Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Biochemistry (Mosc) ; 89(1): 159-172, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38467552

RESUMEN

N6-methyladenosine (m6A) is one of the most common modifications in both eukaryotic and prokaryotic mRNAs. It has been experimentally confirmed that m6A methylation is involved in the regulation of stability and translation of various mRNAs. Until recently, the majority of m6A-related studies have been focused on the cytoplasmic functions of this modification. Here, we review new data on the role of m6A in several key biological processes taking place in the cell nucleus, such as transcription, chromatin organization, splicing, nuclear-cytoplasmic transport, and R-loop metabolism. Based on analysis of these data, we suggest that m6A methylation of nuclear RNAs is another level of gene expression regulation which, together with DNA methylation and histone modifications, controls chromatin structure and functioning in various biological contexts.


Asunto(s)
Adenosina/análogos & derivados , Metiltransferasas , ARN Nuclear , Metiltransferasas/genética , ARN Nuclear/metabolismo , Metilación , Regulación de la Expresión Génica , ARN Mensajero/metabolismo
2.
Int J Mol Sci ; 25(2)2024 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-38255964

RESUMEN

During gastrulation and neurulation, the chordamesoderm and overlying neuroectoderm of vertebrate embryos converge under the control of a specific genetic programme to the dorsal midline, simultaneously extending along it. However, whether mechanical tensions resulting from these morphogenetic movements play a role in long-range feedback signaling that in turn regulates gene expression in the chordamesoderm and neuroectoderm is unclear. In the present work, by using a model of artificially stretched explants of Xenopus midgastrula embryos and full-transcriptome sequencing, we identified genes with altered expression in response to external mechanical stretching. Importantly, mechanically activated genes appeared to be expressed during normal development in the trunk, i.e., in the stretched region only. By contrast, genes inhibited by mechanical stretching were normally expressed in the anterior neuroectoderm, where mechanical stress is low. These results indicate that mechanical tensions may play the role of a long-range signaling factor that regulates patterning of the embryo, serving as a link coupling morphogenesis and cell differentiation.


Asunto(s)
4-Butirolactona , Animales , Estrés Mecánico , Xenopus laevis/genética , Expresión Génica
3.
Int J Mol Sci ; 22(14)2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-34299205

RESUMEN

Gain and loss of DNA methylation in cells is a dynamic process that tends to achieve an equilibrium. Many factors are involved in maintaining the balance between DNA methylation and demethylation. Previously, it was shown that methyl-DNA protein Kaiso may attract NCoR, SMRT repressive complexes affecting histone modifications. On the other hand, the deficiency of Kaiso resulted in reduced methylation of ICR in H19/Igf2 locus and Oct4 promoter in mouse embryonic fibroblasts. However, nothing is known about how Kaiso influences DNA methylation at the genome level. Here we show that deficiency of Kaiso led to whole-genome hypermethylation, using Kaiso deficient human renal cancer cell line obtained via CRISPR/CAS9 genome editing. However, Kaiso serves to protect genic regions, enhancers, and regions with a low level of histone modifications from demethylation. We detected hypomethylation of binding sites for Oct4 and Nanog in Kaiso deficient cells. Kaiso immunoprecipitated with de novo DNA methyltransferases DNMT3a/3b, but not with maintenance methyltransferase DNMT1. Thus, Kaiso may attract methyltransferases to surrounding regions and modulate genome methylation in renal cancer cells apart from being methyl DNA binding protein.


Asunto(s)
Metilación de ADN , Impresión Genómica , Factor II del Crecimiento Similar a la Insulina/metabolismo , Región de Control de Posición , ARN Largo no Codificante/genética , Factores de Transcripción/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Edición Génica , Células HEK293 , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Regiones Promotoras Genéticas , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Proteínas de Motivos Tripartitos/genética , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , ADN Metiltransferasa 3B
4.
Histochem Cell Biol ; 143(1): 29-43, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25182933

