Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nanoscale ; 15(34): 14238-14248, 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37599610

RESUMEN

The efficient functionalization of graphene with sulfonic groups using a sustainable approach facilitates the interaction of biomolecules with its surface. The inclusion of these graphene sheets inside a photopolymerized acrylamide-based hydrogel provides a 3D scaffold with viscoelastic behaviour closer to that found in natural tissues. Cell-culture experiments and differentiation assays with SH-SY5Y cells showed that these hybrid hydrogels are non-cytotoxic, thus making them potentially useful as scaffold materials mimicking the extracellular environment.


Asunto(s)
Grafito , Neuroblastoma , Humanos , Matriz Extracelular , Acrilamida , Hidrogeles
2.
Nat Commun ; 14(1): 3761, 2023 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-37353485

RESUMEN

Pancreatic acinar cells rely on PTF1 and other transcription factors to deploy their transcriptional program. We identify NFIC as a NR5A2 interactor and regulator of acinar differentiation. NFIC binding sites are enriched in NR5A2 ChIP-Sequencing peaks. Nfic knockout mice have a smaller, histologically normal, pancreas with reduced acinar gene expression. NFIC binds and regulates the promoters of acinar genes and those involved in RNA/protein metabolism, and Nfic knockout pancreata show defective ribosomal RNA maturation. NFIC dampens the endoplasmic reticulum stress program through binding to gene promoters and is required for resolution of Tunicamycin-mediated stress. NFIC is down-regulated during caerulein pancreatitis and is required for recovery after damage. Normal human pancreata with low levels of NFIC transcripts display reduced expression of genes down-regulated in Nfic knockout mice. NFIC expression is down-regulated in mouse and human pancreatic ductal adenocarcinoma. Consistently, Nfic knockout mice develop a higher number of mutant Kras-driven pre-neoplastic lesions.


Asunto(s)
Carcinoma Ductal Pancreático , Factores de Transcripción NFI , Neoplasias Pancreáticas , Ribosomas , Animales , Humanos , Ratones , Células Acinares/metabolismo , Carcinoma Ductal Pancreático/patología , Ratones Noqueados , Factores de Transcripción NFI/metabolismo , Páncreas/metabolismo , Neoplasias Pancreáticas/patología
3.
Dev Neurobiol ; 82(6): 495-504, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35796156

RESUMEN

A striking feature of the nervous system pertains to the appearance of different neural cell subtypes at different axial levels. Studies in the Drosophila central nervous system reveal that one mechanism underlying such segmental differences pertains to the segment-specific removal of cells by programmed cell death (PCD). One group of genes involved in segment-specific PCD is the Hox homeotic genes. However, while segment-specific PCD is highly precise, Hox gene expression is evident in gradients, raising the issue of how the Hox gene function is precisely gated to trigger PCD in specific segments at the outer limits of Hox expression. The Drosophila Va neurons are initially generated in all nerve cord segments but removed by PCD in posterior segments. Va PCD is triggered by the posteriorly expressed Hox gene Abdominal-B (Abd-B). However, Va PCD is highly reproducible despite exceedingly weak Abd-B expression in the anterior frontiers of its expression. Here, we found that the transcriptional cofactor Dachshund supports Abd-B-mediated PCD in its anterior domain. In vivo bimolecular fluorescence complementation analysis lends support to the idea that the Dachshund/Abd-B interplay may involve physical interactions. These findings provide an example of how combinatorial codes of transcription factors ensure precision in Hox-mediated PCD in specific segments at the outer limits of Hox expression.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Apoptosis , Sistema Nervioso Central/metabolismo , Perros , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
EMBO J ; 39(9): e102808, 2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32154941

RESUMEN

Defects in transcriptional regulators of pancreatic exocrine differentiation have been implicated in pancreatic tumorigenesis, but the molecular mechanisms are poorly understood. The locus encoding the transcription factor HNF1A harbors susceptibility variants for pancreatic ductal adenocarcinoma (PDAC), while KDM6A, encoding Lysine-specific demethylase 6A, carries somatic mutations in PDAC. Here, we show that pancreas-specific Hnf1a null mutant transcriptomes phenocopy those of Kdm6a mutations, and both defects synergize with KrasG12D to cause PDAC with sarcomatoid features. We combine genetic, epigenomic, and biochemical studies to show that HNF1A recruits KDM6A to genomic binding sites in pancreatic acinar cells. This remodels the acinar enhancer landscape, activates differentiated acinar cell programs, and indirectly suppresses oncogenic and epithelial-mesenchymal transition genes. We also identify a subset of non-classical PDAC samples that exhibit the HNF1A/KDM6A-deficient molecular phenotype. These findings provide direct genetic evidence that HNF1A deficiency promotes PDAC. They also connect the tumor-suppressive role of KDM6A deficiency with a cell-specific molecular mechanism that underlies PDAC subtype definition.


Asunto(s)
Células Acinares/metabolismo , Carcinoma Ductal Pancreático/genética , Factor Nuclear 1-alfa del Hepatocito/genética , Histona Demetilasas/genética , Neoplasias Pancreáticas/genética , Animales , Carcinoma Ductal Pancreático/metabolismo , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Histona Demetilasas/metabolismo , Humanos , Ratones , Mutación , Especificidad de Órganos , Páncreas/metabolismo , Neoplasias Pancreáticas/metabolismo
5.
Nature ; 554(7693): 533-537, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29443959

RESUMEN

Chronic inflammation increases the risk of developing one of several types of cancer. Inflammatory responses are currently thought to be controlled by mechanisms that rely on transcriptional networks that are distinct from those involved in cell differentiation. The orphan nuclear receptor NR5A2 participates in a wide variety of processes, including cholesterol and glucose metabolism in the liver, resolution of endoplasmic reticulum stress, intestinal glucocorticoid production, pancreatic development and acinar differentiation. In genome-wide association studies, single nucleotide polymorphisms in the vicinity of NR5A2 have previously been associated with the risk of pancreatic adenocarcinoma. In mice, Nr5a2 heterozygosity sensitizes the pancreas to damage, impairs regeneration and cooperates with mutant Kras in tumour progression. Here, using a global transcriptomic analysis, we describe an epithelial-cell-autonomous basal pre-inflammatory state in the pancreas of Nr5a2+/- mice that is reminiscent of the early stages of pancreatitis-induced inflammation and is conserved in histologically normal human pancreases with reduced expression of NR5A2 mRNA. In Nr5a2+/-mice, NR5A2 undergoes a marked transcriptional switch, relocating from differentiation-specific to inflammatory genes and thereby promoting gene transcription that is dependent on the AP-1 transcription factor. Pancreatic deletion of Jun rescues the pre-inflammatory phenotype, as well as binding of NR5A2 to inflammatory gene promoters and the defective regenerative response to damage. These findings support the notion that, in the pancreas, the transcriptional networks involved in differentiation-specific functions also suppress inflammatory programmes. Under conditions of genetic or environmental constraint, these networks can be subverted to foster inflammation.


Asunto(s)
Diferenciación Celular/genética , Regulación de la Expresión Génica , Inflamación/genética , Páncreas/metabolismo , Páncreas/patología , Receptores Citoplasmáticos y Nucleares/metabolismo , Transcriptoma , Células Acinares/metabolismo , Células Acinares/patología , Animales , Cromatina/genética , Cromatina/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Redes Reguladoras de Genes/genética , Genes jun/genética , Heterocigoto , Humanos , Ratones , Especificidad de Órganos/genética , Pancreatitis/genética , Regiones Promotoras Genéticas/genética , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores Citoplasmáticos y Nucleares/genética , Factor de Transcripción AP-1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...