Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
1.
Mol Cell ; 66(2): 165-166, 2017 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-28431228

RESUMO

To uncover the precise mechanisms coordinating proliferation and fate choice of stem cells, in this issue of Molecular Cell and in an accompanying paper in Cell Reports, Mazo and colleagues (Petruk et al. 2017a, 2017b) reveal that delayed accumulation of H3K27me3 on nascent DNA is essential to recruit pioneer transcription factors in stem cell differentiation.


Assuntos
Diferenciação Celular/genética , Fatores de Transcrição , Humanos
2.
Mol Cell ; 67(2): 308-321.e6, 2017 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-28732206

RESUMO

Enhancer activation is a critical step for gene activation. Here we report an epigenetic crosstalk at enhancers between the UTX (H3K27 demethylase)-MLL4 (H3K4 methyltransferase) complex and the histone acetyltransferase p300. We demonstrate that UTX, in a demethylase activity-independent manner, facilitates conversion of inactive enhancers in embryonic stem cells to an active (H3K4me1+/H3K27ac+) state by recruiting and coupling the enzymatic functions of MLL4 and p300. Loss of UTX leads to attenuated enhancer activity, characterized by reduced levels of H3K4me1 and H3K27ac as well as impaired transcription. The UTX-MLL4 complex enhances p300-dependent H3K27 acetylation through UTX-dependent stimulation of p300 recruitment, while MLL4-mediated H3K4 monomethylation, reciprocally, requires p300 function. Importantly, MLL4-generated H3K4me1 further enhances p300-dependent transcription. This work reveals a previously unrecognized cooperativity among enhancer-associated chromatin modulators, including a unique function for UTX, in establishing an "active enhancer landscape" and defines a detailed mechanism for the joint deposition of H3K4me1 and H3K27ac.


Assuntos
Cromatina/metabolismo , Proteína p300 Associada a E1A/metabolismo , Células-Tronco Embrionárias/enzimologia , Elementos Facilitadores Genéticos , Histona Desmetilases/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Transcrição Gênica , Ativação Transcricional , Animais , Cromatina/genética , Montagem e Desmontagem da Cromatina , Proteína p300 Associada a E1A/genética , Retroalimentação Fisiológica , Redes Reguladoras de Genes , Células HEK293 , Histona Desmetilases/genética , Histona-Lisina N-Metiltransferase/genética , Histonas/metabolismo , Humanos , Masculino , Metilação , Camundongos , Interferência de RNA , Transfecção
3.
J Cell Physiol ; 239(4): e31178, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38214211

RESUMO

Glioblastoma stem cells (GSCs) exert a crucial influence on glioblastoma (GBM) development, progression, resistance to therapy, and recurrence, making them an attractive target for drug discovery. UTX, a histone H3K27 demethylase, participates in regulating multiple cancer types. However, its functional role in GSCs remains insufficiently explored. This study aims to investigate the role and regulatory mechanism of UTX on GSCs. Analysis of TCGA data revealed heightened UTX expression in glioma, inversely correlating with overall survival. Inhibiting UTX suppressed GBM cell growth and induced apoptosis. Subsequently, we cultured primary GSCs from three patients, observing that UTX inhibition suppressed cell proliferation and induced apoptosis. RNA-seq was performed to analyze the gene expression changes after silencing UTX in GSCs. The results indicated that UTX-mediated genes were strongly correlated with GBM progression and regulatory tumor microenvironment. The transwell co-cultured experiment showed that silencing UTX in the transwell chamber GSCs inhibited the well plate cell proliferation. Protein-protein interaction analysis revealed that periostin (POSTN) played a role in the UTX-mediated transcriptional regulatory network. Replenishing POSTN reversed the effects of UTX inhibition on GSC proliferation and apoptosis. Our study demonstrated that UTX inhibition hindered POSTN expression by enhancing the H3K27me2/3 level, eventually resulting in inhibiting proliferation and promoting apoptosis of patient-derived GSCs. Our findings may provide a novel and effective strategy for the treatment of GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Histona Desmetilases , Células-Tronco Neoplásicas , Humanos , Apoptose/genética , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/patologia , Células-Tronco Neoplásicas/patologia , Periostina , Microambiente Tumoral , Histona Desmetilases/antagonistas & inibidores , Histona Desmetilases/metabolismo
4.
Front Neuroendocrinol ; 71: 101102, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37689249

RESUMO

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.


Assuntos
Epigênese Genética , Neuroesteroides , Feminino , Masculino , Humanos , Neurônios/metabolismo , Neuroesteroides/metabolismo , Testosterona/metabolismo
5.
Biol Reprod ; 110(2): 391-407, 2024 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-37861693

RESUMO

Paternal chromatin undergoes extensive structural and epigenetic changes during mammalian spermatogenesis, producing sperm with an epigenome optimized for the transition to embryogenesis. Lysine demethylase 6a (KDM6A, also called UTX) promotes gene activation in part via demethylation of H3K27me3, a developmentally important repressive modification abundant throughout the epigenome of spermatogenic cells and sperm. We previously demonstrated increased cancer risk in genetically wild-type mice derived from a paternal germ line lacking Kdm6a (Kdm6a cKO), indicating a role for KDM6A in regulating heritable epigenetic states. However, the regulatory function of KDM6A during spermatogenesis is not known. Here, we show that Kdm6a is transiently expressed in spermatogenesis, with RNA and protein expression largely limited to late spermatogonia and early meiotic prophase. Kdm6a cKO males do not have defects in fertility or the overall progression of spermatogenesis. However, hundreds of genes are deregulated upon loss of Kdm6a in spermatogenic cells, with a strong bias toward downregulation coinciding with the time when Kdm6a is expressed. Misregulated genes encode factors involved in chromatin organization and regulation of repetitive elements, and a subset of these genes was persistently deregulated in the male germ line across two generations of offspring of Kdm6a cKO males. Genome-wide epigenetic profiling revealed broadening of H3K27me3 peaks in differentiating spermatogonia of Kdm6a cKO mice, suggesting that KDM6A demarcates H3K27me3 domains in the male germ line. Our findings highlight KDM6A as a transcriptional activator in the mammalian male germ line that is dispensable for spermatogenesis but important for safeguarding gene regulatory state intergenerationally.


Assuntos
Histonas , Meiose , Masculino , Animais , Camundongos , Histonas/genética , Histonas/metabolismo , Sêmen/metabolismo , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Espermatogênese/genética , Espermatogônias/metabolismo , Expressão Gênica , Mamíferos/genética
6.
Cell Commun Signal ; 22(1): 155, 2024 02 29.
Artigo em Inglês | MEDLINE | ID: mdl-38424563

RESUMO

BACKGROUND: Vascular endothelial cells are pivotal in the pathophysiological progression following spinal cord injury (SCI). The UTX (Ubiquitously Transcribed Tetratripeptide Repeat on Chromosome X) serves as a significant regulator of endothelial cell phenotype. The manipulation of endogenous neural stem cells (NSCs) offers a compelling strategy for the amelioration of SCI. METHODS: Two mouse models were used to investigate SCI: NSCs lineage-traced mice and mice with conditional UTX knockout (UTX KO) in endothelial cells. To study the effects of UTX KO on neural differentiation, we harvested extracellular vesicles (EVs) from both UTX KO spinal cord microvascular endothelial cells (SCMECs) and negative control SCMECs. These EVs were then employed to modulate the differentiation trajectory of endogenous NSCs in the SCI model. RESULTS: In our NSCs lineage-traced mice model of SCI, a marked decrease in neurogenesis was observed post-injury. Notably, NSCs in UTX KO SCMECs mice showed enhanced neuronal differentiation compared to controls. RNA sequencing and western blot analyses revealed an upregulation of L1 cell adhesion molecule (L1CAM), a gene associated with neurogenesis, in UTX KO SCMECs and their secreted EVs. This aligns with the observed promotion of neurogenesis in UTX KO conditions. In vivo administration of L1CAM-rich EVs from UTX KO SCMECs (KO EVs) to the mice significantly enhanced neural differentiation. Similarly, in vitro exposure of NSCs to KO EVs resulted in increased activation of the Akt signaling pathway, further promoting neural differentiation. Conversely, inhibiting Akt phosphorylation or knocking down L1CAM negated the beneficial effects of KO EVs on NSC neuronal differentiation. CONCLUSIONS: In conclusion, our findings substantiate that EVs derived from UTX KO SCMECs can act as facilitators of neural differentiation following SCI. This study not only elucidates a novel mechanism but also opens new horizons for therapeutic interventions in the treatment of SCI. Video Abstract.


Assuntos
Vesículas Extracelulares , Molécula L1 de Adesão de Célula Nervosa , Células-Tronco Neurais , Traumatismos da Medula Espinal , Animais , Camundongos , Diferenciação Celular , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Vesículas Extracelulares/metabolismo , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Molécula L1 de Adesão de Célula Nervosa/farmacologia , Células-Tronco Neurais/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/terapia
7.
Genes Dev ; 30(5): 508-21, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26944678

RESUMO

T-cell acute lymphoblastic leukemia (T-ALL) is a heterogeneous group of hematological tumors composed of distinct subtypes that vary in their genetic abnormalities, gene expression signatures, and prognoses. However, it remains unclear whether T-ALL subtypes differ at the functional level, and, as such, T-ALL treatments are uniformly applied across subtypes, leading to variable responses between patients. Here we reveal the existence of a subtype-specific epigenetic vulnerability in T-ALL by which a particular subgroup of T-ALL characterized by expression of the oncogenic transcription factor TAL1 is uniquely sensitive to variations in the dosage and activity of the histone 3 Lys27 (H3K27) demethylase UTX/KDM6A. Specifically, we identify UTX as a coactivator of TAL1 and show that it acts as a major regulator of the TAL1 leukemic gene expression program. Furthermore, we demonstrate that UTX, previously described as a tumor suppressor in T-ALL, is in fact a pro-oncogenic cofactor essential for leukemia maintenance in TAL1-positive (but not TAL1-negative) T-ALL. Exploiting this subtype-specific epigenetic vulnerability, we propose a novel therapeutic approach based on UTX inhibition through in vivo administration of an H3K27 demethylase inhibitor that efficiently kills TAL1-positive primary human leukemia. These findings provide the first opportunity to develop personalized epigenetic therapy for T-ALL patients.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Epigênese Genética , Regulação Neoplásica da Expressão Gênica/genética , Terapia Genética , Histona Desmetilases/genética , Proteínas Nucleares/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/terapia , Proteínas Proto-Oncogênicas/metabolismo , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Histona Desmetilases/metabolismo , Humanos , Proteínas Nucleares/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/fisiopatologia , Proteínas Proto-Oncogênicas/genética , Proteína 1 de Leucemia Linfocítica Aguda de Células T
8.
Cancer Sci ; 114(7): 2787-2797, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37068788

RESUMO

CD8+ T cells play a central role in antitumor immune responses. Epigenetic gene regulation is essential to acquire the effector function of CD8+ T cells. However, the role of Utx, a demethylase of histone H3K27, in antitumor immunity remains unclear. In this study, we examined the roles of Utx in effector CD8+ T-cell differentiation and the antitumor immune response. In a murine tumor-bearing model, an increased tumor size and decreased survival rate were observed in T-cell-specific Utx KO (Utx KO) mice compared with wild-type (WT) mice. The number of CD8+ T cells in tumor-infiltrating lymphocytes (TILs) was significantly decreased in Utx KO mice. We found that the acquisition of effector function was delayed and attenuated in Utx KO CD8+ T cells. RNA sequencing revealed that the expression of effector signature genes was decreased in Utx KO effector CD8+ T cells, while the expression of naïve or memory signature genes was increased. Furthermore, the expression of Cxcr3, which is required for the migration of effector CD8+ T cells to tumor sites, was substantially decreased in Utx KO CD8+ T cells. These findings suggest that Utx promotes CD8+ T-cell-dependent antitumor immune responses partially through epigenetic regulation of the effector function.