RESUMEN

The Kaiso protein was originally described as a BTB/POZ zinc-finger transcription factor and a p120-catenin-binding partner. It is a DNA methylation-dependent transcriptional repressor, but its biological role in mice is still unknown. Here, we characterized a Kaiso-specific antibody by examining Kaiso protein distribution by immunofluorescence microscopy in the following tissues and cell types of adult mice: skin, small intestine, mammary glands, urinary bladder, and others. This study is the first to demonstrate that Kaiso is expressed in most of the examined tissues. Kaiso was localized to the nucleus in almost all tissues. However, it was primarily cytoplasmic in photoreceptor cells in the eye (rods and cones). Furthermore, Kaiso is expressed in a specific subset of male germ cells that are characterized by partly positive PLZF and Bmi-1 staining. In this study, we present the first confirmation of the reliability of expression data using Kaiso knockout mice.


Asunto(s)
Factores de Transcripción/análisis , Animales , Ojo/química , Humanos , Intestino Delgado/química , Masculino , Glándulas Mamarias Humanas/química , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Especificidad de Órganos , Piel/química , Testículo/química , Factores de Transcripción/deficiencia , Vejiga Urinaria/química
5.
Cell Rep ; 33(7): 108396, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33207197

RESUMEN

Zyxin is a cytoskeletal LIM-domain protein that regulates actin cytoskeleton assembly and gene expression. In the present work, we find that zyxin downregulation in Xenopus laevis embryos reduces the expression of numerous genes that regulate cell differentiation, but it enhances that of several genes responsible for embryonic stem cell status, specifically klf4, pou5f3.1, pou5f3.2, pou5f3.3, and vent2.1/2. For pou5f3 family genes (mammalian POU5F1/OCT4 homologs), we show that this effect is the result of mRNA stabilization due to complex formation with the Y-box protein Ybx1. When bound to Ybx1, zyxin interferes with the formation of these complexes, thereby stimulating pou5f3 mRNA degradation. In addition, in zebrafish embryos and human HEK293 cells, zyxin downregulation increases mRNA levels of the pluripotency genes KLF4, NANOG, and POU5F1/OCT4. Our findings indicate that zyxin may play a role as a switch among morphogenetic cell movement, differentiation, and embryonic stem cell status.


Asunto(s)
Células Madre Embrionarias/metabolismo , Zixina/metabolismo , Zixina/fisiología , Animales , Tipificación del Cuerpo/genética , Diferenciación Celular/genética , Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Células Madre Embrionarias/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Células HEK293 , Humanos , Factor 4 Similar a Kruppel , Morfogénesis , Placa Neural/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Xenopus laevis/metabolismo , Pez Cebra/metabolismo
6.
Sci Rep ; 8(1): 10667, 2018 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-30006568

RESUMEN

VHL inactivation is a key oncogenic event for renal carcinomas. In normoxia, VHL suppresses HIF1a-mediated transcriptional response, which is characteristic to hypoxia. It has previously been shown that hypoxic conditions inhibit TET-dependent hydroxymethylation of cytosines and cause DNA hypermethylation at gene promoters. In this work, we performed VHL inactivation by CRISPR/Cas9 and studied its effects on gene expression and DNA methylation. We showed that even without hypoxia, VHL inactivation leads to hypermethylation of the genome. Hypermethylated cytosines were evenly distributed throughout the genome with a slight preference for AP-1 (JUN and FOS) binding sites. Hypermethylated cytosines tended to be enriched within the binding sites of transcription factors that showed increased gene expression after VHL inactivation. We also observed promoter hypermethylation associated with decreased gene expression for several regulators of transcription and DNA methylation including SALL3.


Asunto(s)
Carcinoma de Células Renales/genética , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Neoplasias Renales/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Sistemas CRISPR-Cas/genética , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Citosina/metabolismo , Silenciador del Gen , Genoma Humano/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Renales/patología , Regiones Promotoras Genéticas/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Hipoxia Tumoral , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
7.
Cell Death Differ ; 25(11): 1938-1951, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29472715

RESUMEN

Kaiso is a member of the BTB/POZ zinc finger family, which is involved in cancer progression, cell cycle control, apoptosis, and WNT signaling. Depending on promoter context, it may function as either a transcriptional repressor or activator. Previous studies found that Kaiso might be SUMOylated due to heat shock, but the biological significance of Kaiso SUMOylation is unclear. Here, we find that K42 is the only amino acid within Kaiso that is modified with SUMO. Kaiso is monoSUMOylated at lysine 42 in cell lines of kidney origin under normal physiological conditions. SUMOylated Kaiso can activate transcription from exogenous methylated promoters, wherein the deSUMOylated form of the protein kept the ability to be a repressor. Rapid Kaiso deSUMOylation occurs in response to hyperosmotic stress and is reversible upon return to an isotonic environment. DeSUMOylation occurs within minutes in HEK293 cells treated with 100 mM NaCl and relaxes in 3 h even in a salt-containing medium. Genomic editing of Kaiso by conversion of K42 into R42 (K42R) in HEK293 cells that resulted in fully deSUMOylated endogenous protein led to misregulation of genes associated with ion transport, blood pressure, and the immune response. TRIM25 was significantly repressed in two K42R HEK293 clones. By a series of rescue experiments with K42R and KO HEK293 cells, we show that TRIM25 is a direct transcriptional target for Kaiso. In the absence of Kaiso, the level of TRIM25 is insensitive to hyperosmotic stress. Extending our observations to animal models, we show that in response to a high salt diet, Kaiso knockout mice are characterized by significantly higher blood pressure increases when compared to wild-type animals. Thus, we propose a novel biological role for Kaiso in the regulation of homeostasis.


Asunto(s)
Presión Osmótica , Factores de Transcripción/metabolismo , Animales , Apoptosis/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Sistemas CRISPR-Cas/genética , Línea Celular , Células HEK293 , Humanos , Hipertensión/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Cloruro de Sodio/farmacología , Sumoilación/efectos de los fármacos , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Transcripción Genética , Proteínas de Motivos Tripartitos/genética , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
8.
F1000Res ; 4: 1518, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26925226

RESUMEN

Human cancer cells are subjected to hypoxic conditions in many tumours. Hypoxia causes alterations in the glycolytic pathway activation through stabilization of hypoxia-inducible factor 1. Currently, two approaches are commonly used to model hypoxia: an alternative to generating low-oxygen conditions in an incubator, cells can be treated with CoCl 2. We performed RNA-seq experiments to study transcriptomes of human Caki-1 cells under real hypoxia and after CoCl 2 treatment. Despite causing transcriptional changes of a much higher order of magnitude for the genes in the hypoxia regulation pathway, CoCl 2 treatment fails to induce alterations in the glycolysis / gluconeogenesis pathway. Moreover, CoCl 2 caused aberrant activation of other oxidoreductases in glycine, serine and threonine metabolism pathways.

9.
PLoS One ; 9(10): e109914, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25333971

RESUMEN

In this study, the optimized method for designing IgG-binding magnetosomes based on integration of IgG-binding fusion proteins into magnetosome membrane in vitro is presented. Fusion proteins Mbb and Mistbb consisting of magnetosome membrane protein MamC and membrane associating protein Mistic from Bacillus subtilis as anchors and BB-domains of Staphylococcus aureus protein A as IgG-binding region were used. With Response Surface Methodology (RSM) the highest level of proteins integration into magnetosome membrane was achieved under the following parameters: pH 8.78, without adding NaCl and 55 s of vortexing for Mbb; pH 9.48, 323 mM NaCl and 55 s of vortexing for Mistbb. Modified magnetosomes with Mbb and Mistbb displayed on their surface demonstrated comparable levels of IgG-binding activity, suggesting that both proteins could be efficiently used as anchor molecules. We also demonstrated that such modified magnetosomes are stable in PBS buffer during at least two weeks. IgG-binding magnetosomes obtained by this approach could serve as a multifunctional platform for displaying various types of antibodies.


Asunto(s)
Inmunoglobulina G/metabolismo , Nanopartículas de Magnetita/química , Magnetosomas/química , Proteínas Bacterianas/metabolismo , Proteínas de la Membrana/metabolismo , Unión Proteica , Proteína Estafilocócica A/metabolismo , Staphylococcus aureus/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA