Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 657: 100-107, 2023 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-37001284

RESUMO

The mouse placenta is composed of three different trophoblast layers that are occupied by particular trophoblast subtypes to maintain placental function and pregnancy. Accurate control of trophoblast differentiation is required for proper placental function; however, the molecular mechanisms underlying cell fate decisions in trophoblast stem cells remain poorly understood. Epidermal growth factor (EGF) signaling is involved in multiple biological processes including cell survival, proliferation, and differentiation. The effect of EGF on trophoblast function has been reported in various species; however, the role of EGF signaling in mouse trophoblast specification remains unclear. In this study, we aimed to elucidate the role of EGF signaling in mouse trophoblast differentiation using mouse trophoblast stem cells (mTSCs) in an in vitro culture system. EGF stimulation at the early stage of differentiation repressed mTSC differentiation into spongiotrophoblast cells (SpT). Gene deletion and inhibitor experiments showed that the effect of EGF exposure went through epidermal growth factor receptor (Egfr) activity in mTSCs. EGF stimuli induced acute downstream activation of MAPK/ERK, PI3K/AKT, and JNK pathways, and inhibition of the MAPK/ERK pathway, but not others, alleviated EGF-mediated repression of SpT differentiation. Moreover, expression of Mash2, a master regulator of SpT differentiation, was repressed by EGF stimulation, and MAPK/ERK inhibition counteracted this repression. The Mash2 overexpression recovered SpT marker expression, indicating that the decrease in Mash2 expression was due to abnormal SpT differentiation in EGF-treated mTSCs. Our findings suggest that the EGF-Egfr-MAPK/ERK-Mash2 axis is a core regulatory mechanism for the EGF-mediated repression of SpT differentiation.


Assuntos
Fator de Crescimento Epidérmico , Trofoblastos , Camundongos , Animais , Feminino , Gravidez , Trofoblastos/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Fator de Crescimento Epidérmico/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Placenta/metabolismo , Receptores ErbB/metabolismo , Diferenciação Celular/genética , Células-Tronco/metabolismo
2.
Plast Reconstr Surg ; 151(3): 569-579, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36821571

RESUMO

BACKGROUND: In recent years, structures including the superficial musculoaponeurotic system and retaining ligaments that support the facial soft tissue have been clarified. However, these structures are very difficult to observe in their entirety by the standard gross anatomical procedure (ie, dissection from superficial to deep layers). Furthermore, accurate descriptions of these structures are rare in both anatomical and plastic surgery textbooks. The aim of this study was to clarify the facial fibrous structures in a gross anatomical view. METHODS: The authors' novel method used soft facial tissue and bone. The tissue was fixed in gelatin and sectioned at a thickness of 5 to 10 mm. Each section was placed on a wooden board; the bone was then pinned, and the skin was pulled outward with sutures to hyperextend the soft tissue. Subsequently, the loose connective tissue was torn and fat tissue was removed under a surgical microscope. After the removal of fat tissue, the fibrous facial structures (eg, the superficial musculoaponeurotic system and retaining ligaments) could be observed clearly. RESULTS: The thickness of the sections allowed three-dimensional observation, such that a structure located deep within a section could be clearly observed. The expansion of soft tissue facilitated observation of the facial layer and fibrous structures, and the locations of nerves and vessels. Therefore, the facial layer structure was readily discerned. CONCLUSION: This method is likely to be very useful in the field of plastic surgery because it enabled intuitive identification of facial layers and their characteristics. CLINICAL RELEVANCE STATEMENT: The dissection method developed by the authors reveals the connected morphology of each tissue of the face, thus providing basic data for analyzing soft tissue changes due to aging and gravity. This will be useful for the development of anti-aging medicine.


Assuntos
Procedimentos de Cirurgia Plástica , Ritidoplastia , Sistema Musculoaponeurótico Superficial , Humanos , Face/cirurgia , Sistema Musculoaponeurótico Superficial/cirurgia , Tecido Adiposo/cirurgia , Envelhecimento , Ritidoplastia/métodos
3.
Biochem Biophys Res Commun ; 636(Pt 1): 205-212, 2022 12 25.
Artigo em Inglês | MEDLINE | ID: mdl-36335871

RESUMO

The placenta plays various roles in a healthy pregnancy, and abnormalities in the placenta result in adverse outcomes. Adequate differentiation of trophoblast subtypes is necessary for placental function, but the molecular mechanisms that determine trophoblast cell fate remain unclear. Here, we screened small molecular compound (SMC) libraries (1904 SMCs) to identify particular SMCs which regulate trophoblast differentiation in mouse trophoblast stem cells (mTSCs) to understand the molecular mechanisms underlying cell fate decision in trophoblast cells. The two-step screening revealed a novel effect of N-oleoyldopamine (OLDA), an endogenic vanilloid, to promote differentiation into parietal trophoblast giant cells (P-TGCs) and repress them into spongiotrophoblast cells in mTSCs. Analyses by gene deletion and inhibitor treatments indicated that transient receptor potential cation channel subfamily V member 3 (Trpv3), one of the candidates for targeting by OLDA, was involved in maintaining stem status and P-TGC differentiation in mTSCs. Finally, transcriptome analysis revealed that Fosl1, a key regulatory factor in differentiation into P-TGCs, was upregulated by OLDA treatment, suggesting that OLDA promoted the differentiation of mTSCs into P-TGCs via regulation of Fosl1 expression.


Assuntos
Placenta , Trofoblastos , Camundongos , Animais , Feminino , Gravidez , Trofoblastos/metabolismo , Placenta/metabolismo , Células Gigantes , Diferenciação Celular/genética , Células-Tronco
4.
J Biochem ; 170(1): 131-138, 2021 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-34314486

RESUMO

Understanding the molecular mechanism of neuronal differentiation is important to overcome the incurable diseases caused by nervous system damage. Neurite outgrowth is prerequisite for neuronal differentiation and regeneration, and cAMP response element-binding protein (CREB) is one of the major transcriptional factors positively regulating this process. Neuronal differentiation stimuli activate mammalian target of rapamycin (mTOR) complex 2 (mTORC2)/Akt signalling to phosphorylate CREB; however, the precise molecular mechanism of this event has not been fully understood. In this manuscript, we show that neuronal differentiation stimuli increased a protein level of protein phosphatase 6 (PP6), a member of type 2A Ser/Thr protein phosphatases. PP6 knockdown suppressed mTORC2/Akt/CREB signalling and results in failure of neurite outgrowth. SIN1 is a unique component of mTORC2 that enhances mTORC2 activity towards Akt when it is in dephosphorylated form. We found PP6 knockdown increased SIN1 phosphorylation. These data suggest that PP6 may positively regulate neurite outgrowth by dephosphorylating SIN1 to activate mTORC2/Akt/CREB signalling.


Assuntos
Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Animais , Células Cultivadas , Humanos , Camundongos , Crescimento Neuronal
5.
Exp Neurol ; 345: 113815, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34310943

RESUMO

Amyotrophic lateral sclerosis (ALS) is a fatal neuromuscular disease in which patients gradually become paralyzed due to loss of motor function. Many genetically inheritable mutations have been linked to ALS; however, the majority of ALS patients are considered sporadic. Therefore, there is a need for a common therapy that is effective for all ALS patients. Although there is evidence of the disease beginning in the periphery at the neuromuscular junction (NMJ), the specific processes involved in skeletal muscle and at the NMJ are still largely unknown. To study common disease mechanisms in ALS skeletal muscle, we performed RNA sequencing of skeletal myocytes differentiated from induced pluripotent stem cells (iPSCs) derived from familial ALS (with C9ORF72, SOD1, or TARDBP mutations) and sporadic ALS patients. Compared to healthy control lines, the myocytes from all ALS lines showed downregulation of four genes: BET1L, DCX, GPC3, and HNRNPK. We next measured the expression levels of these four genes in hind limb muscle samples from a rat model of familial ALS (SOD1G93A transgenic) and found that only the Bet1L gene, which encodes Bet1 Golgi Vesicular Membrane Trafficking Protein Like, was commonly downregulated. Bet1L protein appeared to be localized to the basal lamina of the NMJ, with decreased expression over time in SOD1G93A transgenic rats. Importantly, the expression levels began to decrease early in the disease process. Our results indicate that loss of Bet1L at the NMJ could be of interest for better understanding ALS disease progression.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Perfilação da Expressão Gênica/métodos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Junção Neuromuscular/metabolismo , Proteínas Qc-SNARE/deficiência , Adulto , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Animais , Diferenciação Celular/fisiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Junção Neuromuscular/patologia , Proteínas Qc-SNARE/genética , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Análise de Sequência de RNA/métodos
6.
Biochem Biophys Res Commun ; 561: 165-171, 2021 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-34023782

RESUMO

Linker histone H1 is mainly localized in the linker DNA region, between two nucleosome cores, and regulates chromatin structures linking gene expression. Mammalian oocytes contain the histone H1foo, a distinct member with low sequence similarity to other members in the H1 histone family. Although, from various previous studies, evidence related to H1foo function in chromatin structures is being accumulated, the distribution of H1foo at the target gene loci in a genome-wide manner and the molecular mechanism of H1foo-dependent chromatin architecture remain unclear. In this study, we aimed to identify the target loci and the physiological factor bound to H1foo at the loci. Chromatin immunoprecipitation sequencing analysis of H1foo-overexpressing mouse embryonic stem cells showed that H1foo is enriched around the transcriptional start sites of genes such as oocyte-specific genes and that the chromatin structures at these regions were relaxed. We demonstrated that H1foo was physiologically bound to the nuclear receptor estrogen-related receptor beta (Esrrb), and Esrrb was necessary for H1foo activity of chromatin decondensation at the target loci. The specific localization and interaction with Esrrb were validated in endogenous H1foo of oocytes. Overall, H1foo induces chromatin decondensation in a locus-specific manner and this function is achieved by interacting with Esrrb.


Assuntos
Núcleo Celular/metabolismo , Cromatina/metabolismo , Histonas/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Oócitos/metabolismo , Receptores de Estrogênio/metabolismo , Animais , Células Cultivadas , Cromatina/química , Sequenciamento de Cromatina por Imunoprecipitação/métodos , Histonas/genética , Camundongos , Receptores de Estrogênio/genética
7.
Biochem Biophys Res Commun ; 528(4): 685-690, 2020 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-32513538

RESUMO

Linker histone H1 is mainly localized in the linker DNA region, between two nucleosome cores, and regulates chromatin structures linking gene expression. There are 11 variants in histone H1, and each variant has unique functions. Our previous study demonstrates that one of the H1 variants, H1T is mainly localized in the nucleolus and targets the rDNA repeat region. Moreover, H1T condenses the chromatin structures on rDNA to repress pre-rRNA expression. Although H1T is partially localized in the nucleoplasm area, the functions of H1T in the non-repeat genic region are unclear. In this study, we aimed to identify the target loci and the role of H1T in the genic region. Chromatin immunoprecipitation sequencing analysis showed that H1T is localized around the transcriptional start site and the chromatin structures of the region were relaxed. H1T knockdown and overexpression experiments revealed that H1T induced chromatin de-condensation and was associated with the increased expression of target genes. Moreover, we observed H1T co-localization with transcriptional factor SPZ1 on the genic region. Collectively, H1T has opposing roles in the genic region and in rDNA repeats; H1T functions to facilitate chromatin relaxation linked gene activation.


Assuntos
Cromatina/metabolismo , Histonas/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Cromatina/genética , DNA Ribossômico/genética , DNA Ribossômico/metabolismo , Regulação da Expressão Gênica , Histonas/análise , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Espermatogênese , Testículo/metabolismo
8.
Diabetologia ; 63(9): 1836-1846, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32561946

RESUMO

AIMS/HYPOTHESIS: Obesity caused by overeating plays a pivotal role in the development of type 2 diabetes. However, it remains poorly understood how individual meal size differences are determined before the development of obesity. Here, we investigated the underlying mechanisms in determining spontaneous food intake in newly established Oikawa-Nagao Diabetes-Prone (ON-DP) and Diabetes-Resistant (ON-DR) mice. METHODS: Food intake and metabolic phenotypes of ON-DP and ON-DR mice under high-fat-diet feeding were compared from 5 weeks to 10 weeks of age. Differences in leptin status at 5 weeks of age were assessed between the two mouse lines. Adipose tissue explant culture was also performed to evaluate leptin production capacity in vitro. RESULTS: ON-DP mice showed spontaneous overfeeding compared with ON-DR mice. Excessive body weight gain and fat accumulation in ON-DP mice were completely suppressed to the levels seen in ON-DR mice by pair-feeding with ON-DR mice. Deterioration of glucose tolerance in ON-DP mice was also ameliorated under the pair-feeding conditions. While no differences were seen in body weight and adipose tissue mass when comparing the two mouse lines at 5 weeks of age, the ON-DP mice had lower plasma leptin concentrations and adipose tissue leptin gene expression levels. In accordance with peripheral leptin status, ON-DP mice displayed lower anorexigenic leptin signalling in the hypothalamic arcuate nucleus when compared with ON-DR mice without apparent leptin resistance. Explant culture studies revealed that ON-DP mice had lower leptin production capacity in adipose tissue. ON-DP mice also displayed higher DNA methylation levels in the leptin gene promoter region of adipocytes when compared with ON-DR mice. CONCLUSIONS/INTERPRETATION: The results suggest that heritable lower leptin production capacity plays a critical role in overfeeding-induced obesity and subsequent deterioration of glucose tolerance in ON-DP mice. Leptin production capacity in adipocytes, especially before the development of obesity, may have diagnostic potential for predicting individual risk of obesity caused by overeating and future onset of type 2 diabetes. Graphical abstract.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Ingestão de Alimentos/fisiologia , Comportamento Alimentar/fisiologia , Leptina/metabolismo , Obesidade/metabolismo , Adipócitos/metabolismo , Adiponectina/genética , Animais , Proteínas Estimuladoras de Ligação a CCAAT/genética , Ritmo Circadiano , Diabetes Mellitus Tipo 2/etiologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Proteínas de Ligação a Ácido Graxo/genética , Teste de Tolerância a Glucose , Hiperfagia/metabolismo , Hiperfagia/fisiopatologia , Leptina/genética , Locomoção , Camundongos , Obesidade/complicações , PPAR gama/genética
9.
Sci Rep ; 9(1): 19768, 2019 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-31875008

RESUMO

Epigenetic mechanisms can establish and maintain mitotically stable patterns of gene expression while retaining the DNA sequence. These mechanisms can be affected by environmental factors such as nutrients. The importance of intracellular dosages of nutrient metabolites such as acetyl coenzyme A and S-adenosylmethionine, which are utilized as donors for post-translational modifications, is well-known in epigenetic regulation; however, the significance of indirect metabolites in epigenetic regulation is not clear. In this study, we screened for metabolites that function as epigenetic modulators. Because the expression of genes related to hypothalamic function is reportedly affected by nutritional conditions, we used a neural cell culture system and evaluated hypothalamic-linked loci. We supplemented the culture medium with 129 metabolites separately during induction of human-iPS-derived neural cells and used high-throughput ChIP-qPCR to determine the epigenetic status at 37 hypothalamus-linked loci. We found three metabolites (kynurenine, 3-OH-kynurenine, and anthranilate) from tryptophan pathways that increased H3K4 trimethylation and H2AS40 O-GlcNAcylation, resulting in upregulated gene expression at most loci, except those encoding pan-neural markers. Dietary supplementation of these three metabolites and the resulting epigenetic modification were important for stability in gene expression. In conclusion, our findings provide a better understanding of how nutrients play a role in epigenetic mechanisms.


Assuntos
Epigênese Genética , Loci Gênicos , Histonas/metabolismo , Hipotálamo/metabolismo , Cinurenina/análogos & derivados , ortoaminobenzoatos/metabolismo , Glicosilação , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Cinurenina/metabolismo , Metilação
10.
Dis Model Mech ; 12(8)2019 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-31439573

RESUMO

Amyotrophic lateral sclerosis (ALS) is a late-onset neuromuscular disease with no cure and limited treatment options. Patients experience a gradual paralysis leading to death from respiratory complications on average only 2-5 years after diagnosis. There is increasing evidence that skeletal muscle is affected early in the disease process, yet the pathological processes occurring in the skeletal muscle of ALS patients are still mostly unknown. Specifically, the most common genetic cause of ALS, a hexanucleotide repeat expansion in the C9ORF72 gene, has yet to be fully characterized in the context of skeletal muscle. In this study, we used the protocol previously developed in our lab to differentiate skeletal myocytes from induced pluripotent stem cells (iPSCs) of C9ORF72 ALS (C9-ALS) patients in order to create an in vitro disease model of C9-ALS skeletal muscle pathology. Of the three C9ORF72 mutation hallmarks, we did not see any evidence of haploinsufficiency, but we did detect RNA foci and dipeptide repeat (DPR) proteins. Additional abnormalities included changes in the expression of mitochondrial genes and a susceptibility to oxidative stress, indicating that mitochondrial dysfunction may be a critical feature of C9-ALS skeletal muscle pathology. Finally, the C9-ALS myocytes had increased expression and aggregation of TDP-43. Together, these data show that skeletal muscle cells experience pathological changes due to the C9ORF72 mutation. Our in vitro model could facilitate further study of cellular and molecular pathology in ALS skeletal muscle in order to discover new therapeutic targets against this devastating disease.This article has an associated First Person interview with the first author of the paper.


Assuntos
Esclerose Lateral Amiotrófica/patologia , Proteína C9orf72/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Diferenciação Celular , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Haploinsuficiência/genética , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Mitocôndrias/metabolismo , Fibras Musculares Esqueléticas/ultraestrutura , Estresse Oxidativo , Agregados Proteicos , RNA/metabolismo
11.
J Reprod Dev ; 65(1): 19-27, 2019 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-30318498

RESUMO

Mouse trophoblast stem cells (TSCs) have been established and maintained using hyperglycemic conditions (11 mM glucose) for no apparent good reason. Because glucose metabolites are used as resources for cellular energy production, biosynthesis, and epigenetic modifications, differences in extracellular glucose levels may widely affect cellular function. Since the hyperglycemic culture conditions used for TSC culture have not been fully validated, the effect of extracellular glucose levels on the properties of TSCs remains unclear. To address this issue, we investigated the gene expression of stemness-related transcription factors in TSCs cultured in the undifferentiated state under various glucose concentrations. We also examined the expression of trophoblast subtype markers during differentiation, after returning the glucose concentration to the conventional culture concentration (11 mM). As a result, it appeared that the extracellular glucose conditions in the stem state not only affected the gene expression of stemness-related transcription factors before differentiation but also affected the expression of marker genes after differentiation, with some line-to-line variation. In the TS4 cell line, which showed the largest glucose concentration-dependent fluctuations in gene expression among all the lines examined, low glucose (1 mM glucose, LG) augmented H3K27me3 levels. An Ezh2 inhibitor prevented these LG-induced changes in gene expression, suggesting the possible involvement of H3K27me3 in the changes in gene expression seen in LG. These results collectively indicate that the response of the TSCs to the change in the extracellular glucose concentration is cell line-dependent and a part of which may be epigenetically memorized.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Meios de Cultura , Expressão Gênica/efeitos dos fármacos , Glucose/administração & dosagem , Células-Tronco/citologia , Trofoblastos/citologia , Animais , Linhagem Celular , Células Cultivadas , Epigênese Genética/efeitos dos fármacos , Marcadores Genéticos/efeitos dos fármacos , Histonas/metabolismo , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Células-Tronco/efeitos dos fármacos
12.
Sci Rep ; 8(1): 5811, 2018 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-29643413

RESUMO

Trophoblast giant cells (TGCs) are one of the cell types that form the placenta and play multiple essential roles in maintaining pregnancy in rodents. TGCs have large, polyploid nuclei resulting from endoreduplication. While previous studies have shown distinct gene expression profiles of TGCs, their chromatin structure remains largely unknown. An appropriate combination of canonical and non-canonical histones, also known as histone variants, allows each cell to exert its cell type-specific functions. Here, we aimed to reveal the dynamics of histone usage and chromatin structure during the differentiation of trophoblast stem cells (TSCs) into TGCs. Although the expression of most genes encoding canonical histones was downregulated, the expression of a few genes encoding histone variants such as H2AX, H2AZ, and H3.3 was maintained at a relatively high level in TGCs. Both the micrococcal nuclease digestion assay and nucleosome stability assay using a microfluidic device indicated that chromatin became increasingly loose as TSCs differentiated. Combinatorial experiments involving H3.3-knockdown and -overexpression demonstrated that variant H3.3 resulted in the formation of loose nucleosomes in TGCs. In conclusion, our study revealed that TGCs possessed loose nucleosomes owing to alterations in their histone composition during differentiation.


Assuntos
Cromatina/metabolismo , Células Gigantes/química , Histonas/análise , Nucleossomos/química , Poliploidia , Trofoblastos/química , Animais , Diferenciação Celular , Linhagem Celular , Expressão Gênica , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Histonas/genética , Camundongos
13.
Epigenetics Chromatin ; 10(1): 51, 2017 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-29084613

RESUMO

BACKGROUND: We have previously reported a novel O-GlcNAc modification at serine 40 (S40) of H2A (H2AS40Gc). S40-type H2A isoforms susceptible to O-GlcNAcylation are evolutionarily new and restricted to the viviparous animals; however, the biological function of H2AS40Gc is largely unknown. H2A isoforms are consisted of S40 and alanine 40 (A40) type and this residue on H2A is located in the L1 of the globular domain, which is also known as a variable portion that distinguishes between the canonical and non-canonical H2A variants. In this study, by considering the similarity between the S40-type H2A and histone H2A variants, we explored the function of H2AS40Gc in mouse embryonic stem cells (mESCs). RESULTS: We found several similarities between the S40-type H2A isoforms and histone H2A variants such H2AZ and H2AX. mRNA of S40-type H2A isoforms (H2A1 N and H2A3) had a poly(A) tail and was produced throughout the cell cycle in contrast to that of A40-type. Importantly, H2AS40Gc level increased owing to chemical-induced DNA damage, similar to phosphorylated H2AX (γH2AX) and acetylated H2AZ (AcH2AZ). H2AS40Gc was accumulated at the restricted area (± 1.5 kb) of DNA damage sites induced by CRISPR/CAS9 system in contrast to accumulation of γH2AX, which was widely scattered. Overexpression of the wild-type (WT) H2A3, but not the S40 to A40 mutation (S40A-mutant), protected the mESC genome against chemical-induced DNA damage. Furthermore, 3 h after the DNA damage treatment, the genome was almost recovered in WT mESCs, whereas the damage advanced further in the S40A-mutant mESCs, suggesting functions of H2AS40Gc in the DNA repair mechanism. Furthermore, the S40A mutant prevented the accumulation of the DNA repair apparatus such as DNA-PKcs and Rad51 at the damage site. Co-immunoprecipitation experiment in WT and S40A-mutant mESCs revealed that H2AS40Gc physiologically bound to AcH2AZ at the initial phase upon DNA damage, followed by binding with γH2AX during the DNA damage repair process. CONCLUSIONS: These data suggest that H2AS40Gc functions to maintain genome integrity through the DNA repair mechanism in association with AcH2AZ and γH2AX.


Assuntos
Instabilidade Genômica , Histonas/metabolismo , Processamento de Proteína Pós-Traducional , Acetilação , Animais , Linhagem Celular , Reparo do DNA , Células-Tronco Embrionárias/metabolismo , Histonas/genética , Camundongos , Mutação , Fosforilação , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
14.
Epigenetics ; 12(9): 764-778, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28762874

RESUMO

Orexin neurons regulate critical brain activities for controlling sleep, eating, emotions, and metabolism, and impaired orexin neuron function results in several neurologic disorders. Therefore, restoring normal orexin function and understanding the mechanisms of loss or impairment of orexin neurons represent important goals. As a step toward that end, we generated human orexin neurons from induced pluripotent stem cells (hiPSCs) by treatment with N-acetyl-d-mannosamine (ManNAc) and its derivatives. The generation of orexin neurons was associated with DNA hypomethylation, histone H3/H4 hyperacetylation, and hypo-O-GlcNAcylation on the HCRT gene locus, and, thereby, the treatment of inhibitors of SIRT1 and OGT were effective at inducing orexin neurons from hiPSCs. The prolonged exposure of orexin neurons to high glucose in culture caused irreversible silencing of the HCRT gene, which was characterized by H3/H4 hypoacetylation and hyper-O-GlcNAcylation. The DNA hypomethylation status, once established in orexin neurogenesis, was maintained in the HCRT-silenced orexin neurons, indicating that histone modifications, but not DNA methylation, were responsible for the HCRT silencing. Thus, the epigenetic status of the HCRT gene is unique to the hyperglycemia-induced silencing. Intriguingly, treatment of ManNAc and its derivatives reactivated HCRT gene expression, while inhibitors SIRT1 and the OGT did not. The present study revealed that the HCRT gene was silenced by the hyperglycemia condition, and ManNAc and its derivatives were useful for restoring the orexin neurons.


Assuntos
Hexosaminas/farmacologia , Hiperglicemia/genética , Orexinas/genética , Acetilação , Metilação de DNA , Epigênese Genética , Inativação Gênica/efeitos dos fármacos , Histonas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Orexinas/metabolismo
15.
Sci Rep ; 6: 31785, 2016 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-27615797

RESUMO

We report here newly discovered O-linked-N-acetylglucosamine (O-GlcNAc) modification of histone H2A at Ser(40) (H2AS40Gc). The mouse genome contains 18 H2A isoforms, of which 13 have Ser(40) and the other five have Ala(40). The combination of production of monoclonal antibody and mass spectrometric analyses with reverse-phase (RP)-high performance liquid chromatography (HPLC) fractionation indicated that the O-GlcNAcylation is specific to the Ser(40) isoforms. The H2AS40Gc site is in the L1 loop structure where two H2A molecules interact in the nucleosome. Targets of H2AS40Gc are distributed genome-wide and are dramatically changed during the process of differentiation in mouse trophoblast stem cells. In addition to the mouse, H2AS40Gc was also detected in humans, macaques and cows, whereas non-mammalian species possessing only the Ala(40) isoforms, such as silkworms, zebrafish and Xenopus showed no signal. Genome database surveys revealed that Ser(40) isoforms of H2A emerged in Marsupialia and persisted thereafter in mammals. We propose that the emergence of H2A Ser(40) and its O-GlcNAcylation linked a genetic event to genome-wide epigenetic events that correlate with the evolution of placental animals.


Assuntos
Acetilglucosamina/química , Evolução Biológica , Histonas/química , Animais , Anticorpos Monoclonais/química , Sítios de Ligação , Bombyx , Bovinos , Cromatografia Líquida de Alta Pressão , Células-Tronco Embrionárias/citologia , Células HeLa , Humanos , Macaca , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Filogenia , Isoformas de Proteínas/química , Proteínas Recombinantes/química , Serina/química , Especificidade da Espécie , Trofoblastos/metabolismo , Xenopus , Peixe-Zebra
16.
Epigenetics ; 11(4): 288-302, 2016 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-27018843

RESUMO

H1T is a linker histone H1 variant that is highly expressed at the primary spermatocyte stage through to the early spermatid stage of spermatogenesis. While the functions of the somatic types of H1 have been extensively investigated, the intracellular role of H1T is unclear. H1 variants specifically expressed in germ cells show low amino acid sequence homology to somatic H1s, which suggests that the functions or target loci of germ cell-specific H1T differ from those of somatic H1s. Here, we describe the target loci and function of H1T. H1T was expressed not only in the testis but also in tumor cell lines, mouse embryonic stem cells (mESCs), and some normal somatic cells. To elucidate the intracellular localization and target loci of H1T, fluorescent immunostaining and ChIP-seq were performed in tumor cells and mESCs. We found that H1T accumulated in nucleoli and predominantly targeted rDNA repeats, which differ from somatic H1 targets. Furthermore, by nuclease sensitivity assay and RT-qPCR, we showed that H1T repressed rDNA transcription by condensing chromatin structure. Imaging analysis indicated that H1T expression affected nucleolar formation. We concluded that H1T plays a role in rDNA transcription, by distinctively targeting rDNA repeats.


Assuntos
DNA Ribossômico/genética , Histonas/genética , Espermatogênese/genética , Transcrição Gênica , Sequência de Aminoácidos/genética , Animais , Nucléolo Celular/genética , Cromatina/genética , DNA Ribossômico/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/metabolismo , Histonas/biossíntese , Masculino , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Homologia de Sequência de Aminoácidos , Espermátides/crescimento & desenvolvimento , Espermátides/metabolismo , Espermatócitos/crescimento & desenvolvimento , Espermatócitos/metabolismo , Testículo/crescimento & desenvolvimento , Testículo/metabolismo
17.
Biomed Res Int ; 2015: 876047, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26339649

RESUMO

The regulation of transcription and genome stability by epigenetic systems are crucial for the proper development of mammalian embryos. Chemicals that disturb epigenetic systems are termed epimutagens. We previously performed chemical screening that focused on heterochromatin formation and DNA methylation status in mouse embryonic stem cells and identified five epimutagens: diethyl phosphate (DEP), mercury (Hg), cotinine, selenium (Se), and octachlorodipropyl ether (S-421). Here, we used human induced pluripotent stem cells (hiPSCs) to confirm the effects of 20 chemicals, including the five epimutagens, detected at low concentrations in maternal peripheral and cord blood samples. Of note, these individual chemicals did not exhibit epimutagenic activity in hiPSCs. However, because the fetal environment contains various chemicals, we evaluated the effects of combined exposure to chemicals (DEP, Hg, cotinine, Se, and S-421) on hiPSCs. The combined exposure caused a decrease in the number of heterochromatin signals and aberrant DNA methylation status at multiple gene loci in hiPSCs. The combined exposure also affected embryoid body formation and neural differentiation from hiPSCs. Therefore, DEP, Hg, cotinine, Se, and S-421 were defined as an "epimutagen combination" that is effective at low concentrations as detected in maternal peripheral and cord blood.


Assuntos
Metilação de DNA/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Mutagênicos/toxicidade , Animais , Diferenciação Celular/efeitos dos fármacos , Cotinina/toxicidade , Desenvolvimento Embrionário/efeitos dos fármacos , Epigênese Genética/genética , Éteres/toxicidade , Feminino , Sangue Fetal/efeitos dos fármacos , Heterocromatina/efeitos dos fármacos , Heterocromatina/genética , Humanos , Mercúrio/toxicidade , Camundongos , Organofosfatos/toxicidade , Selênio/toxicidade
18.
Curr Protoc Stem Cell Biol ; 32: 1E.4.1-1E.4.32, 2015 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-25640817

RESUMO

The isolation of stable trophoblast stem (TS) cell lines from early mouse embryos has provided a useful cell culture model to study trophoblast development. TS cells are derived from pre-implantation blastocysts or from the extraembryonic ectoderm of early post-implantation embryos. The derivation and maintenance of mouse TS cells is dependent upon continuous fibroblast growth factor (FGF) signaling. Gene expression analysis, differentiation in culture, and chimera formation show that TS cells accurately model the mouse trophoblast lineage. This unit describes how to derive, maintain, and manipulate TS cells, including DNA transfection and chimera formation.


Assuntos
Separação Celular/métodos , Células-Tronco/citologia , Trofoblastos/citologia , Animais , Blastocisto/citologia , Contagem de Células , Células Cultivadas , Quimera , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , DNA/metabolismo , Eletroporação , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Congelamento , Lipídeos , Camundongos , Mitomicina/farmacologia , Células-Tronco/efeitos dos fármacos , Transfecção
19.
Auton Neurosci ; 187: 56-62, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25443216

RESUMO

Aging is associated with a variety of physiological changes originating peripherally and centrally, including within the autonomic nervous system. Sleep-wake disturbances constitute reliable hallmarks of aging in several animal species and humans. Recent studies have been interested in N-acetylmannosamine (ManNAc) a potential therapeutic agent for improving quality of life, as well as preventing age-related cognitive decline. In this study, ManNAc (5.0 mg/ml) was administered in the drinking water of middle-aged male C57BL/6J mice (55 weeks old) for 7 days. Mice were housed under a 12:12 h light:dark cycle at 23-24 °C. We evaluated bio-behavioral activity using electrocardiogram, body temperature and locomotor activity recorded by an implanted telemetry transmitter. To estimate sleep-wake profile, surface electroencephalogram and electromyogram leads connected to a telemetry transmitter were also implanted in mice. Autonomic nervous activity was evaluated using power spectral analysis of heart rate variability. ManNAc-treated mice spent more time in a wakeful state and less time in slow wave sleep during the dark phase. Parasympathetic nervous activity was increased following ManNAc treatment, then the sympatho-vagal balance was shifted predominance of parasympathetic nervous system. Furthermore, improvement in sleep-wake pattern was associated with increased parasympathetic nervous activity. These results suggest that ManNAc treatment can improve bio-behavioral activity and sleep-wake quality in middle-aged mice. This may have implications for improving sleep patterns in elderly humans.


Assuntos
Envelhecimento , Sistema Nervoso Autônomo/efeitos dos fármacos , Hexosaminas/farmacologia , Sono/efeitos dos fármacos , Vigília/efeitos dos fármacos , Animais , Sistema Nervoso Autônomo/fisiologia , Pressão Sanguínea/efeitos dos fármacos , Temperatura Corporal/efeitos dos fármacos , Ondas Encefálicas/efeitos dos fármacos , Eletrocardiografia , Eletroencefalografia , Eletromiografia , Análise de Fourier , Frequência Cardíaca/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Estatística como Assunto , Telemetria
20.
Mech Dev ; 136: 143-54, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25528267

RESUMO

Nodal signaling plays critical roles during embryonic development. The Nodal gene is not expressed in adult tissues but is frequently activated in cancer cells, contributing to progression toward malignancy. Although several regulatory elements of the Nodal gene have been identified, the epigenetic mechanisms by which Nodal expression is regulated over the long term remain unclear. We found a region exhibiting dynamic changes in DNA methylation at approximately -3.0 kb to -0.4 kb upstream from the transcriptional start site (TSS) that we termed the epigenetic regulatory element (ERE). The ERE was unmethylated in mouse embryonic stem cells (mESCs) but became increasingly methylated in differentiated cells and tissues, concomitant with the downregulation of Nodal mRNA expression. In vitro reporter assays identified an Oct3/4 binding motif within the ERE, indicating that the ERE is responsible for the activation of Nodal in mESCs. Furthermore, the ERE was a target of differentiation-associated Polycomb silencing, and the chromatin condensed when mESCs differentiated to embryoid bodies (EBs). Pharmacological inhibition of PRC2 led to the reactivation of Nodal expression in EBs and mouse embryonic fibroblasts (MEFs). The ERE was also targeted by PRC2 in normal human cells. In NODAL-expressing human cancer cells, accumulation of EZH2 and trimethylation of H3K27 at the ERE were diminished. In conclusion, Nodal is epigenetically controlled through the ERE in the mouse embryo and human cells.


Assuntos
Epigênese Genética , Proteína Nodal/genética , Sequências Reguladoras de Ácido Nucleico/genética , Animais , Diferenciação Celular/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Genoma , Genoma Humano , Humanos , Camundongos , Proteína Nodal/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...