Assuntos
Linfócitos T CD8-Positivos , Epigênese Genética , Camundongos , Animais , Linfócitos T CD8-Positivos/metabolismo , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Regulação da Expressão Gênica , Histonas/metabolismo
9.
J Neuroinflammation ; 20(1): 259, 2023 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-37951955

RESUMO

Spinal cord injury (SCI) can prompt an immediate disruption to the blood-spinal cord barrier (BSCB). Restoring the integrity of this barrier is vital for the recovery of neurological function post-SCI. The UTX protein, a histone demethylase, has been shown in previous research to promote vascular regeneration and neurological recovery in mice with SCI. However, it is unclear whether UTX knockout could facilitate the recovery of the BSCB by reducing its permeability. In this study, we systematically studied BSCB disruption and permeability at different time points after SCI and found that conditional UTX deletion in endothelial cells (ECs) can reduce BSCB permeability, decrease inflammatory cell infiltration and ROS production, and improve neurological function recovery after SCI. Subsequently, we used RNA sequencing and ChIP-qPCR to confirm that conditional UTX knockout in ECs can down-regulate expression of myosin light chain kinase (MLCK), which specifically mediates myosin light chain (MLC) phosphorylation and is involved in actin contraction, cell retraction, and tight junctions (TJs) protein integrity. Moreover, we found that MLCK overexpression can increase the ratio of p-MLC/MLC, further break TJs, and exacerbate BSCB deterioration. Overall, our findings indicate that UTX knockout could inhibit the MLCK/p-MLC pathway, resulting in decreased BSCB permeability, and ultimately promoting neurological recovery in mice. These results suggest that UTX is a promising new target for treating SCI.


Assuntos
Cadeias Leves de Miosina , Traumatismos da Medula Espinal , Animais , Camundongos , Ratos , Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Histona Desmetilases/genética , Cadeias Leves de Miosina/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Permeabilidade , Fosforilação , Ratos Sprague-Dawley , Medula Espinal/metabolismo , Traumatismos da Medula Espinal/metabolismo
10.
Cell Immunol ; 386: 104703, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36889216

RESUMO

Epigenetic regulation affects the development and differentiation of iNKT cells. Our previous study found that the number of iNKT cells in thymus of RA mice was reduced and the ratio of subsets was unbalanced, but the related mechanism remains unclear. We adopted an adoptive infusion of iNKT2 cells with specific phenotypes and functions to RA mice and used the α-Galcer treatment group as control. The findings revealed that: 1. Adoptive treatment of iNKT cells decreased the proportion of iNKT1 and iNKT17 subsets in the thymus of RA mice, and increased the proportion of iNKT2 subsets. 2. Following treatment with iNKT cells, the expression of PLZF in thymus DP T cells was increased whereas the expression of T-bet in thymus iNKT cells was decreased in RA mice. 3. Adoptive therapy reduced the modification levels of H3Kb7me3 and H3K4me3 in the promoter regions of Zbtb16 (encoding PLZF) and Tbx21 (encoding T-bet) gene in thymus DP T cells and iNKT cells, and the reduction of H3K4me3 was particularly significant in the cell treatment group. Furthermore, adoptive therapy also upregulated the expression of UTX (histone demethylase) in thymus lymphocytes of RA mice. As a result, it is hypothesized that adoptive therapy of iNKT2 cells may affect the level of histone methylation in the promoter region of important transcription factor genes for iNKT development and differentiation, thereby directly or indirectly correcting the imbalance of iNKT subsets in the thymus of RA mice. These findings offer a fresh rationale and concept for the management of RA that targets.


Assuntos
Epigênese Genética , Células T Matadoras Naturais , Camundongos , Animais , Células T Matadoras Naturais/metabolismo , Timo , Diferenciação Celular , Timócitos , Camundongos Endogâmicos C57BL
11.
FASEB J ; 36(12): e22662, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36412518

RESUMO

Recent studies have demonstrated that epigenetic modifications are deeply involved in neurogenesis; however, the precise mechanisms remain largely unknown. To determine the role of UTX (also known as KDM6A), a demethylase of histone H3K27, in neural development, we generated Utx-deficient mice in neural stem/progenitor cells (NSPCs). Since Utx is an X chromosome-specific gene, the genotypes are sex-dependent; female mice lose both Utx alleles (UtxΔ/Δ ), and male mice lose one Utx allele yet retain one Uty allele, the counterpart of Utx on the Y chromosome (UtxΔ/Uty ). We found that UtxΔ/Δ mice exhibited fetal ventriculomegaly and died soon after birth. Immunofluorescence staining and EdU labeling revealed a significant increase in NSPCs and a significant decrease in intermediate-progenitor and differentiated neural cells. Molecular analyses revealed the downregulation of pathways related to DNA replication and increased H3K27me3 levels around the transcription start sites in UtxΔ/Δ NSPCs. These results indicate that UTX globally regulates the expression of genes required for proper neural development in NSPCs, and UTX deficiency leads to impaired cell cycle exit, reduced differentiation, and neonatal death. Interestingly, although UtxΔ/Uty mice survived the postnatal period, most died of hydrocephalus, a clinical feature of Kabuki syndrome, a congenital anomaly involving UTX mutations. Our findings provide novel insights into the role of histone modifiers in neural development and suggest that UtxΔ/Uty mice are a potential disease model for Kabuki syndrome.


Assuntos
Histonas , Hidrocefalia , Animais , Feminino , Masculino , Camundongos , Desenvolvimento Fetal , Histona Desmetilases/genética , Hidrocefalia/genética , Neurogênese , Células-Tronco , Células-Tronco Neurais
12.
J Nanobiotechnology ; 21(1): 225, 2023 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-37454119

RESUMO

Macrophages polarized to the M2 subtype after spinal cord injury (SCI) are beneficial for promoting neurological recovery. The crosstalk between endothelial cells (ECs) and macrophages is crucial for the imbalance between proinflammatory and pro-resolving responses caused by macrophage heterogeneity; however, this crosstalk is strengthened post-SCI, leading to inflammatory cascades and second damage. As a powerful means to regulate gene expression, epigenetic regulation of the interaction between immune cells and ECs in SCI is still largely unknown. Our previous research demonstrated that the histone demethylase UTX deletion in ECs (UTX-/- ECs) promotes neurological recovery, while the precise mechanism is unrevealed. Here, we discovered that UTX-/- ECs polarize macrophages toward the M2 subtype post-SCI. Macrophage deficiency could block the neurological recovery caused by the knockdown of UTX. The exosomes from UTX-/- ECs mediate this crosstalk. In addition, we found UTX, H3K27, and miR-467b-3p/Sfmbt2 promoters forming a regulatory complex that upregulates the miR-467b-3p in UTX-/- ECs. And then, miR-467b-3p transfers to macrophages by exosomes and activates the PI3K/AKT/mTOR signaling by decreasing PTEN expression, finally polarizing macrophage to the M2 subtype. This study reveals a mechanism by epigenetic regulation of ECs-macrophages crosstalk and identifies potential targets, which may provide opportunities for treating SCI.


Assuntos
MicroRNAs , Traumatismos da Medula Espinal , Humanos , Células Endoteliais/metabolismo , Epigênese Genética , Fosfatidilinositol 3-Quinases/metabolismo , Macrófagos/metabolismo , Traumatismos da Medula Espinal/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo
13.
Adv Exp Med Biol ; 1433: 139-165, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37751139

RESUMO

Histone lysine methylation is a major epigenetic modification that participates in several cellular processes including gene regulation and chromatin structure. This mark can go awry in disease contexts such as cancer. Two decades ago, the discovery of histone demethylase enzymes thirteen years ago sheds light on the complexity of the regulation of this mark. Here we address the roles of lysine demethylases JMJD3 and UTX in physiological and disease contexts. The two demethylases play pivotal roles in many developmental and disease contexts via regulation of di- and trimethylation of lysine 27 on histone H3 (H3K27me2/3) in repressing gene expression programs. JMJD3 and UTX participate in several biochemical settings including methyltransferase and chromatin remodeling complexes. They have histone demethylase-dependent and -independent activities and a variety of context-specific interacting factors. The structure, amounts, and function of the demethylases can be altered in disease due to genetic alterations or aberrant gene regulation. Therefore, academic and industrial initiatives have targeted these enzymes using a number of small molecule compounds in therapeutic approaches. In this chapter, we will touch upon inhibitor formulations, their properties, and current efforts to test them in preclinical contexts to optimize their therapeutic outcomes. Demethylase inhibitors are currently used in targeted therapeutic approaches that might be particularly effective when used in conjunction with systemic approaches such as chemotherapy.


Assuntos
Epigênese Genética , Lisina , Epigenômica , Histona Desmetilases/genética , Histonas/genética
14.
Biochem Biophys Res Commun ; 590: 132-138, 2022 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-34974301

RESUMO

Osteocytes are master regulators of skeletal homeostasis. However, little is known about the molecular mechanism of their differentiation. Epigenetic regulations, especially H3K27me3 modification, play critical roles in cell differentiation. Here, we found that H3K27me3 in the loci of osteocyte-expressing genes decreased during osteocyte differentiation and that H3K27me3 demethylase, Utx, was bound to the loci of those genes. To investigate the physiological functions of Utx in vivo, we generated late osteoblast-to-osteocyte specific Utx knockout mice using Dmp1-cre mice (UtxΔOcy/ΔOcy). Micro CT analyses showed that UtxΔOcy/ΔOcy displayed osteopenic phenotypes with lower bone volume and trabecular number, and greater trabecular separation. Bone histomorphometric analysis showed that bone mineralization and formation were significantly lower in UtxΔOcy/ΔOcy. Furthermore, Dmp1 expression and the number of osteocytes were significantly decreased in UtxΔOcy/ΔOcy. These results suggest that Utx in Dmp1-expressing osteoblast/osteocyte positively regulates osteoblast-to-osteocyte differentiation through H3K27me3 modifications in osteocyte genes. Our results provide new insight into the molecular mechanism of osteocyte differentiation.


Assuntos
Diferenciação Celular , Histona Desmetilases/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Osteoblastos/citologia , Osteócitos/citologia , Animais , Sequência de Bases , Doenças Ósseas Metabólicas/genética , Osso Esponjoso/diagnóstico por imagem , Osso Esponjoso/patologia , Contagem de Células , Diferenciação Celular/genética , Regulação para Baixo/genética , Epigenoma , Loci Gênicos , Histona Desmetilases/deficiência , Metilação , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoblastos/metabolismo , Osteócitos/metabolismo , Fenótipo , Processamento de Proteína Pós-Traducional , Transcriptoma/genética
15.
Pharmacol Res ; 175: 106021, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34883214

RESUMO

Acetaminophen (APAP) overdose is a major cause of acute liver failure, while the underlying mechanisms of APAP hepatotoxicity are not fully understood. Recently, emerging evidence suggests that epigenetic enzymes play roles in APAP-induced liver injury. Here, we found that Utx (ubiquitously transcribed tetratricopeptide repeat, X chromosome, also known as KDM6A), a X-linked histone demethylase which removes the di- and tri-methyl groups from histone H3K27, was markedly induced in the liver of APAP-overdosed female mice. Hepatic deletion of Utx suppressed APAP overdose-induced hepatotoxicity in female but not male mice. RNA-sequencing analysis suggested that Utx deficiency in female mice upregulated antitoxic phase II conjugating enzymes, including sulfotransferase family 2 A member 1 (Sult2a1), thus reduces the amount of toxic APAP metabolites in injured liver; while Utx deficiency also alleviated ER stress through downregulating transcription of ER stress genes including Atf4, Atf3, and Chop. Mechanistically, Utx promoted transcription of ER stress related genes in a demethylase activity-dependent manner, while repressed Sult2a1 expression through mediating H3K27ac levels independent of its demethylase activity. Moreover, overexpression of Sult2a1 in the liver of female mice rescued APAP-overdose induced liver injury. Together, our results indicated a novel UTX-Sult2a1 axis for the prevention or treatment of APAP-induced liver injury.


