Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Biol Rep ; 51(1): 778, 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38904842

RESUMO

BACKGROUND: SETDB1 (SET domain bifurcated-1) is a histone H3-lysine 9 (H3K9)-specific methyltransferase that mediates heterochromatin formation and repression of target genes. Despite the assumed functional link between DNA methylation and SETDB1-mediated H3K9 trimethylations, several studies have shown that SETDB1 operates autonomously of DNA methylation in a region- and cell-specific manner. This study analyzes SETDB1-null HAP1 cells through a linked methylome and transcriptome analysis, intending to explore genes controlled by SETDB1-involved DNA methylation. METHODS AND RESULTS: We investigated SETDB1-mediated regulation of DNA methylation and gene transcription in human HAP1 cells using reduced-representation bisulfite sequencing (RRBS) and RNA sequencing. While two-thirds of differentially methylated CpGs (DMCs) in genic regions were hypomethylated in SETDB1-null cells, we detected a plethora of C2H2-type zinc-finger protein genes (C2H2-ZFP, 223 of 749) among the DMC-associated genes. Most C2H2-ZFPs with DMCs in their promoters were found hypomethylated in SETDB1-KO cells, while other non-ZFP genes with promoter DMCs were not. These C2H2-ZFPs with DMCs in their promoters were significantly upregulated in SETDB1-KO cells. Similarly, C2H2-ZFP genes were upregulated in SETDB1-null 293T cells, suggesting that SETDB1's function in ZFP gene repression is widespread. There are several C2H2-ZFP gene clusters on chromosome 19, which were selectively hypomethylated in SETDB1-KO cells. CONCLUSIONS: SETDB1 collectively and specifically represses a substantial fraction of the C2H2-ZFP gene family. Through the en-bloc silencing of a set of ZFP genes, SETDB1 may help establish a panel of ZFP proteins that are expressed cell-type specifically and thereby can serve as signature proteins for cellular identity.


Assuntos
Metilação de DNA , Histona-Lisina N-Metiltransferase , Dedos de Zinco , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Dedos de Zinco/genética , Metilação de DNA/genética , Regiões Promotoras Genéticas/genética , Regulação para Cima/genética , Desmetilação do DNA , Linhagem Celular , Ilhas de CpG/genética , Deleção de Genes , Histonas/metabolismo , Histonas/genética
2.
Biochem Biophys Res Commun ; 637: 17-22, 2022 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-36375246

RESUMO

The microRNA (miRNA) gene cluster on chromosome 19, C19MC, is the largest primate-specific miRNA gene cluster. The 46 homologous miRNA genes in C19MC are highly expressed in the placenta, but repressed in other tissues by DNA methylation. Here, we found that the SET domain bifurcated 1(SETDB1), a histone H3-lysine 9 (H3K9)-specific methyltransferase 1, transcriptionally controls C19MC miRNA genes in a coordinated manner in human HAP1 cells. SETDB1 knockout (KO) resulted in the overexpression of C19MC miRNA genes, which was accompanied by a reduction of H3K9 trimethylation (H3K9me3) in the cluster. We found that SETDB1 specifically binds to and modifies the upstream promoter locus of C19MC with H3K9me3, suggesting its role as a C19MC repressor. Overexpression of C19MC miRNA genes was not related to DNA methylation because cytosine methylation levels were not altered in the C19MC of SETDB1 KO cells, indicating that SETDB1 KO does not cause DNA demethylation in the C19MC promoter and body regions. In conclusion, our results suggest that SETDB1 binding and H3K9 methylation at the C19MC promoter and body regions are responsible for the coordinated regulation of miRNA genes in the cluster.


Assuntos
Histona-Lisina N-Metiltransferase , Histonas , MicroRNAs , Humanos , Metilação de DNA , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Regiões Promotoras Genéticas
3.
Nucleic Acids Res ; 48(15): 8431-8444, 2020 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-32667642

RESUMO

Genome-wide passive DNA demethylation in cleavage-stage mouse embryos is related to the cytoplasmic localization of the maintenance methyltransferase DNMT1. However, recent studies provided evidences of the nuclear localization of DNMT1 and its contribution to the maintenance of methylation levels of imprinted regions and other genomic loci in early embryos. Using the DNA adenine methylase identification method, we identified Dnmt1-binding regions in four- and eight-cell embryos. The unbiased distribution of Dnmt1 peaks in the genic regions (promoters and CpG islands) as well as the absence of a correlation between the Dnmt1 peaks and the expression levels of the peak-associated genes refutes the active participation of Dnmt1 in the transcriptional regulation of genes in the early developmental period. Instead, Dnmt1 was found to associate with genomic retroelements in a greatly biased fashion, particularly with the LINE1 (long interspersed nuclear elements) and ERVK (endogenous retrovirus type K) sequences. Transcriptomic analysis revealed that the transcripts of the Dnmt1-enriched retroelements were overrepresented in Dnmt1 knockdown embryos. Finally, methyl-CpG-binding domain sequencing proved that the Dnmt1-enriched retroelements, which were densely methylated in wild-type embryos, became demethylated in the Dnmt1-depleted embryos. Our results indicate that Dnmt1 is involved in the repression of retroelements through DNA methylation in early mouse development.


Assuntos
DNA (Citosina-5-)-Metiltransferase 1/genética , Metilação de DNA/genética , Desenvolvimento Embrionário/genética , Genômica , Retroelementos/genética , Animais , Ilhas de CpG/genética , Proteínas de Ligação a DNA/genética , Embrião de Mamíferos , Perfilação da Expressão Gênica , Genoma/genética , Impressão Genômica/genética , Camundongos , Fatores de Transcrição/genética
4.
Bioessays ; 40(9): e1800058, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29897144

RESUMO

In human cancers, histone methyltransferase SETDB1 (SET domain, bifurcated 1) is frequently overexpressed but its significance in carcinogenesis remains elusive. A recent study shows that SETDB1 downregulation induces de-repression of retroelements and innate immunity in cancer cells. The possibility of SETDB1 functioning as a surveillant of retroelement expression is discussed in this study: the cytoplasmic presence of retroelement-derived nucleic acids (RdNAs) drives SETDB1 into the nucleus by the RNA-interference route, rendering the corresponding retroelement transcriptionally inert. These RdNAs could, therefore, be signals of genome instability sent out for SETDB1 present in the cytoplasm to maintain genome integrity.


Assuntos
Histona Metiltransferases/imunologia , Imunidade Inata/imunologia , Vigilância Imunológica/imunologia , Ácidos Nucleicos/imunologia , Proteínas Metiltransferases/imunologia , Retroelementos/imunologia , Animais , Citoplasma/imunologia , Humanos , Neoplasias/imunologia , Transcrição Gênica/imunologia
5.
Nucleic Acids Res ; 42(22): 13545-56, 2014 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-25183519

RESUMO

In mammals, RNA interference is primarily a post-transcriptional mechanism. Evidence has accumulated for additional role in transcriptional gene silencing (TGS) but the question for a good paradigm for small interfering antigene RNA (agRNA)-induced chromatin modification remains unanswered. Here, we show that SETDB1, a histone H3-lysine 9 (H3K9)-specific methyltransferase, cooperates with Argonaute-2 (AGO2) and plays an essential role in agRNA-induced TGS. The androgen receptor (AR) gene was transcriptionally silenced by agRNA targeted to its promoter, and we show that this repression was mitigated by knockdown of SETDB1 or AGO2. Chromatin immunoprecipitation demonstrated that agRNA-driven AGO2 was first targeted to the AR promoter, followed by SETDB1. SIN3A and HDAC1/2, the components of the SIN3-HDAC complex, immunoprecipitated with SETDB1, and localized at the agRNA-targeted promoter. Agreeing with the presence of SETDB1, trimethyl-H3K9 was enriched in the AR promoter. Both EZH2 and trimethyl-H3K27 were also present in the targeted locus; accordingly, EZH2 immunoprecipitated with SETDB1. DNA methylation level was not significantly changed, suggesting the absence of de novo methylating activity in agRNA-induced AR promoter. Our results demonstrate that SETDB1, together with AGO2, plays an essential role in TGS through recruiting chromatin remodeler and/or other modifiers, consequently creating a repressive chromatin milieu at the targeted promoter.


Assuntos
Proteínas Argonautas/metabolismo , Inativação Gênica , Proteínas Metiltransferases/metabolismo , Receptores Androgênicos/genética , Transcrição Gênica , Animais , Linhagem Celular Tumoral , Proteína Potenciadora do Homólogo 2 de Zeste , Histona-Lisina N-Metiltransferase , Histonas/metabolismo , Camundongos , Células NIH 3T3 , Complexo Repressor Polycomb 2/metabolismo , Regiões Promotoras Genéticas , Proteínas Metiltransferases/fisiologia , RNA não Traduzido/análise
6.
Curr Issues Mol Biol ; 17: 1-10, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24698782

RESUMO

The histone methyltransferase SETDB1 contributes to the silencing of local chromatin and the target specificity appears to be determined through various proteins that SETDB1 interacts with. This fundamental function endows SETDB1 with specialized roles in embryonic cells. Keeping the genomic and transcriptomic integrity via proviral silencing and maintaining the pluripotency by repressing the differentiation-associated genes have been demonstrated as the roles of SETDB1 in embryonic stem cells. In early developing embryos, SETDB1 exhibits characteristic nuclear mobilizations that might account for its pleiotropic roles in these rapidly changing cells as well. Early lethality of SETDB1-null embryos, along with other immunolocalization findings, suggests that SETDB1 is necessary for reprogramming and preparing the genomes of zygotes and pluripotent cells for the post-implantation developmental program.


Assuntos
Desenvolvimento Embrionário , Células-Tronco Embrionárias/metabolismo , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Animais , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica no Desenvolvimento , Histona-Lisina N-Metiltransferase/química , Histonas/metabolismo , Humanos , Espaço Intracelular/metabolismo , Transporte Proteico
7.
Genes Cells ; 18(8): 694-703, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23782009

RESUMO

Setdb1 is a histone H3-lysine 9 (H3K9)-specific methyltransferase that interacts with various transcriptional regulators to induce local heterochromatin formation and participates as an indispensable component in building promyelocytic leukemia nuclear body (PML-NB), which is involved in various biological processes. We studied the effects of Setdb1 over-expression. We unexpectedly observed that exogenously expressed GFP-Setdb1 was retained in the cytoplasm, whereas endogenous Setdb1 showed a punctate nuclear signal. Leptomycin B (LMB) treatment, which blocks protein export from the nucleus, showed that entry of GFP-Setdb1 to the nucleus was regulated and that GFP-Setdb1 in the nucleus could localize at PML-NB as endogenous Setdb1. An analysis of Setdb1 deletion constructs showed that the N-terminal region was related to the nuclear export of Setdb1; supporting this, we detected two nuclear export signal motifs in this region. This N-terminal region had a SUMO interaction motif (SIM) whose mutation greatly reduced the ability of Setdb1 to associate with PML-NB and thus resulted in the disaggregation of PML-NB structure. We therefore presume that the cytoplasmic retention of over-expressed Setdb1 occurs as part of a regulatory mechanism to set a tight limit on the nuclear activity of Setdb1, whose excess activity might result in random and haphazard chromatin modifications that cause globally aberrant gene expression.


Assuntos
Transporte Ativo do Núcleo Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Animais , Células Cultivadas , Regulação da Expressão Gênica , Histona-Lisina N-Metiltransferase/química , Camundongos , Células NIH 3T3 , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína da Leucemia Promielocítica , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo
8.
Cell Host Microbe ; 32(2): 244-260.e11, 2024 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-38198924

RESUMO

Although early life colonization of commensal microbes contributes to long-lasting immune imprinting in host tissues, little is known regarding the pathophysiological consequences of postnatal microbial tuning of cutaneous immunity. Here, we show that postnatal exposure to specific skin commensal Staphylococcus lentus (S. lentus) promotes the extent of atopic dermatitis (AD)-like inflammation in adults through priming of group 2 innate lymphoid cells (ILC2s). Early postnatal skin is dynamically populated by discrete subset of primed ILC2s driven by microbiota-dependent induction of thymic stromal lymphopoietin (TSLP) in keratinocytes. Specifically, the indole-3-aldehyde-producing tryptophan metabolic pathway, shared across Staphylococcus species, is involved in TSLP-mediated ILC2 priming. Furthermore, we demonstrate a critical contribution of the early postnatal S. lentus-TSLP-ILC2 priming axis in facilitating AD-like inflammation that is not replicated by later microbial exposure. Thus, our findings highlight the fundamental role of time-dependent neonatal microbial-skin crosstalk in shaping the threshold of innate type 2 immunity co-opted in adulthood.


Assuntos
Dermatite Atópica , Linfopoietina do Estroma do Timo , Humanos , Adulto , Recém-Nascido , Imunidade Inata , Linfócitos , Citocinas/metabolismo , Pele/metabolismo , Inflamação
9.
Reprod Fertil Dev ; 25(4): 691-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22950940

RESUMO

MicroRNA is a short RNA molecule expressed in eukaryotic cells that is involved in multiple processes, including translational repression, target degradation and gene silencing. However, its specific role(s) in these processes remains largely unknown, especially in terms of germ cell development. The present study identified a microRNA, namely miR-335-5p, that is involved in mouse oocyte meiosis. MiR-335-5p was highly expressed in oocytes, but levels decreased markedly shortly after fertilisation. Microinjection of miR-335-5p or its inhibitor into oocytes resulted in a higher proportion of 2-cell-like MII oocytes and oocytes at the germinal vesicle breakdown and/or MI stage, indicating failure of asymmetric oocyte division. This may be due to regulation of actin because perturbation of miR-335-5p resulted in reduced expression of actin nucleator Daam1, a member of the Formin family. Moreover, injection of miR-335-5p or its inhibitor resulted in aberrant spindle morphology, namely an elongated spindle and multiple poles spindle. After injection of oocytes, levels of phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2) decreased, suggesting that miR-335-5p may regulate spindle formation via the mitogen-activated protein kinase pathway. Overexpression and inhibition of miR-335-5p had no effect on embryo development. Together, the results of the present study indicate that miR-335-5p is a novel regulator expressed in oocytes that is involved in cytoskeleton dynamics.


Assuntos
Citoesqueleto/metabolismo , MicroRNAs/metabolismo , Proteínas dos Microfilamentos/metabolismo , Oócitos/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Western Blotting , Primers do DNA/genética , Imunofluorescência , Modelos Lineares , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Meiose , Camundongos , MicroRNAs/farmacologia , Microscopia Confocal , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fuso Acromático/efeitos dos fármacos
10.
Aging (Albany NY) ; 15(14): 6690-6709, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37494662

RESUMO

Age-associated DNA-methylation drift (AMD) manifests itself in two ways in mammals: global decrease (hypomethylation) and local increase of DNA methylation (hypermethylation). To comprehend the principle behind this bidirectional AMD, we studied methylation states of spatially clustered CpG dinucleotides in mouse splenic DNA using reduced-representation-bisulfite-sequencing (RRBS). The mean methylation levels of whole CpGs declined with age. Promoter-resident CpGs, generally weakly methylated (<5%) in young mice, became hypermethylated in old mice, whereas CpGs in gene-body and intergenic regions, initially moderately (~33%) and extensively (>80%) methylated, respectively, were hypomethylated in the old. Chromosome-wise analysis of methylation revealed that inter-individual heterogeneities increase with age. The density of nearby CpGs was used to classify individual CpGs, which found hypermethylation in CpG-rich regions and hypomethylation in CpG-poor regions. When genomic regions were grouped by methylation level, high-methylation regions tended to become hypomethylated whereas low-methylation regions tended to become hypermethylated, regardless of genomic structure/function. Data analysis revealed that while methylation level and CpG density were interdependent, methylation level was a better predictor of the AMD pattern representing a shift toward the mean. Further analysis of gene-expression data showed a decrease in the expression of highly-expressed genes and an increase in the expression of lowly-expressed genes with age. This shift towards the mean in gene-expression changes was correlated with that of methylation changes, indicating a potential link between the two age-associated changes. Our findings suggest that age-associated hyper- and hypomethylation events are stochastic and attributed to malfunctioning intrinsic mechanisms for methylation maintenance in low- and high-methylation regions, respectively.


Assuntos
Metilação de DNA , Baço , Camundongos , Animais , Ilhas de CpG/genética , Envelhecimento/genética , DNA , Mamíferos/genética , Expressão Gênica , Epigênese Genética
11.
J Biol Chem ; 286(47): 41115-24, 2011 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-21921037

RESUMO

Setdb1/Eset is a histone H3 lysine 9 (H3K9)-specific methyltransferase that associates with various transcription factors to regulate gene expression via chromatin remodeling. Here, we report that Setdb1 associates with promyelocytic leukemia (Pml) protein from the early stage of mouse development and is a constitutive member of promyelocytic leukemia (PML)-nuclear bodies (PML-NBs) that have been linked to many cellular processes such as apoptosis, DNA damage responses, and transcriptional regulation. Arsenic treatment, which induces Pml degradation, caused Setdb1 signals to disappear. Setdb1 knockdown resulted in dismantlement of PML-NBs. Immunoprecipitation results demonstrated physical interactions between Setdb1 and Pml. Chromatin immunoprecipitation revealed that, within the frame of PML-NBs, Setdb1 binds the promoter of Id2 and suppresses its expression through installing H3K9 methylation. Our findings suggest that Setdb1 performs dual, but inseparable, functions at PML-NBs to maintain the structural integrity of PML-NBs and to control PML-NB-associated genes transcriptionally.


Assuntos
Núcleo Celular/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Metiltransferases/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Arsênio/farmacologia , Núcleo Celular/efeitos dos fármacos , Implantação do Embrião , Feminino , Técnicas de Silenciamento de Genes , Inativação Gênica , Histona-Lisina N-Metiltransferase , Proteína 2 Inibidora de Diferenciação/deficiência , Proteína 2 Inibidora de Diferenciação/genética , Proteína 2 Inibidora de Diferenciação/metabolismo , Masculino , Camundongos , Células NIH 3T3 , Gravidez , Proteína da Leucemia Promielocítica , Proteínas Metiltransferases/deficiência , Proteínas Metiltransferases/genética
12.
J Biol Chem ; 286(6): 4461-70, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21148561

RESUMO

With the objective of returning cells to their undifferentiated state through alteration of epigenetic states, small molecules have been used that specifically inhibit proteins involved in sustaining the epigenetic system. However, this chemical-based approach can cause chaotic epigenomic states due to random actions of the inhibitors. We investigated whether JHDM3A/JMJD2A, a trimethylated histone H3-lysine 9 (H3K9me3)-specific demethylase, could function as an effector molecule to selectively demethylate target chromatin, with the aid of a guide protein to serve as a delivery vehicle. JHDM3A, which normally locates in euchromatin, spread out to heterochromatin when it was fused to heterochromatin protein-1α (HP1α) or HP1ß; in these cells, demethylation efficiency was also markedly increased. Two truncated modules, JHDM3A(GFP)(406) and JHDM3A(GFP)(701), had contrasting modes and efficiencies of H3K9me3 demethylation; JHDM3A(GFP)(406) showed a very uniform rate (∼80%) of demethylation, whereas JHDM3A(GFP)(701) had a broad methylation range of 4-80%. The methylation values were highly dependent on the presence of the guide proteins OCT4, CTCF, and HP1. Chromatin immunoprecipitation detected reduced H3K9me3 levels at OCT4 regulatory loci in the cells expressing OCT4-tagged JHDM3A(GFP)(701). Derepression of the Sox2 gene was observed in JHDM3A(GFP)(701)OCT4-expressing cells, but not in cells that expressed the JHDM3A(GFP)(701) module alone. JHDM3A(GFP)(701)-assisted OCT4 more efficiently turned on stem cell-related microRNAs than GFP-OCT4 itself. These results suggest that JHDM3A(GFP)(701) is a suitable catalytic module that can be targeted, under the control of a guide protein, to specific loci where the chromatin H3K9me3 status and the milieu of gene expression are to be modified.


Assuntos
Eucromatina/metabolismo , Regulação da Expressão Gênica/fisiologia , Heterocromatina/metabolismo , Histonas/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Sequência de Aminoácidos/genética , Animais , Fator de Ligação a CCCTC , Homólogo 5 da Proteína Cromobox , Eucromatina/genética , Feminino , Loci Gênicos/fisiologia , Células HEK293 , Heterocromatina/genética , Histonas/genética , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Metilação , Camundongos , Células NIH 3T3 , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Deleção de Sequência
13.
Front Cell Dev Biol ; 10: 1069765, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36605719

RESUMO

SETDB1 is a histone H3-lysine 9-specific methyltransferase that fulfills epigenetic functions inside the nucleus; however, when overexpressed, SETDB1 majorily localizes in the cytoplasm. SETDB1 has a single nuclear-localization-signal (NLS) motif and two successive nuclear-export-signal (NES1 and NES2) motifs in the N-terminus, suggesting that SETDB1 localization is the consequence of a balance between the two antithetic motifs. Here, we performed a series of motif deletions to characterize their effects on the cellular movement of SETDB1. Given the cytoplasmic localization of GFP-SETDB1 in the whole form, without the NES motifs, GFP-SETDB1 was not nuclear, and 3xNLS addition plus NES removal held the majority of GFP-SETDB1 within the nucleus. The results indicated that the cytoplasmic localization of GFP-SETDB1 is the combined result of weak NLS and robust NESs. In ATF7IP-overexpressing cells, GFP-SETDB1 entered the nucleus only in the presence of the NES1 motif; neither the NES2 nor NLS motif was necessary. Since subcellular fractionation results showed that ATF7IP was nuclear-only, an intermediary protein may interact specifically with the NES1 motif after stimulation by ATF7IP. When GFP-SETDB1 had either NES1 or NES2, it was precipitated (in immunoprecipitation) and colocalized (in immunofluorescence) with ATF7IP, indicating that GFP-SETDB1 interacts with ATF7IP through the NES motifs in the nucleus. The regulated nuclear entry of SETDB1 is assumed to set a tight restriction on its abundance within the nucleus, thereby ensuring balanced nuclear SETDB1 levels.

14.
Aging (Albany NY) ; 14(1): 143-160, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35017317

RESUMO

With a graying population and increasing longevity, it is essential to identify life transition in later years and discern heterogeneity among older people. Subclassifying the elderly population to inspect the subdivisions for pathophysiological differences is particularly important for the investigation of age-related illnesses. For this purpose, using 24- and 28-month-old mice to represent the "young-old" and "old-old", respectively, we compared their skeletal muscle transcriptomes and found each in a distinct stage: early/gradual (E-aging) and late/accelerated aging phase (L-aging). Principal component analysis showed that the old-old transcriptomes were largely disengaged from the forward transcriptomic trajectory generated in the younger-aged group, indicating a substantial change in gene expression profiles during L-aging. By calculating the transcriptomic distance, it was found that the 28-month group was closer to the two-month group than to the 24-month group. The divergence rate per month for the transcriptomes was the highest in L-aging, twice as fast as the rate in E-aging. Indeed, many of the L-aging genes were significantly altered in transcription, although the changes did not seem random but rather coordinated in a variety of functional gene sets. Of 2,707 genes transcriptionally altered during E-aging, two-thirds were also significantly changed during L-aging, to either downturning or upturning way. The downturn genes were related to mitochondrial function and translational gene sets, while the upturn genes were linked to inflammation-associated gene sets. Our results provide a transcriptomic muscle signature that distinguishes old-old mice from young-old mice. This can help to methodically examine muscle disorders in the elderly.


Assuntos
Envelhecimento/fisiologia , Regulação da Expressão Gênica/fisiologia , Músculo Esquelético/fisiologia , Transcriptoma , Animais , Camundongos
15.
Sci Adv ; 8(43): eabk1239, 2022 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-36306352

RESUMO

Amphibians and fish show considerable regeneration potential via dedifferentiation of somatic cells into blastemal cells. In terms of dedifferentiation, in vitro cellular reprogramming has been proposed to share common processes with in vivo tissue regeneration, although the details are elusive. Here, we identified the cytoskeletal linker protein desmoplakin (Dsp) as a common factor mediating both reprogramming and regeneration. Our analysis revealed that Dsp expression is elevated in distinct intermediate cells during in vitro reprogramming. Knockdown of Dsp impedes in vitro reprogramming into induced pluripotent stem cells and induced neural stem/progenitor cells as well as in vivo regeneration of zebrafish fins. Notably, reduced Dsp expression impairs formation of the intermediate cells during cellular reprogramming and tissue regeneration. These findings suggest that there is a Dsp-mediated evolutionary link between cellular reprogramming in mammals and tissue regeneration in lower vertebrates and that the intermediate cells may provide alternative approaches for mammalian regenerative therapy.


Assuntos
Células-Tronco Pluripotentes Induzidas , Células-Tronco Neurais , Animais , Reprogramação Celular/genética , Desmoplaquinas/genética , Peixe-Zebra , Mamíferos
16.
Genes Cells ; 15(3): 181-92, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20070858

RESUMO

Post-translational modifications of histones play important roles in regulating chromatin dynamics and epigenetic inheritance during mitosis. The epigenetic significance and stability of histone H3-lysine 9 (H3K9) modifications have been well studied in interphase cells, whereas not as much in mitotic cells. Here, we inspected mitosis-coupled alterations in the global modifications of H3K9. Signals for H3K9 mono-, di-methylation and acetylation became invisible as cells entered mitosis in contrast to the pattern observed for H3-serine 10 phosphorylation (H3S10ph). Treatment with the aurora-B inhibitor ZM447439 or expression of the dominant negative mutant Aur-B(K106R) resulted in prometaphase chromosomes that lacked signals for H3S10ph but were positive for H3K9 modifications. Trimethylation was the sole K9 modification that remained consistently detectable throughout the cell cycle. This phenomenon was specific for H3K9-S10, as this pattern was not observed at H3K27-S28. Methylated H3K27 remained detectable throughout the cell cycle, despite phosphorylation of the adjacent H3S28. Contrastingly, our dot-blot experiment using synthetic peptides showed that phosphorylation of serine residue basically kept adjacent lysine from antibody access. Together, these results suggest that phosphorylation of serine residue occurs in a selective manner, being influenced by the types of modifications and the nature of neighboring lysine residues.


Assuntos
Histonas/metabolismo , Lisina/metabolismo , Serina/metabolismo , Animais , Aurora Quinase B/metabolismo , Benzamidas/farmacologia , Ciclo Celular/efeitos dos fármacos , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/metabolismo , Feminino , Masculino , Metilação , Camundongos , Mitose , Células NIH 3T3 , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia
17.
Curr Opin Cell Biol ; 15(3): 290-5, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12787770

RESUMO

Oocyte cytoplasmic factors can reprogramme the sperm genome during fertilisation or the somatic cell genome during cloning. Diverse reprogramming machinery acts sequentially and interdependently on the imported genome to drive it to totipotency, but their three-dimensional interactions in the cytoplasm remain unknown. Aberrant epigenetic phenomena in early cloned embryos indicate that parts of the somatic cell genome are unyielding to reprogramming forces, owing to their 'knotty' epigenetic features. This fastidious nature of the donor genome might prevent completion of epigenetic reprogramming. It might also help to explain the chronic developmental defects seen in many cloned embryos.


Assuntos
Blastocisto/fisiologia , Clonagem de Organismos , Metilação de DNA , Epigênese Genética/fisiologia , Animais
18.
Front Genet ; 11: 573515, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33343623

RESUMO

An increasing volume of evidence suggests that SETDB1 plays a role in the tumorigenesis of various cancers, classifying SETDB1 as an oncoprotein. However, owing to its numerous protein partners and their global-scale effects, the molecular mechanism underlying SETDB1-involved oncogenesis remains ambiguous. In this study, using public transcriptome data of lung adenocarcinoma (ADC) and squamous-cell carcinoma (SCC), we compared tumors with high-level SETDB1 (SH) and those with low-level SETDB1 (comparable with normal samples; SL). The results of principal component analysis revealed a transcriptomic distinction and divergence between the SH and SL samples in both ADCs and SCCs. The results of gene set enrichment analysis indicated that genes involved in the "epithelial-mesenchymal transition," "innate immune response," and "autoimmunity" collections were significantly depleted in SH tumors, whereas those involved in "RNA interference" collections were enriched. Chromatin-modifying genes were highly expressed in SH tumors, and the variance in their expression was incomparably high in SCC-SH, which suggested greater heterogeneity within SCC tumors. DNA methyltransferase genes were also overrepresented in SH samples, and most differentially methylated CpGs (SH/SL) were undermethylated in a highly biased manner in ADCs. We identified interesting molecular signatures associated with the possible roles of SETDB1 in lung cancer. We expect these SETDB1-associated molecular signatures to facilitate the development of biologically relevant targeted therapies for particular types of lung cancer.

19.
Proteomics ; 9(5): 1128-41, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19206105

RESUMO

The ability to generate neural lineages from human embryonic stem cells (hESCs) in a controlled manner would further investigation of human neurogenesis and development of potential cell therapeutic applications to treat neurological diseases; however, generating such neural stem cells (NSCs) remains a challenge. In an attempt to characterize the cellular mechanisms involved in hESC differentiation into neuroprogenitor cells, we performed 2-DE using protein extracts from hESC-derived embryoid bodies (EBs) and neuroectodermal spheres (NESs) bearing neuroprogenitors. Of 47 differentially expressed protein spots, 28 nonredundant spots were shown to be upregulated in the NESs; these protein spots included neurogenesis-related proteins (TAF1, SEPT2, NPH3, and CRABP), as expected. Interestingly, 6 of these 28 protein spots were cytoskeleton-associated proteins (CSAP) such as Fascin-1, Cofilin-1, and Stathmin-1. Western-blot analyses confirmed the increased levels of these proteins in the NESs. Furthermore, immunostaining analysis showed that both Fascin-1 and Stathmin-1 were preferentially expressed in the inner rims of neural rosettes, which are characteristic features of neuroprogenitors in culture. We also confirmed prominent expression of Fascin-1 in (sub-)ventricular zone in E15.5 mouse fetal brain. Our results suggest that, in addition to the induction of those genes involved in neural development, hESC differentiation into the NES is associated with a marked reorganization of the cellular cytoskeleton.


Assuntos
Proteínas do Citoesqueleto/análise , Proteínas do Citoesqueleto/genética , Células-Tronco Embrionárias/citologia , Placa Neural/citologia , Neurogênese , Diferenciação Celular , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Eletroforese em Gel Bidimensional , Células-Tronco Embrionárias/metabolismo , Humanos , Placa Neural/metabolismo , Proteoma/análise , Proteoma/genética , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Espectrometria de Massas em Tandem , Regulação para Cima
20.
BMC Neurosci ; 10: 97, 2009 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-19682396

RESUMO

BACKGROUND: Studies have provided important findings about the roles of Notch signaling in neural development. Unfortunately, however, most of these studies have investigated the neural stem cells (NSCs) of mice or other laboratory animals rather than humans, mainly owing to the difficulties associated with obtaining human brain samples. It prompted us to focus on neuroectodermal spheres (NESs) which are derived from human embryonic stem cell (hESC) and densely inhabited by NSCs. We here investigated the role of Notch signaling with the hESC-derived NESs. RESULTS: From hESCs, we derived NESs, the in-vitro version of brain-derived neurospheres. NES formation was confirmed by increased levels of various NSC marker genes and the emergence of rosette structures in which neuroprogenitors are known to reside. We found that Notch signaling, which maintains stem cell characteristics of in-vivo-derived neuroprogenitors, is active in these hESC-derived NESs, similar to their in-vivo counterpart. Expression levels of Notch signaling molecules such as NICD, DLLs, JAG1, HES1 and HES5 were increased in the NESs. Inhibition of the Notch signaling by a gamma-secretase inhibitor reduced rosette structures, expression levels of NSC marker genes and proliferation potential in the NESs, and, if combined with withdrawal of growth factors, triggered differentiation toward neurons. CONCLUSION: Our results indicate that the hESC-derived NESs, which share biochemical features with brain-derived neurospheres, maintain stem cell characteristics mainly through Notch signaling, which suggests that the hESC-derived NESs could be an in-vitro model for in-vivo neurogenesis.


Assuntos
Diferenciação Celular/fisiologia , Neurogênese/fisiologia , Receptor Notch1/metabolismo , Western Blotting , Linhagem da Célula/fisiologia , Células Cultivadas , Células-Tronco Embrionárias/citologia , Imunofluorescência , Humanos , Imuno-Histoquímica , Células-Tronco Multipotentes/citologia , Placa Neural/citologia , Neurônios/metabolismo , Neurônios/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA