Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
BMC Genomics ; 24(1): 252, 2023 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-37165305

RESUMO

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.


Assuntos
RNA Longo não Codificante , Animais , Camundongos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Retina/metabolismo , Perfilação da Expressão Gênica , Fator 6 Nuclear de Hepatócito/genética , Fator 6 Nuclear de Hepatócito/metabolismo
3.
Commun Biol ; 4(1): 1298, 2021 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-34789845

RESUMO

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.


Assuntos
Fator 6 Nuclear de Hepatócito/genética , Pâncreas/fisiologia , Diferenciação Celular , Linhagem Celular , Regulação da Expressão Gênica no Desenvolvimento , Fator 6 Nuclear de Hepatócito/metabolismo , Células-Tronco Embrionárias Humanas , Humanos , Transcrição Gênica
4.
Nat Med ; 27(11): 1928-1940, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34663987

RESUMO

Genes involved in distinct diabetes types suggest shared disease mechanisms. Here we show that One Cut Homeobox 1 (ONECUT1) mutations cause monogenic recessive syndromic diabetes in two unrelated patients, characterized by intrauterine growth retardation, pancreas hypoplasia and gallbladder agenesis/hypoplasia, and early-onset diabetes in heterozygous relatives. Heterozygous carriers of rare coding variants of ONECUT1 define a distinctive subgroup of diabetic patients with early-onset, nonautoimmune diabetes, who respond well to diabetes treatment. In addition, common regulatory ONECUT1 variants are associated with multifactorial type 2 diabetes. Directed differentiation of human pluripotent stem cells revealed that loss of ONECUT1 impairs pancreatic progenitor formation and a subsequent endocrine program. Loss of ONECUT1 altered transcription factor binding and enhancer activity and NKX2.2/NKX6.1 expression in pancreatic progenitor cells. Collectively, we demonstrate that ONECUT1 controls a transcriptional and epigenetic machinery regulating endocrine development, involved in a spectrum of diabetes, encompassing monogenic (recessive and dominant) as well as multifactorial inheritance. Our findings highlight the broad contribution of ONECUT1 in diabetes pathogenesis, marking an important step toward precision diabetes medicine.


Assuntos
Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Fator 6 Nuclear de Hepatócito/genética , Pâncreas/embriologia , Diferenciação Celular/genética , Anormalidades Congênitas/genética , Retardo do Crescimento Fetal/genética , Vesícula Biliar/anormalidades , Proteína Homeobox Nkx-2.2/biossíntese , Proteínas de Homeodomínio/biossíntese , Humanos , Lactente , Recém-Nascido , Masculino , Herança Multifatorial/genética , Organogênese/genética , Pâncreas/anormalidades , Pancreatopatias/congênito , Pancreatopatias/genética , Células-Tronco Pluripotentes/citologia , Transcrição Gênica/genética
5.
Biomolecules ; 11(8)2021 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-34439790

RESUMO

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.


Assuntos
Linhagem da Célula/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Epigênese Genética , Expressão Gênica , Células Secretoras de Insulina/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Loci Gênicos , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo , Fator 6 Nuclear de Hepatócito/genética , Fator 6 Nuclear de Hepatócito/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Células Secretoras de Insulina/citologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Proteína Homeobox Nanog/genética , Proteína Homeobox Nanog/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Pâncreas/citologia , Pâncreas/metabolismo , Cultura Primária de Células , Transativadores/genética , Transativadores/metabolismo
6.
Development ; 148(9)2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33929509

RESUMO

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.


Assuntos
Dissecação , Fator 6 Nuclear de Hepatócito/metabolismo , Fatores de Transcrição Otx/metabolismo , Retina/crescimento & desenvolvimento , Células Fotorreceptoras Retinianas Cones/metabolismo , Fatores de Transcrição/metabolismo , Animais , Sítios de Ligação , Galinhas , Cromatina , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Fator 6 Nuclear de Hepatócito/genética , Fatores de Transcrição Otx/genética , Retina/metabolismo , Células-Tronco
7.
Bull Exp Biol Med ; 170(1): 40-45, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33222081

RESUMO

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.


Assuntos
Fator 6 Nuclear de Hepatócito/genética , Hepatócitos/metabolismo , Células de Kupffer/metabolismo , Regeneração Hepática/genética , Fígado/metabolismo , Animais , Ductos Biliares/metabolismo , Ductos Biliares/cirurgia , Proliferação de Células , Feminino , Regulação da Expressão Gênica , Hepatectomia/métodos , Fator 6 Nuclear de Hepatócito/metabolismo , Hepatócitos/citologia , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Células de Kupffer/citologia , Fígado/cirurgia , Masculino , Ratos , Ratos Sprague-Dawley
8.
Nat Commun ; 11(1): 5292, 2020 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-33087715

RESUMO

Recent advances have enabled the direct induction of human tissue-specific stem and progenitor cells from differentiated somatic cells. However, it is not known whether human hepatic progenitor cells (hHepPCs) can be generated from other cell types by direct lineage reprogramming with defined transcription factors. Here, we show that a set of three transcription factors, FOXA3, HNF1A, and HNF6, can induce human umbilical vein endothelial cells to directly acquire the properties of hHepPCs. These induced hHepPCs (hiHepPCs) propagate in long-term monolayer culture and differentiate into functional hepatocytes and cholangiocytes by forming cell aggregates and cystic epithelial spheroids, respectively, under three-dimensional culture conditions. After transplantation, hiHepPC-derived hepatocytes and cholangiocytes reconstitute damaged liver tissues and support hepatic function. The defined transcription factors also induce hiHepPCs from endothelial cells circulating in adult human peripheral blood. These expandable and bipotential hiHepPCs may be useful in the study and treatment of human liver diseases.


Assuntos
Técnicas de Reprogramação Celular/métodos , Células Endoteliais/citologia , Hepatócitos/citologia , Células-Tronco/citologia , Animais , Ductos Biliares/citologia , Ductos Biliares/fisiologia , Agregação Celular , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Reprogramação Celular/genética , Reprogramação Celular/fisiologia , Células Endoteliais/fisiologia , Feminino , Fator 1-alfa Nuclear de Hepatócito/genética , Fator 1-alfa Nuclear de Hepatócito/fisiologia , Fator 3-gama Nuclear de Hepatócito/genética , Fator 3-gama Nuclear de Hepatócito/fisiologia , Fator 6 Nuclear de Hepatócito/genética , Fator 6 Nuclear de Hepatócito/fisiologia , Hepatócitos/fisiologia , Hepatócitos/transplante , Xenoenxertos , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Esferoides Celulares/citologia , Esferoides Celulares/fisiologia , Células-Tronco/fisiologia
9.
PLoS One ; 15(8): e0237403, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32790713

RESUMO

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.


Assuntos
Fator 6 Nuclear de Hepatócito/genética , Proteínas de Homeodomínio/genética , Retina/fisiologia , Fatores de Transcrição/genética , Alelos , Células Amácrinas/metabolismo , Células Amácrinas/patologia , Animais , Células Ependimogliais/metabolismo , Células Ependimogliais/patologia , Olho/crescimento & desenvolvimento , Olho/patologia , Loci Gênicos , Genótipo , Fator 6 Nuclear de Hepatócito/metabolismo , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Transgênicos , Retina/citologia , Retina/patologia , Células Bipolares da Retina/metabolismo , Células Bipolares da Retina/patologia , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Fotorreceptoras Retinianas Cones/patologia , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/metabolismo , Fatores de Transcrição/metabolismo
10.
J Mol Endocrinol ; 64(3): 165-179, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31990658

RESUMO

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.


Assuntos
Álcool Desidrogenase/genética , Hidrocarboneto de Aril Hidroxilases/genética , Família 2 do Citocromo P450/genética , Família 7 do Citocromo P450/genética , Hormônio do Crescimento/farmacologia , Fator 6 Nuclear de Hepatócito/genética , Receptores da Prolactina/genética , Esteroide Hidroxilases/genética , Álcool Desidrogenase/metabolismo , Animais , Hidrocarboneto de Aril Hidroxilases/metabolismo , Família 2 do Citocromo P450/metabolismo , Família 7 do Citocromo P450/metabolismo , Epigênese Genética/efeitos dos fármacos , Epigênese Genética/fisiologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônio do Crescimento/metabolismo , Fator 6 Nuclear de Hepatócito/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas/genética , Receptores da Prolactina/metabolismo , Caracteres Sexuais , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Esteroide Hidroxilases/metabolismo
11.
Food Chem Toxicol ; 136: 111070, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31870920

RESUMO

While liver injury is commonly associated with excessive alcohol consumption, how liver injury affects alcohol metabolism and drinking preference remains unclear. To answer these questions, we measured the expression and activity of alcohol dehydrogenase 1 (ADH1) and acetaldehyde dehydrogenase 2 (ALDH2) enzymes, ethanol and acetaldehyde levels in vivo, and binge-like and preferential drinking behaviors with drinking in the dark and two-bottle choice in animal models with liver injury. Acute and chronic carbon tetrachloride (CCl4), and acute LPS-induced liver injury repressed hepatic ALDH2 activity and expression and consequently, blood and liver acetaldehyde concentrations were increased in these models. In addition, chronic CCl4 and acute LPS treatment inhibited hepatic ADH1 expression and activity, leading to increases in blood and liver ethanol concentrations. Consistent with the increase in acetaldehyde levels, alcohol drinking behaviors were reduced in mice with acute or chronic liver injury. Furthermore, oxidative stress induced by hydrogen peroxide attenuated ADH1 and ALDH2 activity post-transcriptionally, while proinflammatory cytokines led to transcriptional repression of ADH1 and ALDH2 in cultured hepatocytes, which correlated with the repression of transcription factor HNF4α. Collectively, our data suggest that alcohol metabolism is suppressed by inflammation and oxidative stress, which is correlated with decreased drinking behavior.


Assuntos
Consumo de Bebidas Alcoólicas/efeitos adversos , Etanol/efeitos adversos , Hepatopatias/imunologia , Fígado/lesões , Aldeído Desidrogenase/genética , Aldeído Desidrogenase/imunologia , Aldeído-Desidrogenase Mitocondrial/genética , Aldeído-Desidrogenase Mitocondrial/imunologia , Animais , Etanol/metabolismo , Fator 6 Nuclear de Hepatócito/genética , Fator 6 Nuclear de Hepatócito/imunologia , Humanos , Fígado/imunologia , Hepatopatias/etiologia , Hepatopatias/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL
12.
Nucleic Acids Res ; 47(11): 5587-5602, 2019 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-31049588

RESUMO

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.


Assuntos
Reprogramação Celular , Cromatina/genética , Regulação da Expressão Gênica , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios/metabolismo , Fatores de Transcrição Onecut/genética , Diferenciação Celular , Linhagem Celular , Cromatina/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Ontologia Genética , Fator 6 Nuclear de Hepatócito/genética , Fator 6 Nuclear de Hepatócito/metabolismo , Proteínas de Homeodomínio , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Neurônios/citologia , Fatores de Transcrição Onecut/metabolismo , Fatores de Transcrição
13.
J Cell Physiol ; 234(4): 3675-3684, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30256389

RESUMO

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.


Assuntos
Movimento Celular , Proliferação de Células , Neoplasias Colorretais/metabolismo , Fator 6 Nuclear de Hepatócito/metabolismo , Neoplasias Hepáticas/metabolismo , Animais , Anoikis , Adesão Celular , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica , Fator 6 Nuclear de Hepatócito/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/secundário , Camundongos Endogâmicos BALB C , Camundongos Nus , Transdução de Sinais , Carga Tumoral , Regulação para Cima
14.
Neural Dev ; 13(1): 26, 2018 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-30466480

RESUMO

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.


Assuntos
Fator 6 Nuclear de Hepatócito/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Animais , Galinhas , Citometria de Fluxo , Genes erbA , Fator 6 Nuclear de Hepatócito/genética , Camundongos , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/metabolismo , Retina/metabolismo , Rodopsina/genética , Rodopsina/metabolismo
15.
Am J Physiol Endocrinol Metab ; 314(4): E308-E321, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29351489

RESUMO

The transcription factors pancreatic and duodenal homeobox 1 (Pdx1) and onecut1 (Oc1) are coexpressed in multipotent pancreatic progenitors (MPCs), but their expression patterns diverge in hormone-expressing cells, with Oc1 expression being extinguished in the endocrine lineage and Pdx1 being maintained at high levels in ß-cells. We previously demonstrated that cooperative function of these two factors in MPCs is necessary for proper specification and differentiation of pancreatic endocrine cells. In those studies, we observed a persistent decrease in expression of the ß-cell maturity factor MafA. We therefore hypothesized that Pdx1 and Oc1 cooperativity in MPCs impacts postnatal ß-cell maturation and function. Here our model of Pdx1-Oc1 double heterozygosity was used to investigate the impact of haploinsufficiency for both of these factors on postnatal ß-cell maturation, function, and adaptability. Examining mice at postnatal day (P) 14, we observed alterations in pancreatic insulin content in both Pdx1 heterozygotes and double heterozygotes. Gene expression analysis at this age revealed significantly decreased expression of many genes important for glucose-stimulated insulin secretion (e.g., Glut2, Pcsk1/2, Abcc8) exclusively in double heterozygotes. Analysis of P14 islets revealed an increase in the number of mixed islets in double heterozygotes. We predicted that double-heterozygous ß-cells would have an impaired ability to respond to stress. Indeed, we observed that ß-cell proliferation fails to increase in double heterozygotes in response to either high-fat diet or placental lactogen. We thus report here the importance of cooperation between regulatory factors early in development for postnatal islet maturation and adaptability.


Assuntos
Fator 6 Nuclear de Hepatócito/fisiologia , Proteínas de Homeodomínio/fisiologia , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/crescimento & desenvolvimento , Células-Tronco Multipotentes/metabolismo , Transativadores/fisiologia , Adaptação Fisiológica/efeitos dos fármacos , Adaptação Fisiológica/genética , Animais , Animais Recém-Nascidos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Dieta Hiperlipídica , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Glucose/farmacologia , Fator 6 Nuclear de Hepatócito/genética , Proteínas de Homeodomínio/genética , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Células-Tronco Multipotentes/efeitos dos fármacos , Células-Tronco Multipotentes/fisiologia , Organogênese/efeitos dos fármacos , Organogênese/genética , Transativadores/genética
16.
Cell Rep ; 20(7): 1533-1542, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28813666

RESUMO

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.


Assuntos
Córtex Cerebral/metabolismo , MicroRNAs/genética , Neovascularização Fisiológica/genética , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Sequência de Bases , Sítios de Ligação , Diferenciação Celular , Proliferação de Células , Córtex Cerebral/crescimento & desenvolvimento , Embrião não Mamífero , Feto , Regulação da Expressão Gênica no Desenvolvimento , Fator 6 Nuclear de Hepatócito/genética , Fator 6 Nuclear de Hepatócito/metabolismo , Humanos , MicroRNAs/metabolismo , Morfogênese/genética , Células-Tronco Neurais/citologia , Neurônios/metabolismo , Neurônios/patologia , Receptores Nucleares Órfãos , Cultura Primária de Células , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Retina/crescimento & desenvolvimento , Retina/metabolismo , Transdução de Sinais , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra
17.
Sci Rep ; 7: 44686, 2017 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-28304379

RESUMO

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.


Assuntos
Membrana Corioalantoide/metabolismo , Avaliação Pré-Clínica de Medicamentos , Medições Luminescentes/métodos , Modelos Biológicos , Neoplasias Pancreáticas/patologia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/patologia , Animais , Carcinogênese/patologia , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Galinhas , Crizotinibe/farmacologia , Crizotinibe/uso terapêutico , Análise Mutacional de DNA , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Fator 6 Nuclear de Hepatócito/genética , Fator 6 Nuclear de Hepatócito/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Análise de Sequência de DNA , Células Tumorais Cultivadas , Gencitabina , Neoplasias Pancreáticas
18.
PLoS One ; 11(12): e0167085, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27936029

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Hormônio do Crescimento/farmacologia , Fator 6 Nuclear de Hepatócito/metabolismo , Fígado/efeitos dos fármacos , Animais , Apoptose/genética , Western Blotting , Caspase 3/genética , Caspase 3/metabolismo , Caspase 8/genética , Caspase 8/metabolismo , Caspase 9/genética , Caspase 9/metabolismo , Células Cultivadas , Análise por Conglomerados , Expressão Gênica/efeitos dos fármacos , Fator 6 Nuclear de Hepatócito/genética , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Proteínas Inibidoras de Apoptose/genética , Proteínas Inibidoras de Apoptose/metabolismo , Lipopolissacarídeos/farmacologia , Fígado/citologia , Fígado/metabolismo , Camundongos Knockout , Substâncias Protetoras/farmacologia , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Genes Dev ; 30(14): 1636-44, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27445394

RESUMO

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.


Assuntos
Regulação da Expressão Gênica/genética , Fator 6 Nuclear de Hepatócito/genética , Fator 6 Nuclear de Hepatócito/metabolismo , Metabolismo dos Lipídeos/genética , Fígado/metabolismo , Fígado/fisiopatologia , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Animais , Fígado Gorduroso/genética , Deleção de Genes , Técnicas de Inativação de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/genética , Ligação Proteica/genética
20.
Cell Cycle ; 15(12): 1602-10, 2016 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-27259983

RESUMO

Autophagy plays important roles in self-renewal and differentiation of stem cells. Hepatic progenitor cells (HPCs) are thought to have the ability of self-renewal as well as possess a bipotential capacity, which allows them to differentiate into both hepatocytes and bile ductular cells. However, how autophagy contributes to self-renewal and differentiation of hepatic progenitor cells is not well understood. In this study, we use a well-established rat hepatic progenitor cell lines called WB-F344, which is treated with 3.75 mM sodium butyrate (SB) to promote the differentiation of WB-F344 along the biliary phenotype. We found that autophagy was decreased in the early stage of biliary differentiation, and maintained a low level at the late stage. Activation of autophagy by rapamycin or starvation suppressed the biliary differentiation of WB-F344. Further study reported that autophagy inhibited Notch1 signaling pathway, which contributed to biliary differentiation and morphogenesis. In conclusions, autophagy regulates biliary differentiation of hepatic progenitor cells through Notch1 signaling pathway.


Assuntos
Ácido Butírico/farmacologia , Células Epiteliais/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Receptor Notch1/genética , Sirolimo/farmacologia , Células-Tronco/efeitos dos fármacos , Animais , Autofagia/efeitos dos fármacos , Autofagia/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Ductos Biliares/citologia , Ductos Biliares/efeitos dos fármacos , Ductos Biliares/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Fator 1-beta Nuclear de Hepatócito/genética , Fator 1-beta Nuclear de Hepatócito/metabolismo , Fator 6 Nuclear de Hepatócito/genética , Fator 6 Nuclear de Hepatócito/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Queratina-19/genética , Queratina-19/metabolismo , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Ratos , Ratos Endogâmicos F344 , Receptor Notch1/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição HES-1/genética , Fatores de Transcrição HES-1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...