Assuntos
Acetaminofen , Analgésicos não Narcóticos , Doença Hepática Induzida por Substâncias e Drogas , Histona Desmetilases , Animais , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Overdose de Drogas/metabolismo , Estresse do Retículo Endoplasmático , Feminino , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Oxidativo , Caracteres Sexuais , Sulfotransferases/genética
16.
Camb Q Healthc Ethics ; 31(1): 73-82, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35049456

RESUMO

This paper argues that uterine transplants are a potentially dangerous distraction from the development of alternative methods of providing reproductive options for women with absolute uterine factor infertility (AUFI). We consider two alternatives in particular: the bioengineering of wombs using stem cells (which would carry fewer risks than uterine transplants) and ectogenesis (which would not require surgical intervention for either the prospective mother with AUFI or a womb donor). Whether biologically or mechanically engineered, these womb replacements could provide a way for women to have children, including genetically related offspring for those who would value this possibility. Most importantly, this alternative would avoid the challenge of sourcing wombs for transplant, a practice that we argue would likely be exploitative and unethical. Continued research into bioengineering and ectogenesis will therefore remain morally important despite the recent development of uterine transplantation, even if the procedure reaches routine clinical application.


Assuntos
Ectogênese , Infertilidade Feminina , Criança , Feminino , Humanos , Infertilidade Feminina/cirurgia , Estudos Prospectivos , Reprodução , Útero/transplante
17.
BMC Cancer ; 21(1): 979, 2021 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-34465286

RESUMO

BACKGROUND: Ubiquitously transcribed tetratricopeptide repeat on chromosome X (UTX) has been identified as a histone 3 lysine 27 (H3K27) demethylase and acted as a tumor suppressor gene or oncogenic function. The current study was to explore the significance of UTX in oral tongue squamous cell carcinoma (OTSCC) patients who received surgical resection. METHODS: A total of 148 OTSCC patients who underwent surgical resection were identified, including 64 patients (43%) with overexpression of UTX and 84 patients (57%) harboring low expression of UTX. We also used two OTSCC cell lines, SAS and Cal 27, to determine the modulation of cancer. Chi-square test was used to investigate the difference of categorical variables between the groups; survival outcome was analyzed using the Kaplan-Meier method in univariate analysis, and a Cox regression model was performed for multivariate analyses. RESULTS: Univariate and multivariate analyses showed overexpression of UTX were significantly related to worse disease-free survival (P = 0.028) and overall survival (P = 0.029). The two OTSCC cell lines were treated with GSK-J4, a potent inhibitor of UTX, and transwell migration and invasion assays showed an inhibitory effect with a dose-dependent manner. In addition, western blot analyses also revealed the inhibition of cell cycle and epithelial-mesenchymal transition. CONCLUSION: Our study suggests that UTX plays an important role in the process of OTSCC and overexpression of UTX may predict poor prognosis in OTSCC patients who received surgical resection.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/patologia , Histona Desmetilases/metabolismo , Neoplasias Bucais/patologia , Neoplasias da Língua/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/cirurgia , Progressão da Doença , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Bucais/metabolismo , Neoplasias Bucais/cirurgia , Prognóstico , Estudos Retrospectivos , Taxa de Sobrevida , Neoplasias da Língua/metabolismo , Neoplasias da Língua/cirurgia
18.
FASEB J ; 34(3): 3956-3968, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31961023

RESUMO

Heterochromatin protein (HP) 1γ, a component of heterochromatin in eukaryotes, is involved in H3K9 methylation. Although HP1γ is expressed strongly in neural tissues and neural stem cells, its functions are unclear. To elucidate the roles of HP1γ, we analyzed HP1γ -deficient (HP1γ KO) mouse embryonic neurospheres and determined that HP1γ KO neurospheres tended to differentiate after quaternary culture. Several genes normally expressed in neuronal cells were upregulated in HP1γ KO undifferentiated neurospheres, but not in the wild type (WT). Compared to that in the control neurospheres, the occupancy of H3K27me3 was lower around the transcription start sites (TSSs) of these genes in HP1γ KO neurospheres, while H3K9me2/3, H3K4me3, and H3K27ac amounts remained unchanged. Moreover, amounts of the H3K27me2/3 demethylases, UTX, and JMJD3, were increased around the TSSs of these genes. Treatment with GSK-J4, an inhibitor of H3K27 demethylases, decreased the expression of genes upregulated in HP1γ KO neurospheres, along with an increase of H3K27me3 amounts. Therefore, in murine neurospheres, HP1γ protected the promoter sites of differentiated cell-specific genes against H3K27 demethylases to repress the expression of these genes. A better understanding of central cellular processes such as histone methylation will help elucidate critical events such as cell-specific gene expression, epigenetics, and differentiation.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Histonas/metabolismo , Animais , Proliferação de Células/genética , Proliferação de Células/fisiologia , Imunoprecipitação da Cromatina , Proteínas Cromossômicas não Histona/genética , Imunofluorescência , Ontologia Genética , Imageamento por Ressonância Magnética , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sítio de Iniciação de Transcrição/fisiologia
19.
Proc Natl Acad Sci U S A ; 115(17): E3978-E3986, 2018 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-29632194

RESUMO

Lung cancer is the leading cause of cancer-related death worldwide. Inactivation of tumor suppressor genes (TSGs) promotes lung cancer malignant progression. Here, we take advantage of the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9-mediated somatic gene knockout in a KrasG12D/+ mouse model to identify bona fide TSGs. From individual knockout of 55 potential TSGs, we identify five genes, including Utx, Ptip, Acp5, Acacb, and Clu, whose knockout significantly promotes lung tumorigenesis. These candidate genes are frequently down-regulated in human lung cancer specimens and significantly associated with survival in patients with lung cancer. Through crossing the conditional Utx knockout allele to the KrasG12D/+ mouse model, we further find that Utx deletion dramatically promotes lung cancer progression. The tumor-promotive effect of Utx knockout in vivo is mainly mediated through an increase of the EZH2 level, which up-regulates the H3K27me3 level. Moreover, the Utx-knockout lung tumors are preferentially sensitive to EZH2 inhibitor treatment. Collectively, our study provides a systematic screening of TSGs in vivo and identifies UTX as an important epigenetic regulator in lung tumorigenesis.


Assuntos
Sistemas CRISPR-Cas , Transformação Celular Neoplásica/metabolismo , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Histona Desmetilases/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Experimentais/metabolismo , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Histona Desmetilases/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Knockout , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
20.
Mol Ther ; 27(12): 2134-2146, 2019 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31495776

RESUMO

The regeneration of the blood vessel system post spinal cord injury (SCI) is essential for the repair of neurological function. As a significant means to regulate gene expression, epigenetic regulation of angiogenesis in SCI is still largely unknown. Here, we found that Ubiquitously Transcribed tetratricopeptide repeat on chromosome X (UTX), the histone H3K27 demethylase, increased significantly in endothelial cells post SCI. Knockdown of UTX can promote the migration and tube formation of endothelial cells. The specific knockout of UTX in endothelial cells enhanced angiogenesis post SCI accompanied with improved neurological function. In addition, we found regulation of UTX expression can change the level of microRNA 24 (miR-24) in vitro. The physical binding of UTX to the promotor of miR-24 was indicated by chromatin immunoprecipitation (ChIP) assay. Meanwhile, methylation sequencing of endothelial cells demonstrated that UTX could significantly decrease the level of methylation in the miR-24 promotor. Furthermore, miR-24 significantly abolished the promoting effect of UTX deletion on angiogenesis in vitro and in vivo. Finally, we predicted the potential target mRNAs of miR-24 related to angiogenesis. We indicate that UTX deletion can epigenetically promote the vascular regeneration and functional recovery post SCI by forming a regulatory network with miR-24.


Assuntos
Células Endoteliais/citologia , Epigênese Genética , Deleção de Genes , Histona Desmetilases/fisiologia , Regeneração , Traumatismos da Medula Espinal/terapia , Animais , Movimento Celular , Proliferação de Células , Metilação de DNA , Células Endoteliais/fisiologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , Regiões Promotoras Genéticas , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/patologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa