Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 38(22): e101681, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31583744

RESUMEN

Epigenetic modifications operate in concert to maintain cell identity, yet how these interconnected networks suppress alternative cell fates remains unknown. Here, we uncover a link between the removal of repressive histone H3K9 methylation and DNA methylation during the reprogramming of somatic cells to pluripotency. The H3K9me2 demethylase, Kdm3b, transcriptionally controls DNA hydroxymethylase Tet1 expression. Unexpectedly, in the absence of Kdm3b, loci that must be DNA demethylated are trapped in an intermediate hydroxymethylated (5hmC) state and do not resolve to unmethylated cytosine. Ectopic 5hmC trapping precludes the chromatin association of master pluripotency factor, POU5F1, and pluripotent gene activation. Increased Tet1 expression is important for the later intermediates of the reprogramming process. Taken together, coordinated removal of distinct chromatin modifications appears to be an important mechanism for altering cell identity.


Asunto(s)
Linaje de la Célula/genética , Reprogramación Celular , Cromatina/genética , Metilación de ADN , Epigénesis Genética , Histonas/genética , Células Madre Pluripotentes Inducidas/citología , Animales , Células Cultivadas , Proteínas de Unión al ADN/fisiología , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Madre Pluripotentes Inducidas/metabolismo , Histona Demetilasas con Dominio de Jumonji/fisiología , Ratones , Ratones Noqueados , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Proto-Oncogénicas/fisiología
2.
Blood ; 137(5): 610-623, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33538795

RESUMEN

This study was conducted to determine the dosage effect of c-Myc on hematopoiesis and its distinct role in mediating the Wnt/ß-catenin pathway in hematopoietic stem cell (HSC) and bone marrow niche cells. c-Myc haploinsufficiency led to ineffective hematopoiesis by inhibiting HSC self-renewal and quiescence and by promoting apoptosis. We have identified Nr4a1, Nr4a2, and Jmjd3, which are critical for the maintenance of HSC functions, as previously unrecognized downstream targets of c-Myc in HSCs. c-Myc directly binds to the promoter regions of Nr4a1, Nr4a2, and Jmjd3 and regulates their expression. Our results revealed that Nr4a1 and Nr4a2 mediates the function of c-Myc in regulating HSC quiescence, whereas all 3 genes contribute to the function of c-Myc in the maintenance of HSC survival. Adenomatous polyposis coli (Apc) is a negative regulator of the Wnt/ß-catenin pathway. We have provided the first evidence that Apc haploinsufficiency induces a blockage of erythroid lineage differentiation through promoting secretion of IL6 in bone marrow endothelial cells. We found that c-Myc haploinsufficiency failed to rescue defective function of Apc-deficient HSCs in vivo but it was sufficient to prevent the development of severe anemia in Apc-heterozygous mice and to significantly prolong the survival of those mice. Furthermore, we showed that c-Myc-mediated Apc loss induced IL6 secretion in endothelial cells, and c-Myc haploinsufficiency reversed the negative effect of Apc-deficient endothelial cells on erythroid cell differentiation. Our studies indicate that c-Myc has a context-dependent role in mediating the function of Apc in hematopoiesis.


Asunto(s)
Genes myc , Hematopoyesis/fisiología , Proteínas Proto-Oncogénicas c-myb/fisiología , Proteína de la Poliposis Adenomatosa del Colon/fisiología , Anemia/genética , Anemia/prevención & control , Animales , Apoptosis/fisiología , Trasplante de Médula Ósea , Autorrenovación de las Células/fisiología , Ensayo de Unidades Formadoras de Colonias , Células Endoteliales/patología , Células Eritroides/patología , Eliminación de Gen , Genes APC , Haploinsuficiencia , Hematopoyesis/genética , Células Madre Hematopoyéticas , Interleucina-6/fisiología , Histona Demetilasas con Dominio de Jumonji/fisiología , Ratones Mutantes , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/fisiología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/fisiología , Poli I-C/farmacología , Quimera por Radiación , Vía de Señalización Wnt/fisiología
3.
Biochem Soc Trans ; 49(1): 93-105, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33492339

RESUMEN

Alterations in global epigenetic signatures on chromatin are well established to contribute to tumor initiation and progression. Chromatin methylation status modulates several key cellular processes that maintain the integrity of the genome. KDM4A, a demethylase that belongs to the Fe-II dependent dioxygenase family that uses α-ketoglutarate and molecular oxygen as cofactors, is overexpressed in several cancers and is associated with an overall poor prognosis. KDM4A demethylates lysine 9 (H3K9me2/3) and lysine 36 (H3K36me3) methyl marks on histone H3. Given the complexity that exists with these marks on chromatin and their effects on transcription and proliferation, it naturally follows that demethylation serves an equally important role in these cellular processes. In this review, we highlight the role of KDM4A in transcriptional modulation, either dependent or independent of its enzymatic activity, arising from the amplification of this demethylase in cancer. KDM4A modulates re-replication of distinct genomic loci, activates cell cycle inducers, and represses proteins involved in checkpoint control giving rise to proliferative damage, mitotic disturbances and chromosomal breaks, ultimately resulting in genomic instability. In parallel, emerging evidence of non-nuclear substrates of epigenetic modulators emphasize the need to investigate the role of KDM4A in regulating non-nuclear substrates and evaluate their contribution to genomic instability in this context. The existence of promising KDM-specific inhibitors makes these demethylases an attractive target for therapeutic intervention in cancers.


Asunto(s)
Inestabilidad Genómica/genética , Histona Demetilasas con Dominio de Jumonji/fisiología , Animales , Transformación Celular Neoplásica/genética , Histonas/metabolismo , Humanos , Metilación , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Procesamiento Proteico-Postraduccional/genética , Transducción de Señal/genética
4.
PLoS Biol ; 16(11): e2006145, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30500822

RESUMEN

The circadian oscillator is a molecular feedback circuit whose orchestration involves posttranslational control of the activity and protein levels of its components. Although controlled proteolysis of circadian proteins is critical for oscillator function, our understanding of the underlying mechanisms remains incomplete. Here, we report that JmjC domain-containing protein 5 (JMJD5) interacts with CRYPTOCHROME 1 (CRY1) in an F-box/leucine-rich repeat protein 3 (FBXL3)-dependent manner and facilitates targeting of CRY1 to the proteasome. Genetic deletion of JMJD5 results in greater CRY1 stability, reduced CRY1 association with the proteasome, and disruption of circadian gene expression. We also report that in the absence of JMJD5, AMP-regulated protein kinase (AMPK)-induced CRY1 degradation is impaired, establishing JMJD5 as a key player in this mechanism. JMJD5 cooperates with CRY1 to repress circadian locomotor output cycles protein kaput (CLOCK)-brain and muscle ARNT-like protein 1 (BMAL1), thus linking CRY1 destabilization to repressive function. Finally, we find that ablation of JMJD5 impacts FBXL3- and CRY1-related functions beyond the oscillator.


Asunto(s)
Criptocromos/fisiología , Histona Demetilasas con Dominio de Jumonji/fisiología , Complejo de la Endopetidasa Proteasomal/metabolismo , Factores de Transcripción ARNTL/metabolismo , Animales , Relojes Circadianos/genética , Ritmo Circadiano/genética , Criptocromos/genética , Proteínas F-Box/fisiología , Células HEK293 , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Ratones , Ratones Endogámicos C57BL , Células Madre Embrionarias de Ratones , Complejo de la Endopetidasa Proteasomal/fisiología , Dominios Proteicos , Proteolisis
5.
PLoS Biol ; 16(8): e2006134, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30080846

RESUMEN

Cyclic GMP-AMP (cGAMP) synthase (cGAS) stimulator of interferon genes (STING) senses pathogen-derived or abnormal self-DNA in the cytosol and triggers an innate immune defense against microbial infection and cancer. STING agonists induce both innate and adaptive immune responses and are a new class of cancer immunotherapy agents tested in multiple clinical trials. However, STING is commonly silenced in cancer cells via unclear mechanisms, limiting the application of these agonists. Here, we report that the expression of STING is epigenetically suppressed by the histone H3K4 lysine demethylases KDM5B and KDM5C and is activated by the opposing H3K4 methyltransferases. The induction of STING expression by KDM5 blockade triggered a robust interferon response in a cytosolic DNA-dependent manner in breast cancer cells. This response resulted in resistance to infection by DNA and RNA viruses. In human tumors, KDM5B expression is inversely associated with STING expression in multiple cancer types, with the level of intratumoral CD8+ T cells, and with patient survival in cancers with a high level of cytosolic DNA, such as human papilloma virus (HPV)-positive head and neck cancer. These results demonstrate a novel epigenetic regulatory pathway of immune response and suggest that KDM5 demethylases are potential targets for antipathogen treatment and anticancer immunotherapy.


Asunto(s)
Histona Demetilasas/fisiología , Histona Demetilasas con Dominio de Jumonji/fisiología , Proteínas de la Membrana/fisiología , Proteínas Nucleares/fisiología , Proteínas Represoras/fisiología , Línea Celular , Citosol/metabolismo , ADN/metabolismo , Histona Metiltransferasas/fisiología , Histonas/fisiología , Humanos , Inmunidad Innata/fisiología , Inmunoterapia , Interferones/metabolismo , Interferones/fisiología , Células MCF-7 , Proteínas de la Membrana/metabolismo , Transducción de Señal
6.
Mol Cell ; 49(6): 1134-46, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23395003

RESUMEN

Polycomb repressive complex 1 (PRC1) catalyzes lysine 119 monoubiquitylation on H2A (H2AK119ub1) and regulates pluripotency in embryonic stem cells (ESCs). However, the mechanisms controlling the binding of PRC1 to genomic sites and its catalytic activity are poorly understood. Here, we show that Fbxl10 interacts with Ring1B and Nspc1, forming a noncanonical PRC1 that is required for H2AK119ub1 in mouse ESCs. Genome-wide analyses reveal that Fbxl10 preferentially binds to CpG islands and colocalizes with Ring1B on Polycomb target genes. Notably, Fbxl10 depletion causes a decrease in Ring1B binding to target genes and a major loss of H2AK119ub1. Furthermore, genetic analyses demonstrate that Fbxl10 DNA binding capability and integration into PRC1 are required for H2AK119 ubiquitylation. ESCs lacking Fbxl10, like previously characterized Polycomb mutants, cannot differentiate properly. These results demonstrate that Fbxl10 has a key role in regulating Ring1B recruitment to its target genes and H2AK119 ubiquitylation in ESCs.


Asunto(s)
Islas de CpG , Proteínas F-Box/fisiología , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/fisiología , Complejo Represivo Polycomb 1/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Animales , Diferenciación Celular , Línea Celular , Células Madre Embrionarias/enzimología , Células Madre Embrionarias/fisiología , Epigénesis Genética , Genoma , Humanos , Ratones , Modelos Moleculares , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Sitio de Iniciación de la Transcripción , Transcripción Genética
7.
Nucleic Acids Res ; 47(17): 9005-9023, 2019 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-31363749

RESUMEN

CpG islands (CGIs) are associated with the majority of mammalian gene promoters and function to recruit chromatin modifying enzymes. It has therefore been proposed that CGIs regulate gene expression through chromatin-based mechanisms, however in most cases this has not been directly tested. Here, we reveal that the histone H3 lysine 36 (H3K36) demethylase activity of the CGI-binding KDM2 proteins contributes only modestly to the H3K36me2-depleted state at CGI-associated gene promoters and is dispensable for normal gene expression. Instead, we discover that KDM2 proteins play a widespread and demethylase-independent role in constraining gene expression from CGI-associated gene promoters. We further show that KDM2 proteins shape RNA Polymerase II occupancy but not chromatin accessibility at CGI-associated promoters. Together this reveals a demethylase-independent role for KDM2 proteins in transcriptional repression and uncovers a new function for CGIs in constraining gene expression.


Asunto(s)
Islas de CpG/genética , Proteínas F-Box/fisiología , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/fisiología , Regiones Promotoras Genéticas , Transcripción Genética , Animales , Cromatina/enzimología , Cromatina/metabolismo , Metilación de ADN , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Lisina/metabolismo , Ratones , Modelos Genéticos , Células Madre Embrionarias de Ratones/enzimología , Células Madre Embrionarias de Ratones/metabolismo , ARN Polimerasa II/metabolismo
8.
Proc Natl Acad Sci U S A ; 115(16): E3741-E3748, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29610306

RESUMEN

Inactivation of the retinoblastoma gene (RB1) product, pRB, is common in many human cancers. Targeting downstream effectors of pRB that are central to tumorigenesis is a promising strategy to block the growth of tumors harboring loss-of-function RB1 mutations. One such effector is retinoblastoma-binding protein 2 (RBP2, also called JARID1A or KDM5A), which encodes an H3K4 demethylase. Binding of pRB to RBP2 has been linked to the ability of pRB to promote senescence and differentiation. Importantly, genetic ablation of RBP2 is sufficient to phenocopy pRB's ability to induce these cellular changes in cell culture experiments. Moreover, germline Rbp2 deletion significantly impedes tumorigenesis in Rb1+/- mice. The value of RBP2 as a therapeutic target in cancer, however, hinges on whether loss of RBP2 could block the growth of established tumors as opposed to simply delaying their onset. Here we show that conditional, systemic ablation of RBP2 in tumor-bearing Rb1+/- mice is sufficient to slow tumor growth and significantly extend survival without causing obvious toxicity to the host. These findings show that established Rb1-null tumors require RBP2 for growth and further credential RBP2 as a therapeutic target in human cancers driven by RB1 inactivation.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Código de Histonas/fisiología , Histona Demetilasas con Dominio de Jumonji/fisiología , Terapia Molecular Dirigida/métodos , Proteínas de Neoplasias/fisiología , Neoplasias Hipofisarias/enzimología , Proteína de Retinoblastoma/deficiencia , Neoplasias de la Tiroides/enzimología , Alelos , Animales , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Ecocardiografía , Activación Enzimática/efectos de los fármacos , Fibroblastos , Genes de Retinoblastoma , Defectos de los Tabiques Cardíacos/genética , Código de Histonas/efectos de los fármacos , Integrasas/efectos de los fármacos , Histona Demetilasas con Dominio de Jumonji/deficiencia , Histona Demetilasas con Dominio de Jumonji/genética , Ratones , Ratones Endogámicos C57BL , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/terapia , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Tamoxifeno/farmacología , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/terapia , Transgenes/efectos de los fármacos
9.
Int J Mol Sci ; 22(2)2021 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-33478063

RESUMEN

Cancer is a major cause of death worldwide. Epigenetic changes in response to external (diet, sports activities, etc.) and internal events are increasingly implicated in tumor initiation and progression. In this review, we focused on post-translational changes in histones and, more particularly, the tri methylation of lysine from histone 3 (H3K27me3) mark, a repressive epigenetic mark often under- or overexpressed in a wide range of cancers. Two actors regulate H3K27 methylation: Jumonji Domain-Containing Protein 3 demethylase (JMJD3) and Enhancer of zeste homolog 2 (EZH2) methyltransferase. A number of studies have highlighted the deregulation of these actors, which is why this scientific review will focus on the role of JMJD3 and, consequently, H3K27me3 in cancer development. Data on JMJD3's involvement in cancer are classified by cancer type: nervous system, prostate, blood, colorectal, breast, lung, liver, ovarian, and gastric cancers.


Asunto(s)
Histona Demetilasas con Dominio de Jumonji/fisiología , Neoplasias/genética , Animales , Metilación de ADN/genética , Proteína Potenciadora del Homólogo Zeste 2/fisiología , Epigénesis Genética/genética , Femenino , Histona Demetilasas/fisiología , Histonas/metabolismo , Humanos , Masculino , Neoplasias/metabolismo , Neoplasias/patología
10.
Plant Mol Biol ; 102(4-5): 417-430, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31898146

RESUMEN

KEY MESSAGE: We genome-wide identified 28 JmjC domain-containing genes, further spatio-temporal expression profiling and genetic analysis defined them as epigenetic regulators in flowering initiation of Rosa chinensis. The JmjC domain-containing histone demethylases play critical roles in maintaining homeostasis of histone methylations, thus are vital for plant growth and development. Genome-wide identification of the JmjC domain-containing genes have been reported in several species, however, no systematic study has been performed in rose plants. In this paper, we identified 28 JmjC domain-containing genes from the newly published genome database of Rosa chinensis. The JmjC domain-containing proteins in R. chinensis were divided into seven groups, KDM3 was the largest group with 13 members, and JmjC domain-only A and KDM5B were the smallest clades both with only one member. Although all the JmjC domain proteins having a conserved JmjC domain, the gene and protein structure experienced differentiation and specification during the evolution, especially in KDM3 clade, one gene (RcJMJ40) was found carrying site deletions for cofactors Fe (II) and α-KG binding which were crucial for demethylase activities, three genes (RcJMJ41, RcJMJ43 and RcJMJ44) had no intron while two of them had tandem JmjC domains. Spatial expression pattern analysis of these JmjC domain-containing genes in different tissues showed most of them were highly expressed in reproductive tissues such as floral meristem and closed flowers than vegetative tissues, demonstrating their important functions in developmental switch from vegetative to reproductive growth of roses. Temporal expression profiling indicated majority of JmjC domain-containing genes from R. chinensis fluctuated along with floral bud differentiation and development, further proving their essential roles in flower organogenesis. VIGS induced silencing of RcJMJ12 led to delayed flowering time, and decreased the expression levels of flowering integrator such as RcFT, RcSOC1, RcFUL, RcLFY and RcAP1, therefore providing the genetic evidence of RcJMJ12 in flowering initiation. Collectively, spatio-temporal expression profiling and genetic analysis defined the JmjC domain-containing genes as important epigenetic regulators in flower development of R. chinensis.


Asunto(s)
Epigénesis Genética , Flores/genética , Regulación de la Expresión Génica de las Plantas , Histona Demetilasas con Dominio de Jumonji/genética , Meristema/genética , Rosa/genética , Eliminación de Gen , Perfilación de la Expresión Génica , Silenciador del Gen , Genoma de Planta , Histona Demetilasas con Dominio de Jumonji/fisiología , Metilación , Filogenia , Proteínas de Plantas/genética , Dominios Proteicos , Transcriptoma
11.
Development ; 144(4): 567-579, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28087629

RESUMEN

Jmjd2 H3K9 demethylases cooperate in promoting mouse embryonic stem cell (ESC) identity. However, little is known about their importance at the exit of ESC pluripotency. Here, we reveal that Jmjd2c facilitates this process by stabilising the assembly of mediator-cohesin complexes at lineage-specific enhancers. Functionally, we show that Jmjd2c is required in ESCs to initiate appropriate gene expression programs upon somatic multi-lineage differentiation. In the absence of Jmjd2c, differentiation is stalled at an early post-implantation epiblast-like stage, while Jmjd2c-knockout ESCs remain capable of forming extra-embryonic endoderm derivatives. Dissection of the underlying molecular basis revealed that Jmjd2c is re-distributed to lineage-specific enhancers during ESC priming for differentiation. Interestingly, Jmjd2c-bound enhancers are co-occupied by the H3K9-methyltransferase G9a (also known as Ehmt2), independently of its H3K9-modifying activity. Loss of Jmjd2c abrogates G9a recruitment and further destabilises loading of the mediator and cohesin components Med1 and Smc1a at newly activated and poised enhancers in ESC-derived epiblast-like cells. These findings unveil Jmjd2c and G9a as novel enhancer-associated factors, and implicate Jmjd2c as a molecular scaffold for the assembly of essential enhancer-protein complexes with an impact on timely gene activation.


Asunto(s)
Células Madre Embrionarias/citología , Elementos de Facilitación Genéticos , N-Metiltransferasa de Histona-Lisina/fisiología , Histona Demetilasas con Dominio de Jumonji/fisiología , Animales , Proteínas de Ciclo Celular/fisiología , Diferenciación Celular , Linaje de la Célula , Proteínas Cromosómicas no Histona/fisiología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Estratos Germinativos/citología , Histonas/metabolismo , Ratones , Ratones Noqueados , Células Madre Pluripotentes/citología , Unión Proteica , Análisis de Secuencia de ARN , Cohesinas
12.
Biol Reprod ; 103(1): 114-125, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32318688

RESUMEN

Correct reprogramming of epigenetic marks in the donor nucleus is a prerequisite for successful cloning by somatic cell transfer (SCT). In several mammalian species, repressive histone (H) lysine (K) trimethylation (me3) marks, in particular H3K9me3, form a major barrier to somatic cell reprogramming into pluripotency and totipotency. We engineered bovine embryonic fibroblasts (BEFs) for the doxycycline-inducible expression of a biologically active, truncated form of murine Kdm4b, a demethylase that removes H3K9me3 and H3K36me3 marks. Upon inducing Kdm4b, H3K9me3 and H3K36me3 levels were reduced about 3-fold and 5-fold, respectively, compared with noninduced controls. Donor cell quiescence has been previously associated with reduced somatic trimethylation levels and increased cloning efficiency in cattle. Simultaneously inducing Kdm4b expression (via doxycycline) and quiescence (via serum starvation) further reduced global H3K9me3 and H3K36me3 levels by a total of 18-fold and 35-fold, respectively, compared with noninduced, nonstarved control fibroblasts. Following SCT, Kdm4b-BEFs reprogrammed significantly better into cloned blastocysts than noninduced donor cells. However, detrimethylated donors and sustained Kdm4b-induction during embryo culture did not increase the rates of postblastocyst development from implantation to survival into adulthood. In summary, overexpressing Kdm4b in donor cells only improved their reprogramming into early preimplantation stages, highlighting the need for alternative experimental approaches to reliably improve somatic cloning efficiency in cattle.


Asunto(s)
Blastocisto/fisiología , Bovinos/embriología , Reprogramación Celular/fisiología , Clonación de Organismos , Histonas/metabolismo , Técnicas de Transferencia Nuclear , Animales , Reprogramación Celular/genética , Desmetilación , Desarrollo Embrionario/fisiología , Epigénesis Genética , Femenino , Expresión Génica , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/fisiología , Ratones , Regulación hacia Arriba
13.
FASEB J ; 33(9): 10515-10527, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31251083

RESUMEN

Histone Lys-specific demethylases (KDMs) play a key role in many biological processes through epigenetic mechanisms. However, the role of KDMs in inflammatory responses to oral bacterial infection is poorly understood. Here, we show a novel regulatory role of KDM3C in inflammatory responses to oral bacterial infection. KDM3C expression is transiently suppressed in human and mouse macrophages exposed to LPS from Porphyromonas gingivalis (Pg LPS). Loss of KDM3C in both human and mouse macrophages led to notable induction of proinflammatory cytokines in response to Pg LPS stimulation. Also, KDM3C depletion led to strong induction of p65 phosphorylation and accelerated nuclear translocation in cells exposed to Pg LPS. Kdm3C knockout (KO) in mice led to increased alveolar bone destruction upon induction of experimental periodontitis or pulp exposure compared with those of the wild-type (WT) littermates. The Kdm3C KO mice also revealed an increased number of osteoclasts juxtaposed to the bony lesions. We also confirmed enhanced osteoclastogenesis by bone marrow-derived macrophages isolated from the Kdm3C KO compared with the WT controls. These findings suggest an anti-inflammatory function of KDM3C in regulating the inflammatory responses against oral bacterial infection through suppression of NF-κB signaling and osteoclastogenesis.-Lee, J. Y., Mehrazarin, S., Alshaikh, A., Kim, S., Chen, W., Lux, R., Gwack, Y., Kim, R. H., Kang, M. K. Histone Lys demethylase KDM3C demonstrates anti-inflammatory effects by suppressing NF-κB signaling and osteoclastogenesis.


Asunto(s)
Inflamación/prevención & control , Histona Demetilasas con Dominio de Jumonji/fisiología , Enfermedades de la Boca/prevención & control , FN-kappa B/antagonistas & inhibidores , Osteogénesis , Porphyromonas gingivalis/patogenicidad , Animales , Infecciones por Bacteroidaceae/complicaciones , Infecciones por Bacteroidaceae/microbiología , Diferenciación Celular , Citocinas , Histonas , Humanos , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos/toxicidad , Macrófagos/metabolismo , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Noqueados , Enfermedades de la Boca/etiología , Enfermedades de la Boca/metabolismo , Enfermedades de la Boca/patología , FN-kappa B/genética , FN-kappa B/metabolismo , Osteoclastos/metabolismo , Osteoclastos/microbiología , Osteoclastos/patología , Fosforilación , Transducción de Señal
14.
Inflamm Res ; 69(7): 657-666, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32394143

RESUMEN

OBJECTIVES: This study aimed to explore the effects and relative mechanism of JMJD3 on knee osteoarthritis (OA). METHODS: In this study, we first analyzed the expression of JMJD3 in OA cartilage using western blot and immunohistochemistry. In an in vitro study, the effects of GSK-J4, JMJD3 inhibitor, on ATDC-5 chondrocytes were evaluated by CCK-8 assay. Real-time PCR and western blot were used to examine the inhibitory effect of GSK-J4 on the inflammation and ECM degradation of chondrocytes. NF-κB p65 phosphorylation and nuclear translocation were measured by western blot and immunofluorescence. In the animal study, twenty mice were randomized into four experimental groups: sham group, DMM-induced OA + DMSO group, OA + low-dose GSK-J4 group, and OA + high-dose GSK-J4 group. After the treatment, hematoxylin-eosin and safranin O/fast green staining were used to evaluate cartilage degradation of knee joint, with OARSI scores for quantitative assessment of cartilage damage. RESULTS: Our results revealed that JMJD3 was overexpressed in OA cartilage and GSK-J4 could suppress the IL-1ß-induced production of pro-inflammatory cytokines and catabolic enzymes, including IL-6, IL-8, MMP-9 and ADAMTS-5. Consistent with these findings, GSK-J4 could inhibit IL-1ß-induced degradation of collagen II and aggrecan. Mechanistically, GSK-J4 dramatically suppressed IL-1ß-stimulated NF-κB signal pathway activation. In vivo, GSK-J4 prevented cartilage damage in mouse DMM-induced OA model. CONCLUSIONS: This study elucidates the important role of JMJD3 in cartilage degeneration in OA, and our results indicate that JDJM3 may become a novel therapeutic target in OA therapy.


Asunto(s)
Benzazepinas/farmacología , Cartílago/efectos de los fármacos , Condrocitos/efectos de los fármacos , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Osteoartritis/prevención & control , Pirimidinas/farmacología , Agrecanos/metabolismo , Animales , Cartílago/fisiopatología , Línea Celular , Condrocitos/fisiología , Colágeno/metabolismo , Expresión Génica , Humanos , Inflamación/prevención & control , Interleucina-1beta/farmacología , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/fisiología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Osteoartritis/fisiopatología , Ratas , Transducción de Señal/fisiología
15.
Zhonghua Bing Li Xue Za Zhi ; 49(10): 1041-1045, 2020 Oct 08.
Artículo en Zh | MEDLINE | ID: mdl-32992420

RESUMEN

Objective: To observe the expression level of histone demethyltransferase Jmjd3 in patients with pre-eclampsia (PE), and to investigate the possible mechanism of its epigenetic modification in regulating Th1/Th2 imbalance in PE patients. Methods: The mRNA levels of histone demethyltransferase Jmjd3 from peripheral blood mononuclear cells (PBMC) of PE patients and normal pregnant women were detected by RT-PCR. Peripheral serum IFN-γ and IL-4  were detected by ELISA. RT-PCR was used to detect the mRNA levels of Jmjd3, Tbx21 and Cxcr3 in the spleen of PE and control mice. Immunomagnetic beads were used to sort out the initial CD4+ T cells in the spleen of control and PE mice. Western blot was used to detect H3K27me1 and H3K27me3 levels. ChIP analysis was used for H3K27me3 demethylation modification in spleens of PE mice. Results: Compared with normal pregnant women, the mRNA level of Jmjd3 in PBMC of PE patients was significantly increased, the level of IFN-γ in serum was significantly increased, and the level of IL-4 was significantly reduced (P<0.01). Compared with normal control mice, the mRNA level of Jmjd3 in the spleen of PE mice was significantly increased, and the expression of Tbx21 and Cxcr3 was significantly increased in PE mice (P<0.01); the H3K27me3 level of CD4+ T cells in PE mice was significantly reduced (P<0.05), but H3K27me1 was not changed. ChIP analysis showed that CD4+ T cells H3K27me3 in PE group mice were in the Ifng promoter region, compared with control mice. Recruitment was significantly reduced, while recruitment in the promoter region of Il4 was significantly increased (P<0.01). Conclusions: In both PE patients and mice with PE model, the relative expression level of histone demethyltransferase Jmjd3 is significantly up-regulated, which further induces the demethylation of H3K27me3 in the Ifng promoter region and promotes the initial CD4+ T cells to Th1 cell differentiation and development, leading to an imbalance of Th1/Th2, which may be one of the important reasons for the development of preeclampsia.


Asunto(s)
Histonas , Histona Demetilasas con Dominio de Jumonji/fisiología , Preeclampsia/genética , Animales , Epigénesis Genética , Femenino , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Leucocitos Mononucleares , Ratones , Embarazo
16.
Plant J ; 95(6): 961-975, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29923261

RESUMEN

Plant somatic cells can be reprogrammed by in vitro tissue culture methods, and massive genome-wide chromatin remodeling occurs, particularly during callus formation. Since callus tissue resembles root primordium, conversion of tissue identity is essentially required when leaf explants are used. Consistent with the fact that the differentiation state is defined by chromatin structure, which permits limited gene profiles, epigenetic changes underlie cellular reprogramming for changes to tissue identity. Although a histone methylation process suppressing leaf identity during leaf-to-callus transition has been demonstrated, the epigenetic factor involved in activation of root identity remains elusive. Here, we report that JUMONJI C DOMAIN-CONTAINING PROTEIN 30 (JMJ30) stimulates callus formation by promoting expression of a subset of LATERAL ORGAN BOUNDARIES-DOMAIN (LBD) genes that establish root primordia. The JMJ30 protein binds to promoters of the LBD16 and LBD29 genes along with AUXIN RESPONSE FACTOR 7 (ARF7) and ARF19 and activates LBD expression. Consistently, the JMJ30-deficient mutant displays reduced callus formation with low LBD transcript levels. The ARF-JMJ30 complex catalyzes the removal of methyl groups from H3K9me3, especially at the LBD16 and LBD29 loci to activate their expression during leaf-to-callus transition. Moreover, the ARF-JMJ30 complex further recruits ARABIDOPSIS TRITHORAX-RELATED 2 (ATXR2), which promotes deposition of H3K36me3 at the LBD16 and LBD29 promoters, and the tripartite complex ensures stable LBD activation during callus formation. These results indicate that the coordinated epigenetic modifications promote callus formation by establishing root primordium identity.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Cromatina/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Arabidopsis/fisiología , Proteínas de Arabidopsis/fisiología , Reprogramación Celular , Cromatina/fisiología , Desmetilación , Regulación de la Expresión Génica de las Plantas , Histona Demetilasas con Dominio de Jumonji/fisiología , Hojas de la Planta/metabolismo , Hojas de la Planta/fisiología , Raíces de Plantas/metabolismo , Raíces de Plantas/fisiología , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología
17.
Biochem Biophys Res Commun ; 508(2): 576-582, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30514438

RESUMEN

Hepatocellular carcinoma (HCC) is the fifth most common cancer and the third most lethal cancer worldwide. Although gene mutations associated with HCC development have been intensively studied, how epigenetic factors specifically modulate the functional properties of HCC by regulating target gene expression is unclear. Here we demonstrated the overexpression of KDM3B in liver tissue of HCC patients using public RNA-seq data. Ablation of KDM3B by CRISPR/Cas9 retarded the cell cycle and proliferation of hepatocarcinoma HepG2 cells. Approximately 30% of KDM3B knockout cells exhibited mitotic spindle multipolarity as a chromosome instability (CIN) phenotype. RNA-seq analysis of KDM3B knockout revealed significantly down-regulated expression of cell cycle related genes, especially cell proliferation factor CDC123. Furthermore, the expression level of Cyclin D1 was reduced in KDM3B knockout by proteosomal degradation without any change in the expression of CCND1, which encodes Cyclin D1. The results implicate KDM3B as a crucial epigenetic factor in cell cycle regulation that manipulates chromatin dynamics and transcription in HCC, and identifies a potential gene therapy target for effective treatment of HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Redes Reguladoras de Genes , Genes cdc/genética , Histona Demetilasas con Dominio de Jumonji/fisiología , Neoplasias Hepáticas/genética , Carcinoma Hepatocelular/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclina D1/metabolismo , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Histona Demetilasas con Dominio de Jumonji/análisis , Histona Demetilasas con Dominio de Jumonji/genética , Hígado/metabolismo , Neoplasias Hepáticas/metabolismo , Transcripción Genética
18.
Genes Cells ; 23(9): 767-777, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30073721

RESUMEN

Obesity is a serious global health issue; however, the roles of genetics and epigenetics in the onset and progression of obesity are still not completely understood. The aim of this study was to determine the role of Kdm4b, which belongs to a subfamily of histone demethylases, in adipogenesis and fat metabolism in vivo. We established conditional Kdm4b knockout mice. Inactivation of Kdm4b in adipocytes (K4bKO) induced profound obesity in mice on a high fat diet (HFD). The HFD-fed K4bKO mice exhibited an increased volume of fat mass and higher expression levels of adipogenesis-related genes. In contrast, the genes involved in energy expenditure and mitochondrial functions were down-regulated. Supporting these findings, the energy expenditure of Kdm4b-deficient cells was markedly decreased. In addition, progression of glucose intolerance and hepatic steatosis with hepatocellular damages was observed. These data indicate that Kdm4b is a critical regulator of systemic metabolism via enhancing energy expenditure in adipocytes.


Asunto(s)
Tejido Adiposo/patología , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético , Histona Demetilasas con Dominio de Jumonji/fisiología , Enfermedades Metabólicas/patología , Obesidad/patología , Adipogénesis , Tejido Adiposo/metabolismo , Animales , Células Cultivadas , Femenino , Metabolismo de los Lípidos , Masculino , Enfermedades Metabólicas/etiología , Enfermedades Metabólicas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/etiología , Obesidad/metabolismo
19.
Nucleic Acids Res ; 45(11): 6427-6441, 2017 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-28402433

RESUMEN

Epigenetic regulation of chromatin plays a critical role in controlling embryonic stem (ES) cell self-renewal and pluripotency. However, the roles of histone demethylases and activating histone modifications such as trimethylated histone 3 lysine 4 (H3K4me3) in transcriptional events such as RNA polymerase II (RNAPII) elongation and alternative splicing are largely unknown. In this study, we show that KDM5B, which demethylates H3K4me3, plays an integral role in regulating RNAPII occupancy, transcriptional initiation and elongation, and alternative splicing events in ES cells. Depletion of KDM5B leads to altered RNAPII promoter occupancy, and decreased RNAPII initiation and elongation rates at active genes and at genes marked with broad H3K4me3 domains. Moreover, our results demonstrate that spreading of H3K4me3 from promoter to gene body regions, which is mediated by depletion of KDM5B, modulates RNAPII elongation rates and RNA splicing in ES cells. We further show that KDM5B is enriched nearby alternatively spliced exons, and depletion of KDM5B leads to altered levels of H3K4 methylation in alternatively spliced exon regions, which is accompanied by differential expression of these alternatively splice exons. Altogether, our data indicate an epigenetic role for KDM5B in regulating RNAPII elongation and alternative splicing, which may support the diverse mRNA repertoire in ES cells.


Asunto(s)
Empalme Alternativo , Proteínas de Unión al ADN/fisiología , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/fisiología , Células Madre Embrionarias de Ratones/metabolismo , Elongación de la Transcripción Genética , Animales , Línea Celular , Epigénesis Genética , Metilación , Ratones , Regiones Promotoras Genéticas , Unión Proteica , Procesamiento Proteico-Postraduccional , ARN Polimerasa II/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
20.
J Cell Sci ; 129(4): 788-803, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26759175

RESUMEN

Pluripotent embryonic stem cells (ESCs) are characterised by their capacity to self-renew indefinitely while maintaining the potential to differentiate into all cell types of an adult organism. Both the undifferentiated and differentiated states are defined by specific gene expression programs that are regulated at the chromatin level. Here, we have analysed the contribution of the H3K27me2- and H3K27me23-specific demethylases KDM6A and KDM6B to murine ESC differentiation by employing the GSK-J4 inhibitor, which is specific for KDM6 proteins, and by targeted gene knockout (KO) and knockdown. We observe that inhibition of the H3K27 demethylase activity induces DNA damage along with activation of the DNA damage response (DDR) and cell death in differentiating but not in undifferentiated ESCs. Laser microirradiation experiments revealed that the H3K27me3 mark, but not the KDM6B protein, colocalise with γH2AX-positive sites of DNA damage in differentiating ESCs. Lack of H3K27me3 attenuates the GSK-J4-induced DDR in differentiating Eed-KO ESCs. Collectively, our findings indicate that differentiating ESCs depend on KDM6 and that the H3K27me3 demethylase activity is crucially involved in DDR and survival of differentiating ESCs.


Asunto(s)
Histona Demetilasas/fisiología , Histona Demetilasas con Dominio de Jumonji/fisiología , Células Madre Embrionarias de Ratones/fisiología , Animales , Apoptosis , Puntos de Control del Ciclo Celular , Diferenciación Celular , Núcleo Celular/enzimología , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Daño del ADN , Humanos , Ratones , Transporte de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA