


Asunto(s)
Diferenciación Celular/fisiología , Regulación de la Expresión Génica/fisiología , Células Madre Embrionarias Humanas/metabolismo , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Proteínas Represoras/metabolismo , Línea Celular , Células Madre Embrionarias Humanas/citología , Humanos , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas Represoras/genética
14.
Genet Mol Res ; 14(4): 12723-32, 2015 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-26505423

RESUMEN

Mutations in melanocortin 1 receptor (MC1R) gene and v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog (KIT) gene have been shown to affect coat color patterns in pigs. Additional functional marker genes, such as insulin like growth factor-2 (IGF2) and orphan nuclear receptor, germ cell nuclear factor (NR6A1), have been described for variations in factors such as fat deposition, litter size, and vertebra number in pigs. In this study, we investigated 129 pigs representing 4 breeds: Thai indigenous, classified into black (similar to Raad or Ka done pig) and black and white (similar to the Hailum and Kwai pig) coat color types; wild boar; Duroc; and Chinese Meishan. Mutations of MC1R, KIT, IGF2, and NR6A1 were detected using polymerase chain reaction-restriction fragment length polymorphism. The genotypes variation in MC1R and KIT genes could be used to differentiate four groups of coat color: solid black, black and white, red, and wild type. For IGF2, the GG genotype was present in wild boar only; for NR6A1 the TT genotype was found only in Duroc pigs. We identified novel 14-bp deletions in KIT that were associated with black and white coat color in Thai indigenous pigs. Insights into variations in genes presented in this study will be useful in future developmental breeding programs for the Thai native pig.


Asunto(s)
Factor II del Crecimiento Similar a la Insulina/genética , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/genética , Proteínas Proto-Oncogénicas c-kit/genética , Receptor de Melanocortina Tipo 1/genética , Animales , Genotipo , Reacción en Cadena de la Polimerasa , Polimorfismo de Longitud del Fragmento de Restricción/genética , Sus scrofa , Porcinos
15.
Mol Endocrinol ; 29(12): 1792-804, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26484580

RESUMEN

Gonadotropin-regulated testicular RNA helicase (GRTH) (GRTH/DDX25), is a testis-specific protein essential for completion of spermatogenesis. Transgenic mice carrying 5'-flanking regions of the GRTH gene/green fluorescence protein (GFP) reporter revealed a region (-6.4/-3.6 kb) which directs its expression in germ cells (GCs) via androgen action. This study identifies a functional cis-binding element on the GRTH gene for GC nuclear factor (GCNF) (GCNF/RTR) required to regulate GRTH gene expression in postmeiotic testis GCs and explore the action of androgen on GCNF and GRTH transcription/expression. GCNF expression decreased in mice testis upon flutamide (androgen receptor antagonist) treatment, indicating the presence of an androgen/GCNF network to direct GRTH expression in GC. Binding studies and chromatin immunoprecipitation demonstrated specific association of GCNF to a consensus half-site (-5270/-5252) of the GRTH gene in both round spermatids and spermatocytes, which was abolished by flutamide treatment in round spermatids. Moreover, flutamide treatment of wild-type mice caused selective reduction of GCNF and GRTH in round spermatids. GCNF knock-down in seminiferous tubules from GRTH-transgenic mice (dark zone, round spermatid rich) caused decreased GFP expression. Exposure of tubules to flutamide caused decrease in GCNF and GFP expression, whereas androgen exposure induced significant increase. Our studies provide evidence for actions of androgen on GCNF cell-specific regulation of GRTH expression in GC. GRTH associates with GCNF mRNA, its absence caused increase on GCNF expression and mRNA stability indicative of a negative autocrine regulation of GCNF by GRTH. These in vivo/in vitro models link androgen actions to GC through GCNF, as regulated transfactor that controls transcription/expression of GRTH.


Asunto(s)
Andrógenos/metabolismo , ARN Helicasas DEAD-box/genética , Gonadotropinas/metabolismo , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/metabolismo , Testículo/metabolismo , Animales , ARN Helicasas DEAD-box/metabolismo , Masculino , Ratones , Ratones Transgénicos , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/genética , Espermátides/metabolismo , Espermatocitos/metabolismo , Espermatogénesis/genética , Espermatogénesis/fisiología
16.
Meat Sci ; 98(4): 781-4, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25134014

RESUMEN

Wild boar meat cannot be easily distinguished from domestic pig meat, especially in processed products, thus it can be fraudulently substituted with cheaper domestic pork. In this study we genotyped polymorphisms in two genes (MC1R, affecting coat color and NR6A1, associated with number of vertebrae) in 293 domestic pigs of five commercial breeds, 111 wild boars sampled in Italy, and 90 in Slovenia and other Western Balkan regions. Allele and genotype frequency data were used to set up a DNA-based method to distinguish meat of wild boars and domestic pigs. Genotyping results indicated that domesticated genes were introgressed into wild boar populations. This complicated the determination of the origin of the meat and would cause a high error rate if markers of only one gene were used. The combined use of polymorphisms in the two analyzed genes substantially reduced false negative results.


Asunto(s)
Carne , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/genética , Polimorfismo Genético/genética , Receptor de Melanocortina Tipo 1/genética , Animales , Femenino , Marcadores Genéticos/genética , Masculino , Reacción en Cadena de la Polimerasa , Análisis de Secuencia de ADN , Sus scrofa , Porcinos
17.
PLoS One ; 9(8): e103985, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25140725

RESUMEN

Expression of germ cell nuclear factor (GCNF; Nr6a1), an orphan member of the nuclear receptor gene family of transcription factors, during gastrulation and neurulation is critical for normal embryogenesis in mice. Gcnf represses the expression of the POU-domain transcription factor Oct4 (Pou5f1) during mouse post-implantation development. Although Gcnf expression is not critical for the embryonic segregation of the germ cell lineage, we found that sexually dimorphic expression of Gcnf in germ cells correlates with the expression of pluripotency-associated genes, such as Oct4, Sox2, and Nanog, as well as the early meiotic marker gene Stra8. To elucidate the role of Gcnf during mouse germ cell differentiation, we generated an ex vivo Gcnf-knockdown model in combination with a regulated CreLox mutation of Gcnf. Lack of Gcnf impairs normal spermatogenesis and oogenesis in vivo, as well as the derivation of germ cells from embryonic stem cells (ESCs) in vitro. Inactivation of the Gcnf gene in vivo leads to loss of repression of Oct4 expression in both male and female gonads.


Asunto(s)
Gametogénesis/fisiología , Gónadas/crecimiento & desarrollo , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/metabolismo , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Gónadas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo
18.
Semin Cell Dev Biol ; 24(10-12): 679-86, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24029702

RESUMEN

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.


Asunto(s)
Desarrollo Embrionario , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/metabolismo , Secuencia de Aminoácidos , Animales , ADN/metabolismo , Humanos , Datos de Secuencia Molecular , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/química , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/genética , Multimerización de Proteína , Proteínas Represoras/metabolismo , Elementos de Respuesta/genética
19.
Genes Dev ; 27(8): 941-54, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23630078

RESUMEN

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.


Asunto(s)
Fibroblastos/citología , Regulación de la Expresión Génica , MicroARNs/genética , MicroARNs/metabolismo , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/genética , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Línea Celular , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Fibroblastos/metabolismo , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Unión Proteica , Ribonucleasa III/genética , Ribonucleasa III/metabolismo
20.
Prostate ; 73(10): 1103-14, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23532770

RESUMEN

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.


Asunto(s)
Biomarcadores de Tumor/genética , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/genética , Próstata/metabolismo , Neoplasias de la Próstata/genética , Testículo/metabolismo , Anciano , Biomarcadores de Tumor/metabolismo , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/metabolismo , Orquiectomía , Próstata/patología , Próstata/cirugía , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/cirugía , Testículo/patología , Testículo/cirugía , Análisis de Matrices Tisulares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...