Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Front Neuroendocrinol ; 71: 101102, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37689249

RESUMEN

The brain synthesizes a variety of neurosteroids, including neuroestradiol. Inhibition of neuroestradiol synthesis results in alterations in basic neurodevelopmental processes, such as neurogenesis, neuroblast migration, neuritogenesis and synaptogenesis. Although the neurodevelopmental actions of neuroestradiol are exerted in both sexes, some of them are sex-specific, such as the well characterized effects of neuroestradiol derived from the metabolism of testicular testosterone during critical periods of male brain development. In addition, recent findings have shown sex-specific actions of neuroestradiol on neuroblast migration, neuritic growth and synaptogenesis in females. Among other factors, the epigenetic regulation exerted by X linked genes, such as Kdm6a/Utx, may determine sex-specific actions of neuroestradiol in the female brain. This review evidences the impact of neuroestradiol on brain formation in both sexes and highlights the interaction of neural steriodogenesis, hormones and sex chromosomes in sex-specific brain development.


Asunto(s)
Epigénesis Genética , Neuroesteroides , Femenino , Masculino , Humanos , Neuronas/metabolismo , Neuroesteroides/metabolismo , Testosterona/metabolismo
2.
Front Cell Dev Biol ; 10: 937875, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36268511

RESUMEN

Kdm6a is an X-chromosome-linked H3K27me2/3 demethylase that promotes chromatin accessibility and gene transcription and is critical for tissue/cell-specific differentiation. Previous results showed higher Kdm6a levels in XX than in XY hypothalamic neurons and a female-specific requirement for Kdm6a in mediating increased axogenesis before brain masculinization. Here, we explored the sex-specific role of Kdm6a in the specification of neuronal subtypes in the developing hypothalamus. Hypothalamic neuronal cultures were established from sex-segregated E14 mouse embryos and transfected with siRNAs to knockdown Kdm6a expression (Kdm6a-KD). We evaluated the effect of Kdm6a-KD on Ngn3 expression, a bHLH transcription factor regulating neuronal sub-specification in hypothalamus. Kdm6a-KD decreased Ngn3 expression in females but not in males, abolishing basal sex differences. Then, we analyzed Kdm6a-KD effect on Ascl1, Pomc, Npy, Sf1, Gad1, and Th expression by RT-qPCR. While Kdm6a-KD downregulated Ascl1 in both sexes equally, we found sex-specific effects for Pomc, Npy, and Th. Pomc and Th expressed higher in female than in male neurons, and Kdm6a-KD reduced their levels only in females, while Npy expressed higher in male than in female neurons, and Kdm6a-KD upregulated its expression only in females. Identical results were found by immunofluorescence for Pomc and Npy neuropeptides. Finally, using ChIP-qPCR, we found higher H3K27me3 levels at Ngn3, Pomc, and Npy promoters in male neurons, in line with Kdm6a higher expression and demethylase activity in females. At all three promoters, Kdm6a-KD induced an enrichment of H3K27me3 only in females. These results indicate that Kdm6a plays a sex-specific role in controlling the expression of transcription factors and neuropeptides critical for the differentiation of hypothalamic neuronal populations regulating food intake and energy homeostasis.

3.
Theranostics ; 11(6): 2691-2705, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33456567

RESUMEN

Rationale: Despite landmark therapy of chronic myelogenous leukemia (CML) with tyrosine kinase inhibitors (TKIs), drug resistance remains problematic. Cancer pathogenesis involves epigenetic dysregulation and in particular, histone lysine demethylases (KDMs) have been implicated in TKI resistance. We sought to identify KDMs with altered expression in CML and define their contribution to imatinib resistance. Methods: Bioinformatics screening compared KDM expression in CML versus normal bone marrow with shRNA knockdown and flow cytometry used to measure effects on imatinib-induced apoptosis in K562 cells. Transcriptomic analyses were performed against KDM6A CRISPR knockout/shRNA knockdown K562 cells along with gene rescue experiments using wildtype and mutant demethylase-dead KDM6A constructs. Co-immunoprecipitation, luciferase reporter and ChIP were employed to elucidate mechanisms of KDM6A-dependent resistance. Results: Amongst five KDMs upregulated in CML, only KDM6A depletion sensitized CML cells to imatinib-induced apoptosis. Re-introduction of demethylase-dead KDM6A as well as wild-type KDM6A restored imatinib resistance. RNA-seq identified NTRK1 gene downregulation after depletion of KDM6A. Moreover, NTRK1 expression positively correlated with KDM6A in a subset of clinical CML samples and KDM6A knockdown in fresh CML isolates decreased NTRK1 encoded protein (TRKA) expression. Mechanistically, KDM6A was recruited to the NTRK1 promoter by the transcription factor YY1 with subsequent TRKA upregulation activating down-stream survival pathways to invoke imatinib resistance. Conclusion: Contrary to its reported role as a tumor suppressor and independent of its demethylase function, KDM6A promotes imatinib-resistance in CML cells. The identification of the KDM6A/YY1/TRKA axis as a novel imatinib-resistance mechanism represents an unexplored avenue to overcome TKI resistance in CML.


Asunto(s)
Histona Demetilasas/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Receptor trkA/genética , Transcripción Genética/genética , Regulación hacia Arriba/genética , Factor de Transcripción YY1/genética , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Células HEK293 , Humanos , Mesilato de Imatinib/farmacología , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Inhibidores de Proteínas Quinasas/farmacología , ARN Interferente Pequeño/genética , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
4.
Mol Cell Biol ; 40(20)2020 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-32817139

RESUMEN

Lysine demethylase 6A (KDM6A), also known as UTX, belongs to the KDM6 family of histone H3 lysine 27 (H3K27) demethylases, which also includes UTY and KDM6B (JMJD3). The KDM6A protein contains six tetratricopeptide repeat (TPR) domains and an enzymatic Jumonji C (JmjC) domain that catalyzes the removal of di- and trimethylation on H3K27. KDM6A physically associates with histone H3 lysine 4 monomethyltransferases MLL3 (KMT2C) and MLL4 (KMT2D). Since its identification as an H3K27 demethylase in 2007, studies have reported KDM6A's critical roles in cell differentiation, development, and cancer. KDM6A is important for differentiation of embryonic stem cells and development of various tissues. Mutations of KDM6A cause Kabuki syndrome. KDM6A is frequently mutated in cancers and functions as a tumor suppressor. KDM6A is redundant with UTY and functions largely independently of its demethylase activity. It regulates gene expression, likely through the associated transcription factors and MLL3/4 on enhancers. However, KDM6A enzymatic activity is required in certain cellular contexts. Functional redundancy between H3K27 demethylase activities of KDM6A and KDM6B in vivo has yet to be determined. Further understanding of KDM6A functions and working mechanisms will provide more insights into enhancer regulation and may help generate novel therapeutic approaches to treat KDM6A-related diseases.


Asunto(s)
Diferenciación Celular/genética , Regulación de la Expresión Génica/genética , Histona Demetilasas/genética , Neoplasias/genética , Dominio Catalítico/genética , Ensamble y Desensamble de Cromatina/genética , Células Madre Embrionarias/citología , Genes Supresores de Tumor , Histona Demetilasas/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji/metabolismo , Proteínas Nucleares/metabolismo
5.
Theriogenology ; 133: 10-21, 2019 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-31051389

RESUMEN

Lysine demethylase 6A (KDM6A, also known as UTX), which belongs a type of histone demethylase, is essential for spermatogenesis and embryo development. However, in pig, the coding sequence, mRNA expression and insertion/deletion (indel) variants of KDM6A remain unclear. Herein, the open-reading frame (ORF) of pig KDM6A gene was cloned and characterized. The entire ORF sequence was 4206 bp in length encoding a deduced protein of 1401 amino acid residues. The phylogenetic tree and signatures of selection on the pig KDM6A gene were calculated, and the results revealed that the pig KDM6A gene has been under strong positively selection. Expression analysis results showed that KDM6A gene was expressed in all tissues tested both in male piglets and adult boars. Meanwhile, with testicular development, the KDM6A expression levels in testis were significantly increased. Additionally, three intronic indels of 11-bp insertion, 17-bp deletion and 16-bp insertion were identified. Association analyses revealed that the 11-bp indel was associated with testicular short perimeter (TSP) (P < 0.01), testicular long perimeter (TLP) (P < 0.01) and testicular weight (TW) (P < 0.01) in 15-day-old Yorkshire and TLP (P < 0.01) in 40-day-old Yorkshire. The 16-bp indel was associated with TLP, TSP and TW (P < 0.05) in 15-day-old Yorkshire. All these findings would provide a foundation for the further research of KDM6A gene and the application of marker-assisted selection to pig breeding.


Asunto(s)
Histona Demetilasas/genética , Mutación INDEL , Porcinos/genética , Secuencia de Aminoácidos , Animales , Clonación Molecular , Estudios de Asociación Genética , Histona Demetilasas/química , Histona Demetilasas/metabolismo , Masculino , Sistemas de Lectura Abierta , Filogenia , ARN Mensajero/metabolismo , Testículo/anatomía & histología , Testículo/crecimiento & desarrollo , Testículo/metabolismo
6.
Stem Cells ; 36(10): 1589-1602, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29873146

RESUMEN

Epicardial adipose tissues (EATs) and vascular tissues may both belong to the mesoepithelial lineage that develops from epicardium-derived progenitor cells (EPDCs) in developing and injured hearts. Very little is known of the molecular mechanisms of EPDC contribution in EAT development and neovascularization in adult heart, which the topic remains a subject of intense therapeutic interest and scientific debate. Here we studied the epigenetic control of stemness and anti-adipogenic and pro-vasculogenic fate of human EPDCs (hEPDCs), through investigating an angiogenic hormone, prokineticin-2 (PK2) signaling via its receptor PKR1. We found that hEPDCs spontaneously undergoes epithelial-to-mesenchymal transformation (EMT), and are not predestined for the vascular lineages. However, PK2 via a histone demethylase KDM6A inhibits EMT, and induces asymmetric division, leading to self-renewal and formation of vascular and epithelial/endothelial precursors with angiogenic potential capable of differentiating into vascular smooth muscle and endothelial cells. PK2 upregulates and activates KDM6A to inhibit repressive histone H3K27me3 marks on promoters of vascular genes (Flk-1 and SM22α) involved in vascular lineage commitment and maturation. In PK2-mediated anti-adipogenic signaling, KDM6A stabilizes and increases cytoplasmic ß-catenin levels to repress peroxisome proliferator-activated receptor-γ expression and activity. Our findings offer additional molecular targets to manipulate hEPDCs-involved tissue repair/regeneration in cardiometabolic and ischemic heart diseases. Stem Cells 2018;36:1589-1602.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/metabolismo , Hormonas Gastrointestinales/metabolismo , Neuropéptidos/metabolismo , Pericardio/citología , Pericardio/metabolismo , Diferenciación Celular/fisiología , Epigénesis Genética , Transición Epitelial-Mesenquimal , Hormonas Gastrointestinales/genética , Histona Demetilasas/metabolismo , Humanos , Neuropéptidos/genética , Proteínas Nucleares/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo
7.
Immunol Res ; 64(2): 345-59, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26411453

RESUMEN

Kabuki syndrome (KS) is a rare multi-systemic disorder characterized by a distinct face, postnatal growth deficiency, mild-to-moderate intellectual disability, skeletal and visceral (mainly cardiovascular, renal, and skeletal) malformations, dermatoglyphic abnormalities. Its cause is related to mutations of two genes: KMT2D (histone-lysine N-methyltransferase 2D) and KDM6A (lysine-specific demethylase 6A), both functioning as epigenetic modulators through histone modifications in the course of embryogenesis and in several biological processes. Epigenetic regulation is defined as the complex of hereditable modifications to DNA and histone proteins that modulates gene expression in the absence of DNA nucleotide sequence changes. Different human disorders are caused by mutations of genes involved in the epigenetic regulation, and not surprisingly, all these share developmental defects, disturbed growth (in excess or defect), multiple congenital organ malformations, and also hematological and immunological defects. In particular, most KS patients show increased susceptibility to infections and have reduced serum immunoglobulin levels, while some suffer also from autoimmune manifestations, such as idiopathic thrombocytopenic purpura, hemolytic anemia, autoimmune thyroiditis, and vitiligo. Herein we review the immunological aspects of KS and propose a novel model to account for the immune dysfunction observed in this condition.


Asunto(s)
Epigénesis Genética , Regulación de la Expresión Génica , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Anomalías Múltiples/genética , Anomalías Múltiples/inmunología , Autoinmunidad , Cara/anomalías , Estudios de Asociación Genética , Enfermedades Hematológicas/complicaciones , Enfermedades Hematológicas/genética , Enfermedades Hematológicas/inmunología , Histonas/metabolismo , Humanos , Infecciones/etiología , Metilación , Mutación , Enfermedades Vestibulares/complicaciones , Enfermedades Vestibulares/genética , Enfermedades Vestibulares/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA