Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
BMC Genomics ; 24(1): 252, 2023 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-37165305

RESUMEN

BACKGROUND: The long non-coding RNAs (lncRNAs) are critical regulators of diverse biological processes. Nevertheless, a global view of its expression and function in the mouse retina, a crucial model for neurogenesis study, still needs to be made available. RESULTS: Herein, by integrating the established gene models and the result from ab initio prediction using short- and long-read sequencing, we characterized 4,523 lncRNA genes (MRLGs) in developing mouse retinas (from the embryonic day of 12.5 to the neonatal day of P28), which was so far the most comprehensive collection of retinal lncRNAs. Next, derived from transcriptomics analyses of different tissues and developing retinas, we found that the MRLGs were highly spatiotemporal specific in expression and played essential roles in regulating the genesis and function of mouse retinas. In addition, we investigated the expression of MRLGs in some mouse mutants and revealed that 97 intergenic MRLGs might be involved in regulating differentiation and development of retinal neurons through Math5, Isl1, Brn3b, NRL, Onecut1, or Onecut2 mediated pathways. CONCLUSIONS: In summary, this work significantly enhanced our knowledge of lncRNA genes in mouse retina development and provided valuable clues for future exploration of their biological roles.


Asunto(s)
ARN Largo no Codificante , Animales , Ratones , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Retina/metabolismo , Perfilación de la Expresión Génica , Factor Nuclear 6 del Hepatocito/genética , Factor Nuclear 6 del Hepatocito/metabolismo
2.
Commun Biol ; 4(1): 1298, 2021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34789845

RESUMEN

Cell type specification during pancreatic development is tightly controlled by a transcriptional and epigenetic network. The precise role of most transcription factors, however, has been only described in mice. To convey such concepts to human pancreatic development, alternative model systems such as pancreatic in vitro differentiation of human pluripotent stem cells can be employed. Here, we analyzed stage-specific RNA-, ChIP-, and ATAC-sequencing data to dissect transcriptional and regulatory mechanisms during pancreatic development. Transcriptome and open chromatin maps of pancreatic differentiation from human pluripotent stem cells provide a stage-specific pattern of known pancreatic transcription factors and indicate ONECUT1 as a crucial fate regulator in pancreas progenitors. Moreover, our data suggest that ONECUT1 is also involved in preparing pancreatic progenitors for later endocrine specification. The dissection of the transcriptional and regulatory circuitry revealed an important role for ONECUT1 within such network and will serve as resource to study human development and disease.


Asunto(s)
Factor Nuclear 6 del Hepatocito/genética , Páncreas/fisiología , Diferenciación Celular , Línea Celular , Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 6 del Hepatocito/metabolismo , Células Madre Embrionarias Humanas , Humanos , Transcripción Genética
3.
Biomolecules ; 11(8)2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34439790

RESUMEN

The identification of the molecular mechanisms controlling early cell fate decisions in mammals is of paramount importance as the ability to determine specific lineage differentiation represents a significant opportunity for new therapies. Pancreatic Progenitor Cells (PPCs) constitute a regenerative reserve essential for the maintenance and regeneration of the pancreas. Besides, PPCs represent an excellent model for understanding pathological pancreatic cellular remodeling. Given the lack of valid markers of early endoderm, the identification of new ones is of fundamental importance. Both products of the Ink4a/Arf locus, in addition to being critical cell-cycle regulators, appear to be involved in several disease pathologies. Moreover, the locus' expression is epigenetically regulated in ES reprogramming processes, thus constituting the ideal candidates to modulate PPCs homeostasis. In this study, starting from mouse embryonic stem cells (mESCs), we analyzed the early stages of pancreatic commitment. By inducing mESCs commitment to the pancreatic lineage, we observed that both products of the Cdkn2a locus, Ink4a and Arf, mark a naïve pancreatic cellular state that resembled PPC-like specification. Treatment with epi-drugs suggests a role for chromatin remodeling in the CDKN2a (Cycline Dependent Kinase Inhibitor 2A) locus regulation in line with previous observations in other cellular systems. Our data considerably improve the comprehension of pancreatic cellular ontogeny, which could be critical for implementing pluripotent stem cells programming and reprogramming toward pancreatic lineage commitment.


Asunto(s)
Linaje de la Célula/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Epigénesis Genética , Expresión Génica , Células Secretoras de Insulina/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Sitios Genéticos , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/metabolismo , Factor Nuclear 6 del Hepatocito/genética , Factor Nuclear 6 del Hepatocito/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Células Secretoras de Insulina/citología , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Páncreas/citología , Páncreas/metabolismo , Cultivo Primario de Células , Transactivadores/genética , Transactivadores/metabolismo
4.
Development ; 148(9)2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33929509

RESUMEN

The vertebrate retina is generated by retinal progenitor cells (RPCs), which produce >100 cell types. Although some RPCs produce many cell types, other RPCs produce restricted types of daughter cells, such as a cone photoreceptor and a horizontal cell (HC). We used genome-wide assays of chromatin structure to compare the profiles of a restricted cone/HC RPC and those of other RPCs in chicks. These data nominated regions of regulatory activity, which were tested in tissue, leading to the identification of many cis-regulatory modules (CRMs) active in cone/HC RPCs and developing cones. Two transcription factors, Otx2 and Oc1, were found to bind to many of these CRMs, including those near genes important for cone development and function, and their binding sites were required for activity. We also found that Otx2 has a predicted autoregulatory CRM. These results suggest that Otx2, Oc1 and possibly other Onecut proteins have a broad role in coordinating cone development and function. The many newly discovered CRMs for cones are potentially useful reagents for gene therapy of cone diseases.


Asunto(s)
Disección , Factor Nuclear 6 del Hepatocito/metabolismo , Factores de Transcripción Otx/metabolismo , Retina/crecimiento & desarrollo , Células Fotorreceptoras Retinianas Conos/metabolismo , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Pollos , Cromatina , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Factor Nuclear 6 del Hepatocito/genética , Factores de Transcripción Otx/genética , Retina/metabolismo , Células Madre
5.
Gastric Cancer ; 24(1): 103-116, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32705446

RESUMEN

BACKGROUND: Gastric intestinal metaplasia (IM) is considered a precancerous lesion, and bile acids (BA) play a critical role in the induction of IM. Ectopic expression of HNF4α was observed in a BA-induced IM cell model. However, the mechanisms underlying the upregulation of the protein in IM cells remains to be elucidated. METHODS: The effects of HNF4α on gastric mucosal cells in vivo were identified by a transgenic mouse model and RNA-seq was used to screen downstream targets of deoxycholic acid (DCA). The expression of pivotal molecules and miR-1 was detected by immunohistochemistry and in situ hybridization in normal, gastritis and IM tissue slides or microarrays. The transcriptional regulation of HDAC6 was investigated by chromatin immunoprecipitation (ChIP) and luciferase reporter assays. RESULTS: The transgenic mouse model validated that HNF4α stimulated the HDAC6 expression and mucin secretion in gastric mucosa. Increased HDAC6 and HNF4α expression was also detected in the gastric IM cell model and patient specimens. HNF4α could bind to and activate HDAC6 promoter. In turn, HDAC6 enhanced the HNF4α protein level in GES-1 cells. Furthermore, miR-1 suppressed the expression of downstream intestinal markers by targeting HDAC6 and HNF4α. CONCLUSIONS: Our findings show that the HDAC6/HNF4α loop regulated by miR-1 plays a critical role in gastric IM. Blocking the activation of this loop could be a potential approach to preventing BA-induced gastric IM or even gastric cancer (GC).


Asunto(s)
Mucosa Gástrica/patología , Factor Nuclear 6 del Hepatocito/metabolismo , Histona Desacetilasa 6/metabolismo , MicroARNs/metabolismo , Neoplasias Gástricas/genética , Animales , Ácidos y Sales Biliares/metabolismo , Modelos Animales de Enfermedad , Mucosa Gástrica/metabolismo , Gastritis/genética , Regulación de la Expresión Génica/genética , Humanos , Inmunohistoquímica , Hibridación in Situ , Metaplasia/genética , Ratones , Lesiones Precancerosas/genética , Regiones Promotoras Genéticas/genética , Neoplasias Gástricas/patología , Transcripción Genética/genética
6.
Bull Exp Biol Med ; 170(1): 40-45, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33222081

RESUMEN

Hepatocyte nuclear factor (HNF-6) is a liver-specific protein and a key component in the differentiation process during the development of mature liver. The immunohistochemical staining and RT-PCR techniques were employed to examine the expression of HNF-6 and proliferation of Ki-67+ cells during the early regeneration of the liver on postsurgery in 3, 6, 12, and 24 h in original model of partial hepatectomy in rats. The earliest proliferating (Ki-67+) cells were observed in 3 h after surgery in liver sinusoids (liver macrophages) and then in liver parenchyma. Expression of HNF-6 in hepatocytes and epithelial cells of the bile ducts attained maximum in 6 h after surgery. At later terms, this parameter somewhat decreased, but still surpassed the control level.


Asunto(s)
Factor Nuclear 6 del Hepatocito/genética , Hepatocitos/metabolismo , Macrófagos del Hígado/metabolismo , Regeneración Hepática/genética , Hígado/metabolismo , Animales , Conductos Biliares/metabolismo , Conductos Biliares/cirugía , Proliferación Celular , Femenino , Regulación de la Expresión Génica , Hepatectomía/métodos , Factor Nuclear 6 del Hepatocito/metabolismo , Hepatocitos/citología , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Macrófagos del Hígado/citología , Hígado/cirugía , Masculino , Ratas , Ratas Sprague-Dawley
7.
PLoS One ; 15(8): e0237403, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32790713

RESUMEN

Genome duplication leads to an emergence of gene paralogs that are essentially free to undergo the process of neofunctionalization, subfunctionalization or degeneration (gene loss). Onecut1 (Oc1) and Onecut2 (Oc2) transcription factors, encoded by paralogous genes in mammals, are expressed in precursors of horizontal cells (HCs), retinal ganglion cells and cone photoreceptors. Previous studies have shown that ablation of either Oc1 or Oc2 gene in the mouse retina results in a decreased number of HCs, while simultaneous deletion of Oc1 and Oc2 leads to a complete loss of HCs. Here we study the genetic redundancy between Oc1 and Oc2 paralogs and focus on how the dose of Onecut transcription factors influences abundance of individual retinal cell types and overall retina physiology. Our data show that reducing the number of functional Oc alleles in the developing retina leads to a gradual decrease in the number of HCs, progressive thinning of the outer plexiform layer and diminished electrophysiology responses. Taken together, these observations indicate that in the context of HC population, the alleles of Oc1/Oc2 paralogous genes are mutually interchangeable, function additively to support proper retinal function and their molecular evolution does not follow one of the typical routes after gene duplication.


Asunto(s)
Factor Nuclear 6 del Hepatocito/genética , Proteínas de Homeodominio/genética , Retina/fisiología , Factores de Transcripción/genética , Alelos , Células Amacrinas/metabolismo , Células Amacrinas/patología , Animales , Células Ependimogliales/metabolismo , Células Ependimogliales/patología , Ojo/crecimiento & desarrollo , Ojo/patología , Sitios Genéticos , Genotipo , Factor Nuclear 6 del Hepatocito/metabolismo , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Transgénicos , Retina/citología , Retina/patología , Células Bipolares de la Retina/metabolismo , Células Bipolares de la Retina/patología , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Conos/patología , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/metabolismo , Factores de Transcripción/metabolismo
8.
Neural Dev ; 15(1): 5, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-32192535

RESUMEN

BACKGROUND: The vertebrate retina consists of six major classes of neuronal cells. During development, these cells are generated from a pool of multipotent retinal progenitor cells (RPCs) that express the gene Vsx2. Fate-restricted RPCs have recently been identified, with limited mitotic potential and cell fate possibilities compared to multipotent RPCs. One population of fate-restricted RPCs, marked by activity of the regulatory element ThrbCRM1, gives rise to both cone photoreceptors and horizontal cells. These cells do not express Vsx2, but co-express the transcription factors (TFs) Onecut1 and Otx2, which bind to ThrbCRM1. The components of the gene regulatory networks that control the transition from multipotent to fate-restricted gene expression are not known. This work aims to identify and evaluate cis-regulatory elements proximal to Onecut1 to identify the gene regulatory networks involved in RPC fate-restriction. METHOD: We identified regulatory elements through ATAC-seq and conservation, followed by reporter assays to screen for activity based on temporal and spatial criteria. The regulatory elements of interest were subject to deletion and mutation analysis to identify functional sequences and evaluated by quantitative flow cytometry assays. Finally, we combined the enhancer::reporter assays with candidate TF overexpression to evaluate the relationship between the TFs, the enhancers, and early vertebrate retinal development. Statistical tests included ANOVA, Kruskal-Wallis, or unpaired t-tests. RESULTS: Two regulatory elements, ECR9 and ECR65, were identified to be active in ThrbCRM1(+) restricted RPCs. Candidate bHLH binding sites were identified as critical sequences in both elements. Overexpression of candidate bHLH TFs revealed specific enhancer-bHLH interactions. Nhlh1 overexpression expanded ECR65 activity into the Vsx2(+) RPC population, and overexpression of NeuroD1/NeuroG2/NeuroD4 had a similar effect on ECR9. Furthermore, bHLHs that were able to activate ectopic ECR9 reporter were able to induce endogenous Otx2 expression. CONCLUSIONS: This work reports a large-scale screen to identify spatiotemporally specific regulatory elements near the Onecut1 locus. These elements were used to identify distinct populations in the developing retina. In addition, fate-restricted regulatory elements responded differentially to bHLH factors, and suggest a role for retinal bHLHs upstream of the Otx2 and Onecut1 genes during the formation of restricted RPCs from multipotent RPCs.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 6 del Hepatocito/metabolismo , Factores de Transcripción Otx/metabolismo , Retina/metabolismo , Células Madre/metabolismo , Animales , Embrión de Pollo , Embrión de Mamíferos , Ratones
9.
J Mol Endocrinol ; 64(3): 165-179, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31990658

RESUMEN

Many sex differences in liver gene expression originate in the brain, depend on GH secretion and may underlie sex disparities in hepatic disease. Because epigenetic mechanisms may contribute, we studied promoter methylation and microRNA abundance in the liver, associated with expression of sexual dimorphic genes in mice with selective disruption of the dopamine D2 receptor in neurons (neuroDrd2KO), which decreases hypothalamic Ghrh, pituitary GH, and serum IGFI and in neonatally androgenized female mice which have increased pituitary GH content and serum IGFI. We evaluated mRNA levels of the female predominant genes prolactin receptor (Prlr), alcohol dehydrogenase 1 (Adh1), Cyp2a4, and hepatocyte nuclear transcription factor 6 (Hnf6) and the male predominant gene, Cyp7b1. Female predominant genes had higher mRNA levels compared to males, but lower methylation was only detected in the Prlr and Cyp2a4 female promoters. In neuroDrd2KO mice, sexual dimorphism was lost for all genes; the upregulation (feminization) of Prlr and Cyp2a4 in males correlated with decreased methylation of their promoters, and the downregulation (masculinization) of Hnf-6 mRNA in females correlated inversely with its promoter methylation. Neonatal androgenization of females evoked a loss of sexual dimorphism only for the female predominant Hnf6 and Adh1 genes, but no differences in promoter methylation were found. Finally, mmu-miR-155-5p, predicted to target Cyp7b1 expression, was lower in males in association with higher Cyp7b1 mRNA levels compared to females and was not modified in neuroDrd2KO or TP mice. Our results suggest specific regulation of gene sexually dimorphic expression in the liver by methylation or miRNAs.


Asunto(s)
Alcohol Deshidrogenasa/genética , Hidrocarburo de Aril Hidroxilasas/genética , Familia 2 del Citocromo P450/genética , Familia 7 del Citocromo P450/genética , Hormona del Crecimiento/farmacología , Factor Nuclear 6 del Hepatocito/genética , Receptores de Prolactina/genética , Esteroide Hidroxilasas/genética , Alcohol Deshidrogenasa/metabolismo , Animales , Hidrocarburo de Aril Hidroxilasas/metabolismo , Familia 2 del Citocromo P450/metabolismo , Familia 7 del Citocromo P450/metabolismo , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/fisiología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hormona del Crecimiento/metabolismo , Factor Nuclear 6 del Hepatocito/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas/genética , Receptores de Prolactina/metabolismo , Caracteres Sexuales , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Esteroide Hidroxilasas/metabolismo
10.
Nucleic Acids Res ; 47(11): 5587-5602, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31049588

RESUMEN

Remodeling of chromatin accessibility is necessary for successful reprogramming of fibroblasts to neurons. However, it is still not fully known which transcription factors can induce a neuronal chromatin accessibility profile when overexpressed in fibroblasts. To identify such transcription factors, we used ATAC-sequencing to generate differential chromatin accessibility profiles between human fibroblasts and iNeurons, an in vitro neuronal model system obtained by overexpression of Neurog2 in induced pluripotent stem cells (iPSCs). We found that the ONECUT transcription factor sequence motif was strongly associated with differential chromatin accessibility between iNeurons and fibroblasts. All three ONECUT transcription factors associated with this motif (ONECUT1, ONECUT2 and ONECUT3) induced a neuron-like morphology and expression of neuronal genes within two days of overexpression in fibroblasts. We observed widespread remodeling of chromatin accessibility; in particular, we found that chromatin regions that contain the ONECUT motif were in- or lowly accessible in fibroblasts and became accessible after the overexpression of ONECUT1, ONECUT2 or ONECUT3. There was substantial overlap with iNeurons, still, many regions that gained accessibility following ONECUT overexpression were not accessible in iNeurons. Our study highlights both the potential and challenges of ONECUT-based direct neuronal reprogramming.


Asunto(s)
Reprogramación Celular , Cromatina/genética , Regulación de la Expresión Génica , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/metabolismo , Factores de Transcripción Onecut/genética , Diferenciación Celular , Línea Celular , Cromatina/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Ontología de Genes , Factor Nuclear 6 del Hepatocito/genética , Factor Nuclear 6 del Hepatocito/metabolismo , Proteínas de Homeodominio , Humanos , Células Madre Pluripotentes Inducidas/citología , Neuronas/citología , Factores de Transcripción Onecut/metabolismo , Factores de Transcripción
11.
Cell Mol Gastroenterol Hepatol ; 7(4): 841-856, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30831323

RESUMEN

BACKGROUND & AIMS: The Onecut 1 transcription factor (Oc1, a.k.a. HNF6) promotes differentiation of endocrine and duct cells of the pancreas; however, it has no known role in acinar cell differentiation. We sought to better understand the role of Oc1 in exocrine pancreas development and to identify its direct transcriptional targets. METHODS: Pancreata from Oc1Δpanc (Oc1fl/fl;Pdx1-Cre) mouse embryos and neonates were analyzed morphologically. High-throughput RNA-sequencing was performed on control and Oc1-deficient pancreas; chromatin immunoprecipitation sequencing was performed on wild-type embryonic mouse pancreata to identify direct Oc1 transcriptional targets. Immunofluorescence labeling was used to confirm the RNA-sequencing /chromatin immunoprecipitation sequencing results and to further investigate the effects of Oc1 loss on acinar cells. RESULTS: Loss of Oc1 from the developing pancreatic epithelium resulted in disrupted duct and acinar cell development. RNA-sequencing revealed decreased expression of acinar cell regulatory factors (Nr5a2, Ptf1a, Gata4, Mist1) and functional genes (Amylase, Cpa1, Prss1, Spink1) at embryonic day (e) 18.5 in Oc1Δpanc samples. Approximately 1000 of the altered genes were also identified as direct Oc1 targets by chromatin immunoprecipitation sequencing, including most of the previously noted genes. By immunolabeling, we confirmed that Amylase, Mist1, and GATA4 protein levels are significantly decreased by P2, and Spink1 protein levels were significantly reduced and mislocalized. The pancreatic duct regulatory factors Hnf1ß and FoxA2 were also identified as direct Oc1 targets. CONCLUSIONS: These findings confirm that Oc1 is an important regulator of both duct and acinar cell development in the embryonic pancreas. Novel transcriptional targets of Oc1 have now been identified and provide clarity into the mechanisms of Oc1 transcriptional regulation in the developing exocrine pancreas. Oc1 can now be included in the gene-regulatory network of acinar cell regulatory genes. Oc1 regulates other acinar cell regulatory factors and acinar cell functional genes directly, and it can also regulate some acinar cell regulatory factors (eg, Mist1) indirectly. Oc1 therefore plays an important role in acinar cell development.


Asunto(s)
Diferenciación Celular , Factor Nuclear 6 del Hepatocito/metabolismo , Morfogénesis , Páncreas Exocrino/crecimiento & desarrollo , Páncreas Exocrino/patología , Células Acinares/patología , Animales , Animales Recién Nacidos , Secuencia de Bases , Proliferación Celular , Embrión de Mamíferos/patología , Epitelio/crecimiento & desarrollo , Epitelio/patología , Regulación del Desarrollo de la Expresión Génica , Ratones , Páncreas Exocrino/metabolismo
12.
J Cell Physiol ; 234(4): 3675-3684, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30256389

RESUMEN

Hepatocyte nuclear factor 6 (HNF6), as a transcription factor, has been reported to be involved in cell proliferation, carcinogenesis, and tumor metastasis. Here, we demonstrated the role of HNF6 in tumor growth and liver metastasis in colorectal cancer (CRC). Through bioinformatics and clinical samples analysis, we found HNF6 messenger RNA was upregulated both in CRC primary sites and liver metastases, and its high expression indicated poor survival in CRC patients. In vitro studies confirmed that HNF6 promoted cell proliferation and colony formation. What is more, in mouse models, the xenografts grew significantly faster and liver metastasis rate was nearly 45% higher in mice injected with HNF6-overexpressing cells. Further mechanism exploration showed that HNF6 expression affected cell adhesion and conferred resistance to anoikis in CRC cells. Taken together, HNF6 expression was upregulated in CRC and closely correlated with poor survival. HNF6 promoted CRC cell proliferation and tumor growth, and may contribute to liver metastasis via conferring cell resistance to anoikis.


Asunto(s)
Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Factor Nuclear 6 del Hepatocito/metabolismo , Neoplasias Hepáticas/metabolismo , Animales , Anoicis , Adhesión Celular , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 6 del Hepatocito/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/secundario , Ratones Endogámicos BALB C , Ratones Desnudos , Transducción de Señal , Carga Tumoral , Regulación hacia Arriba
13.
Neural Dev ; 13(1): 26, 2018 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-30466480

RESUMEN

BACKGROUND: Cone and rod photoreceptors are two of the primary cell types affected in human retinal disease. Potential strategies to combat these diseases are the use of gene therapy to rescue compromised photoreceptors or to generate new functional photoreceptors to replace those lost in the diseased retina. Cis-regulatory elements specific to cones, rods, or both types of photoreceptors are critical components of successful implementation of these two strategies. The purpose of this study was to identify and characterize the cell type specificity and activity of cis-regulatory elements active in developing photoreceptors. METHODS: Cis-regulatory elements were introduced into the developing chicken and mouse retina by electroporation. Characterization of reporter activity in relation with cell type markers was determined using confocal microscopy. In addition, two high-throughput flow cytometry assay were developed to assess whether these elements were downstream of Onecut1 in the photoreceptor specification network. RESULTS: The majority of cis-regulatory elements were active in both cone and rod photoreceptors and were largely uninfluenced by a Onecut1 dominant-negative construct. Elements associated with the Thrb, Nr2e3, and Rhodopsin genes showed highly enriched activity in cones or rods, and were affected by interference in Onecut1 signaling. Rhodopsin promoter activity was the most highly influenced by Onecut1 activity and its induction could be modulated by the Maf family transcription factor L-Maf. Nr2e3 elements were observed to have activity in cone photoreceptors and Nr2e3 protein was expressed in developing cone photoreceptors, suggesting a role for this predominant rod gene in cone photoreceptor development. CONCLUSIONS: The analysis presented here provides an experimental framework to determine the specificity and strength of photoreceptor elements within specific genetic networks during development. The Onecut1 transcription factor is one such factor that influences the gene regulatory networks specific to cones and rods, but not those that are common to both.


Asunto(s)
Factor Nuclear 6 del Hepatocito/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Animales , Pollos , Citometría de Flujo , Genes erbA , Factor Nuclear 6 del Hepatocito/genética , Ratones , Receptores Nucleares Huérfanos/genética , Receptores Nucleares Huérfanos/metabolismo , Retina/metabolismo , Rodopsina/genética , Rodopsina/metabolismo
14.
Cell Rep ; 20(7): 1533-1542, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28813666

RESUMEN

In the developing brain, neurons expressing VEGF-A and blood vessels grow in close apposition, but many of the molecular pathways regulating neuronal VEGF-A and neurovascular system development remain to be deciphered. Here, we show that miR-9 links neurogenesis and angiogenesis through the formation of neurons expressing VEGF-A. We found that miR-9 directly targets the transcription factors TLX and ONECUTs to regulate VEGF-A expression. miR-9 inhibition leads to increased TLX and ONECUT expression, resulting in VEGF-A overexpression. This untimely increase of neuronal VEGF-A signal leads to the thickening of blood vessels at the expense of the normal formation of the neurovascular network in the brain and retina. Thus, this conserved transcriptional cascade is critical for proper brain development in vertebrates. Because of this dual role on neural stem cell proliferation and angiogenesis, miR-9 and its downstream targets are promising factors for cellular regenerative therapy following stroke and for brain tumor treatment.


Asunto(s)
Corteza Cerebral/metabolismo , MicroARNs/genética , Neovascularización Fisiológica/genética , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Secuencia de Bases , Sitios de Unión , Diferenciación Celular , Proliferación Celular , Corteza Cerebral/crecimiento & desarrollo , Embrión no Mamífero , Feto , Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 6 del Hepatocito/genética , Factor Nuclear 6 del Hepatocito/metabolismo , Humanos , MicroARNs/metabolismo , Morfogénesis/genética , Células-Madre Neurales/citología , Neuronas/metabolismo , Neuronas/patología , Receptores Nucleares Huérfanos , Cultivo Primario de Células , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Retina/crecimiento & desarrollo , Retina/metabolismo , Transducción de Señal , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra
15.
Stem Cell Reports ; 8(6): 1659-1674, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28552606

RESUMEN

The loss of cone photoreceptors that mediate daylight vision represents a leading cause of blindness, for which cell replacement by transplantation offers a promising treatment strategy. Here, we characterize cone differentiation in retinas derived from mouse embryonic stem cells (mESCs). Similar to in vivo development, a temporal pattern of progenitor marker expression is followed by the differentiation of early thyroid hormone receptor ß2-positive precursors and, subsequently, photoreceptors exhibiting cone-specific phototransduction-related proteins. We establish that stage-specific inhibition of the Notch pathway increases cone cell differentiation, while retinoic acid signaling regulates cone maturation, comparable with their actions in vivo. MESC-derived cones can be isolated in large numbers and transplanted into adult mouse eyes, showing capacity to survive and mature in the subretinal space of Aipl1-/- mice, a model of end-stage retinal degeneration. Together, this work identifies a robust, renewable cell source for cone replacement by purified cell suspension transplantation.


Asunto(s)
Células Madre Embrionarias de Ratones/trasplante , Células Fotorreceptoras Retinianas Conos/citología , Degeneración Retiniana/terapia , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/antagonistas & inhibidores , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Diferenciación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Proteínas del Ojo/antagonistas & inhibidores , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Factor Nuclear 6 del Hepatocito/metabolismo , Factor Inhibidor de Leucemia/farmacología , Ratones , Ratones Noqueados , Células Madre Embrionarias de Ratones/citología , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Opsinas/metabolismo , Receptores Nucleares Huérfanos/antagonistas & inhibidores , Receptores Nucleares Huérfanos/genética , Receptores Nucleares Huérfanos/metabolismo , Factores de Transcripción Otx/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores Notch/antagonistas & inhibidores , Receptores Notch/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Degeneración Retiniana/patología , Transducción de Señal , Tretinoina/metabolismo , Tretinoina/farmacología
16.
Sci Rep ; 7: 44686, 2017 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-28304379

RESUMEN

The aim of the present study was to develop chick-embryo chorioallantoic membrane (CAM) bioluminescent tumor models employing low passage cell cultures obtained from primary pancreatic ductal adenocarcinoma (PDAC) cells. Primary PDAC cells transduced with lentivirus expressing Firefly-luciferase (Fluc) were established and inoculated onto the CAM membrane, with >80% engraftment. Fluc signal reliably correlated with tumor growth. Tumor features were evaluated by immunohistochemistry and genetic analyses, including analysis of mutations and mRNA expression of PDAC pivotal genes, as well as microRNA (miRNA) profiling. These studies showed that CAM tumors had histopathological and genetic characteristic comparable to the original tumors. We subsequently tested the modulation of key miRNAs and the activity of gemcitabine and crizotinib on CAM tumors, showing that combination treatment resulted in 63% inhibition of tumor growth as compared to control (p < 0.01). These results were associated with reduced expression of miR-21 and increased expression of miR-155. Our study provides the first evidence that transduced primary PDAC cells can form tumors on the CAM, retaining several histopathological and (epi)genetic characteristics of original tumors. Moreover, our results support the use of these models for drug testing, providing insights on molecular mechanisms underlying antitumor activity of new drugs/combinations.


Asunto(s)
Membrana Corioalantoides/metabolismo , Evaluación Preclínica de Medicamentos , Mediciones Luminiscentes/métodos , Modelos Biológicos , Neoplasias Pancreáticas/patología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Carcinogénesis/patología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Pollos , Crizotinib/farmacología , Crizotinib/uso terapéutico , Análisis Mutacional de ADN , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 6 del Hepatocito/genética , Factor Nuclear 6 del Hepatocito/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Análisis de Secuencia de ADN , Células Tumorales Cultivadas , Gemcitabina , Neoplasias Pancreáticas
17.
PLoS One ; 11(12): e0167085, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27936029

RESUMEN

BACKGROUND AND AIMS: Growth hormone (GH) not only supports hepatic metabolism but also protects against hepatocyte cell death. Hnf6 (or Oc1) belonging to the Onecut family of hepatocyte transcription factors known to regulate differentiated hepatic function, is a GH-responsive gene. We evaluate if GH mediates Hnf6 activity to attenuate hepatic apoptotic injury. METHODS: We used an animal model of hepatic apoptosis by bile duct ligation (BDL) with Hnf6 -/- (KO) mice in which hepatic Hnf6 was conditionally inactivated. GH was administered to adult wild type WT and KO mice for the 7 days of BDL to enhance Hnf6 expression. In vitro, primary hepatocytes derived from KO and WT liver were treated with LPS and hepatocyte apoptosis was assessed with and without GH treatment. RESULTS: In WT mice, GH treatment enhanced Hnf6 expression during BDL, inhibited Caspase -3, -8 and -9 responses and diminished hepatic apoptotic and fibrotic injury. GH-mediated upregulation of Hnf6 expression and parallel suppression of apoptosis and fibrosis in WT BDL liver were abrogated in KO mice. LPS activated apoptosis and suppressed Hnf6 expression in primary hepatocytes. GH/LPS co-treatment enhanced Hnf6 expression with corresponding attenuation of apoptosis in WT-derived hepatocytes, but not in KO hepatocytes. ChiP-on-ChiP and electromobility shift assays of KO and WT liver nuclear extracts identified Ciap1 (or Birc2) as an Hnf6-bound target gene. Ciap1 expression patterns closely follow Hnf6 expression in the liver and in hepatocytes. CONCLUSION: GH broad protective actions on hepatocytes during liver injury are effected through Hnf6, with Hnf6 transcriptional activation of Ciap1 as an underlying molecular mediator.


Asunto(s)
Apoptosis/efectos de los fármacos , Hormona del Crecimiento/farmacología , Factor Nuclear 6 del Hepatocito/metabolismo , Hígado/efectos de los fármacos , Animales , Apoptosis/genética , Western Blotting , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 8/genética , Caspasa 8/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Células Cultivadas , Análisis por Conglomerados , Expresión Génica/efectos de los fármacos , Factor Nuclear 6 del Hepatocito/genética , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Lipopolisacáridos/farmacología , Hígado/citología , Hígado/metabolismo , Ratones Noqueados , Sustancias Protectoras/farmacología , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Neuron ; 92(6): 1252-1265, 2016 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-27939581

RESUMEN

Generic spinal motor neuron identity is established by cooperative binding of programming transcription factors (TFs), Isl1 and Lhx3, to motor-neuron-specific enhancers. How expression of effector genes is maintained following downregulation of programming TFs in maturing neurons remains unknown. High-resolution exonuclease (ChIP-exo) mapping revealed that the majority of enhancers established by programming TFs are rapidly deactivated following Lhx3 downregulation in stem-cell-derived hypaxial motor neurons. Isl1 is released from nascent motor neuron enhancers and recruited to new enhancers bound by clusters of Onecut1 in maturing neurons. Synthetic enhancer reporter assays revealed that Isl1 operates as an integrator factor, translating the density of Lhx3 or Onecut1 binding sites into transient enhancer activity. Importantly, independent Isl1/Lhx3- and Isl1/Onecut1-bound enhancers contribute to sustained expression of motor neuron effector genes, demonstrating that outwardly stable expression of terminal effector genes in postmitotic neurons is controlled by a dynamic relay of stage-specific enhancers.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 6 del Hepatocito/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Neuronas Motoras/metabolismo , Neurogénesis/genética , Factores de Transcripción/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Inmunoprecipitación de Cromatina , Regulación hacia Abajo , Elementos de Facilitación Genéticos , Ratones , Células Madre Embrionarias de Ratones , Proteínas del Tejido Nervioso/metabolismo
19.
J Agric Food Chem ; 64(36): 6826-37, 2016 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-27538679

RESUMEN

Glucose transporters (GLUTs) are required for glucose uptake in malignant cells, and they can be used as molecular targets for cancer therapy. An RT-PCR analysis was performed to investigate the mRNA levels of 14 subtypes of GLUTs in human colorectal cancer (COLO 205 and HT-29) and normal (FHC) cells. RT-PCR (n = 27) was used to assess the differences in paired tissue samples (tumor vs normal) isolated from colorectal cancer patients. GLUT2 was detected in all tested cells. The average GLUT2 mRNA level in 12 of 27 (44.4%) cases was 2.4-fold higher in tumor compared to normal tissues (*, p = 0.027). Higher GLUT2 mRNA expression was preferentially detected in advanced-stage tumors (stage 0 vs 3 = 16.38-fold, 95% CI = 9.22-26.54-fold; *, p = 0.029). The apple polyphenol phloretin (Ph) and siRNA methods were used to inhibit GLUT2 protein expression. Ph (0-100 µM, for 24 h) induced COLO 205 cell growth cycle arrest in a p53-dependent manner, which was confirmed by pretreatment of the cells with a p53-specific dominant negative expression vector. Hepatocyte nuclear factor 6 (HNF6), which was previously reported to be a transcription factor that activates GLUT2 and p53, was also induced by Ph (0-100 µM, for 24 h). The antitumor effect of Ph (25 mg/kg or DMSO twice a week for 6 weeks) was demonstrated in vivo using BALB/c nude mice bearing COLO 205 tumor xenografts. In conclusion, targeting GLUT2 could potentially suppress colorectal tumor cell invasiveness.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Transportador de Glucosa de Tipo 2/metabolismo , Malus/química , Floretina/química , Polifenoles/química , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Anciano , Animales , Antioxidantes/metabolismo , Apoptosis , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Femenino , Glucosa/metabolismo , Factor Nuclear 6 del Hepatocito/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Estadificación de Neoplasias , ARN Mensajero/metabolismo
20.
Genes Dev ; 30(14): 1636-44, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27445394

RESUMEN

Hepatocyte nuclear factor 6 (HNF6) is required for liver development, but its role in adult liver metabolism is not known. Here we show that deletion of HNF6 in livers of adult C57Bl/6 mice leads to hepatic steatosis in mice fed normal laboratory chow. Although HNF6 is known mainly as a transcriptional activator, hepatic loss of HNF6 up-regulated many lipogenic genes bound directly by HNF6. Many of these genes are targets of the circadian nuclear receptor Rev-erbα, and binding of Rev-erbα at these sites was lost when HNF6 was ablated in the liver. While HNF6 and Rev-erbα coordinately regulate hepatic lipid metabolism, each factor also affects additional gene sets independently. These findings highlight a novel mechanism of transcriptional repression by HNF6 and demonstrate how overlapping and distinct mechanisms of transcription factor function contribute to the integrated physiology of the liver.


Asunto(s)
Regulación de la Expresión Génica/genética , Factor Nuclear 6 del Hepatocito/genética , Factor Nuclear 6 del Hepatocito/metabolismo , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Hígado/fisiopatología , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Animales , Hígado Graso/genética , Eliminación de Gen , Técnicas de Inactivación de Genes , Masculino , Ratones , Ratones Endogámicos C57BL , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Unión Proteica/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA