Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
Reprod Med Biol ; 16(4): 337-348, 2017 10.
Article in English | MEDLINE | ID: mdl-29259487

ABSTRACT

Purpose: Prenatal exposure to environmental chemicals is a growing concern, because such exposures have been shown to be associated with various diseases. The levels of chemicals and heavy metals in maternal blood, cord blood, maternal urine and amniotic fluid in Japanese pregnant women were investigated. Methods: A total of 145 women, including 14 fetal growth restriction cases, were included in the present study. The levels of phthalates (di[2-ethylhexyl]phthalate and mono[2-ethylhexyl]phthalate), perfluorinated compounds (perfluorooctane sulfonate, perfluorohexanoic acid, perfluorooctanoic acid, and perfluorononanoic acid), pesticides (dimethylphosphate, dimethylthiophosphate, diethylphosphate, diethylthiophosphate, 3-phenoxybenzoic acid, and octachlorodipropyl ether), bisphenol A, nicotine (nicotine, nornicotine, cotinine, norcotinine, and trans-3'-hydroxycotinine), polybrominated diphenyl ethers, and heavy metals were measured. The relationship between fetal growth and the levels of chemicals and heavy metals were investigated. Results: Phthalates, perfluorinated compounds, pesticides, polybrominated diphenyl ethers, and heavy metals were detected in high frequency, whereas nicotine and bisphenol A were almost negative. Phthalates, perfluorinated compounds, and several heavy metals were transferred to the fetus. High perfluorononanoic acid levels in the maternal blood and cord blood, and low perfluorooctanoic acid level in the cord blood were significantly and negatively associated with fetal growth. Conclusions: The present study showed that pregnant women in Japan and their fetuses are exposed to a variety of chemicals and heavy metals.

2.
Epigenetics Chromatin ; 10(1): 51, 2017 10 30.
Article in English | MEDLINE | ID: mdl-29084613

ABSTRACT

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.


Subject(s)
Genomic Instability , Histones/metabolism , Protein Processing, Post-Translational , Acetylation , Animals , Cell Line , DNA Repair , Embryonic Stem Cells/metabolism , Histones/genetics , Mice , Mutation , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism
3.
Epigenetics ; 12(9): 764-778, 2017 09.
Article in English | MEDLINE | ID: mdl-28762874

ABSTRACT

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.


Subject(s)
Hexosamines/pharmacology , Hyperglycemia/genetics , Orexins/genetics , Acetylation , DNA Methylation , Epigenesis, Genetic , Gene Silencing/drug effects , Histones/metabolism , Humans , Induced Pluripotent Stem Cells , Neurons/drug effects , Neurons/metabolism , Orexins/metabolism
4.
J Endocrinol ; 234(2): 217-232, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28576872

ABSTRACT

Mammals share common strategies for regulating reproduction, including a conserved hypothalamic-pituitary-gonadal axis; yet, individual species exhibit differences in reproductive performance. In this report, we describe the discovery of a species-restricted homeostatic control system programming testis growth and function. Prl3c1 is a member of the prolactin gene family and its protein product (PLP-J) was discovered as a uterine cytokine contributing to the establishment of pregnancy. We utilized mouse mutagenesis of Prl3c1 and revealed its involvement in the regulation of the male reproductive axis. The Prl3c1-null male reproductive phenotype was characterized by testiculomegaly and hyperandrogenism. The larger testes in the Prl3c1-null mice were associated with an expansion of the Leydig cell compartment. Prl3c1 locus is a template for two transcripts (Prl3c1-v1 and Prl3c1-v2) expressed in a tissue-specific pattern. Prl3c1-v1 is expressed in uterine decidua, while Prl3c1-v2 is expressed in Leydig cells of the testis. 5'RACE, chromatin immunoprecipitation and DNA methylation analyses were used to define cell-specific promoter usage and alternative transcript expression. We examined the Prl3c1 locus in five murid rodents and showed that the testicular transcript and encoded protein are the result of a recent retrotransposition event at the Mus musculus Prl3c1 locus. Prl3c1-v1 encodes PLP-J V1 and Prl3c1-v2 encodes PLP-J V2. Each protein exhibits distinct intracellular targeting and actions. PLP-J V2 possesses Leydig cell-static actions consistent with the Prl3c1-null testicular phenotype. Analysis of the biology of the Prl3c1 gene has provided insight into a previously unappreciated homeostatic setpoint control system programming testicular growth and function.


Subject(s)
Gene Expression Regulation/physiology , Glycoproteins/metabolism , Prolactin/metabolism , Testis/physiology , Animals , Female , Glycoproteins/genetics , Homeostasis , Intercellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Multigene Family , Prolactin/genetics , Protein Isoforms , Rats , Testis/growth & development
5.
Sci Rep ; 6: 31785, 2016 09 12.
Article in English | MEDLINE | ID: mdl-27615797

ABSTRACT

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.


Subject(s)
Acetylglucosamine/chemistry , Biological Evolution , Histones/chemistry , Animals , Antibodies, Monoclonal/chemistry , Binding Sites , Bombyx , Cattle , Chromatography, High Pressure Liquid , Embryonic Stem Cells/cytology , HeLa Cells , Humans , Macaca , Mass Spectrometry , Mice , Mice, Inbred C57BL , Phylogeny , Protein Isoforms/chemistry , Recombinant Proteins/chemistry , Serine/chemistry , Species Specificity , Trophoblasts/metabolism , Xenopus , Zebrafish
6.
Epigenetics ; 11(4): 288-302, 2016 04 02.
Article in English | MEDLINE | ID: mdl-27018843

ABSTRACT

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.


Subject(s)
DNA, Ribosomal/genetics , Histones/genetics , Spermatogenesis/genetics , Transcription, Genetic , Amino Acid Sequence/genetics , Animals , Cell Nucleolus/genetics , Chromatin/genetics , DNA, Ribosomal/biosynthesis , Gene Expression Regulation, Developmental , Germ Cells/metabolism , Histones/biosynthesis , Male , Mice , Mouse Embryonic Stem Cells/metabolism , Sequence Homology, Amino Acid , Spermatids/growth & development , Spermatids/metabolism , Spermatocytes/growth & development , Spermatocytes/metabolism , Testis/growth & development , Testis/metabolism
7.
Biomed Res Int ; 2015: 876047, 2015.
Article in English | MEDLINE | ID: mdl-26339649

ABSTRACT

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.


Subject(s)
DNA Methylation/drug effects , Epigenesis, Genetic/drug effects , Induced Pluripotent Stem Cells/drug effects , Mutagens/toxicity , Animals , Cell Differentiation/drug effects , Cotinine/toxicity , Embryonic Development/drug effects , Epigenesis, Genetic/genetics , Ethers/toxicity , Female , Fetal Blood/drug effects , Heterochromatin/drug effects , Heterochromatin/genetics , Humans , Mercury/toxicity , Mice , Organophosphates/toxicity , Selenium/toxicity
8.
Biochem Biophys Res Commun ; 462(4): 332-8, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-25960295

ABSTRACT

In mammalian livers, sexual dimorphisms are observed in tissue-specific functions and diseases such as hepatocellular carcinoma. We identified sex-dependent differentially methylated regions (S-DMRs) which had been previously been characterized as growth hormone- STAT5 dependent. In this study, we performed genome-wide screening and identified ten additional hypomethylated S-DMR gene regions in male livers. Of these S-DMRs, Uggt2 and Sarnp were hypomethylated in both male and female livers compared to brain and embryonic stem (ES) cells. Similarly, Adam2, Uggt2, and Scp2 were hypomethylated in female embryonic germ (EG) cells and not in male EG cells, indicating that these S-DMRs are liver-specific male hypo-S-DMRs. Interestingly, the five S-DMRs were free from STAT5 chromatin immunoprecipitation (ChIP) signals, suggesting that S-DMRs are independent of the growth hormone-STAT5-pathway. Instead, the DNA methylation statuses of the S-DMRs of Adam2, Snx29, Uggt2, Sarnp, and Rnpc3 genes were under the control of testosterone. Importantly, the hypomethylated S-DMRs of the Adam2 and Snx29 regions showed chromatin decondensation. Epigenetic factors could be responsible for the sexual dimorphisms in DNA methylation status and chromatin structure, as the expression of Dnmt1, Dnmt3b, and Tet2 genes was lower in male mice compared to female mice and TET2 expression recovered following orchidectomy by testosterone treatment. In conclusion, we identified novel male-specific hypomethylated S-DMRs that contribute to chromatin decondensation in the liver. S-DMRs were tissue-specific and the hypomethylation is testosterone-dependent.


Subject(s)
DNA Methylation , Liver/metabolism , Animals , Chromatin/metabolism , DNA Methylation/drug effects , Fatty Liver/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Sex Characteristics , Testosterone/pharmacology
9.
J Am Soc Nephrol ; 26(10): 2388-97, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25653098

ABSTRACT

Epigenetic mechanisms may underlie the progression of diabetic kidney disease. Because the kidney is a heterogeneous organ with different cell types, we investigated DNA methylation status of the kidney in a cell type-specific manner. We first identified genes specifically demethylated in the normal proximal tubules obtained from control db/m mice, and next delineated the candidate disease-modifying genes bearing aberrant DNA methylation induced by diabetes using db/db mice. Genes involved in glucose metabolism, including Sglt2, Pck1, and G6pc, were selectively hypomethylated in the proximal tubules in control mice. Hnf4a, a transcription factor regulating transporters for reabsorption, was also selectively demethylated. In diabetic mice, aberrant hypomethylation of Agt, Abcc4, Cyp4a10, Glut5, and Met and hypermethylation of Kif20b, Cldn18, and Slco1a1 were observed. Time-dependent demethylation of Agt, a marker of diabetic kidney disease, was accompanied by histone modification changes. Furthermore, inhibition of DNA methyltransferase or histone deacetylase increased Agt mRNA in cultured human proximal tubular cells. Aberrant DNA methylation and concomitant changes in histone modifications and mRNA expression in the diabetic kidney were resistant to antidiabetic treatment with pioglitazone. These results suggest that an epigenetic switch involving aberrant DNA methylation causes persistent mRNA expression of select genes that may lead to phenotype changes of the proximal tubules in diabetic kidney disease.


Subject(s)
DNA Methylation , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Kidney Tubules, Proximal/metabolism , Animals , Male , Mice , Mice, Inbred C57BL
10.
Auton Neurosci ; 187: 56-62, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25443216

ABSTRACT

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.


Subject(s)
Aging , Autonomic Nervous System/drug effects , Hexosamines/pharmacology , Sleep/drug effects , Wakefulness/drug effects , Animals , Autonomic Nervous System/physiology , Blood Pressure/drug effects , Body Temperature/drug effects , Brain Waves/drug effects , Electrocardiography , Electroencephalography , Electromyography , Fourier Analysis , Heart Rate/drug effects , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Statistics as Topic , Telemetry
11.
Mech Dev ; 136: 143-54, 2015 May.
Article in English | MEDLINE | ID: mdl-25528267

ABSTRACT

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.


Subject(s)
Epigenesis, Genetic , Nodal Protein/genetics , Regulatory Sequences, Nucleic Acid/genetics , Animals , Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Genome , Genome, Human , Humans , Mice , Nodal Protein/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Signal Transduction/genetics
12.
PLoS One ; 9(12): e114414, 2014.
Article in English | MEDLINE | ID: mdl-25514169

ABSTRACT

Extensive studies have been conducted to characterize the unique phenomena of equine pregnancy. Most studies have focused on embryo transmigration when the embryo is covered with a mucin-like glycoprotein capsule and on the characterization of the chorionic girdle and chorionic gonadotropin (CG) secretion. However, the events preceding and following capsule disappearance have not been well studied. In this study, the mRNA expression in conceptus membranes at days 19, 21, and 25 (day 0 = day of ovulation) was analyzed by RNA-seq (SOLiD3), and transcript levels on these three days and day 13 were confirmed by real-time PCR. Of the 26,416 equine genes registered, 20,436 transcripts were aligned to sequences in the Ensembl database, from which 4,625 transcripts were registered in both Ensembl and the KEGG pathway. Each of the 4,625 transcripts was examined through KEGG pathway analysis, and 12 transcripts of integrins (ITGs) and collagens (COLs) were confirmed through real-time PCR. Our data indicated that extracellular matrix (ECM)-related mRNAs were highly expressed in day 19, 21, and 25 conceptus membranes. In combination with previous results, which confirmed a lack of laminin and fibronectin transcript expression in the endometrium, these observations suggest that in contrast to attachment through focal adhesion, conceptus chorionic membrane ECMs function as a scaffold-like structure to possibly maintain the shape of the conceptus and a separation between chorionic membranes and the uterine luminal epithelium.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Horses/embryology , Horses/genetics , Zygote/chemistry , Animals , Base Sequence , DNA Primers/genetics , Extracellular Matrix Proteins/metabolism , Female , Gene Expression Profiling/veterinary , Molecular Sequence Data , Pregnancy , Real-Time Polymerase Chain Reaction/veterinary , Sequence Analysis, RNA/veterinary
13.
Int J Dev Biol ; 58(2-4): 231-8, 2014.
Article in English | MEDLINE | ID: mdl-25023689

ABSTRACT

DNA methylation functions as cellular memory beyond generations of cells and is involved in many biological processes. Because of its relatively stable nature compared with the transcriptome, the DNA methylation profile of cells can also be used to evaluate developmental similarity and cellular phenotypes. Recent insights into 5-hydroxymethylcytosine have started to reshape our view of the epigenetic regulation of mammalian development. Both global DNA methylation and hydroxymethylation levels change dynamically during preimplantation embryogenesis. It is known that DNA methylation plays an essential role in embryonic cell fate restriction, whereas its role in trophoblast development requires further research. Two distinct blastocyst-derived stem cell lines, embryonic stem (ES) cells and trophoblast stem (TS) cells, are used to study the epigenetic mechanisms underlying cell lineage maintenance and the regulation of cell differentiation. Such studies will allow us to understand the details of the epigenetic landscape of trophoblast development, which should offer valuable information for managing pregnancy-related diseases in humans.


Subject(s)
Cell Lineage/genetics , DNA Methylation , Embryo, Mammalian/cytology , Stem Cells/cytology , Trophoblasts/cytology , Animals , Cell Differentiation , Embryo, Mammalian/physiology , Epigenesis, Genetic , Female , Humans , Pregnancy , Stem Cells/metabolism , Trophoblasts/metabolism
14.
Genesis ; 51(11): 763-76, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23913699

ABSTRACT

Porcine induced pluripotent stem cells (iPSCs) provide useful information for translational research. The quality of iPSCs can be assessed by their ability to differentiate into various cell types after chimera formation. However, analysis of chimera formation in pigs is a labor-intensive and costly process, necessitating a simple evaluation method for porcine iPSCs. Our previous study identified mouse embryonic stem cell (ESC)-specific hypomethylated loci (EShypo-T-DMRs), and, in this study, 36 genes selected from these were used to evaluate porcine iPSC lines. Based on the methylation profiles of the 36 genes, the iPSC line, Porco Rosso-4, was found closest to mouse pluripotent stem cells among 5 porcine iPSCs. Moreover, Porco Rosso-4 more efficiently contributed to the inner cell mass (ICM) of blastocysts than the iPSC line showing the lowest reprogramming of the 36 genes (Porco Rosso-622-14), indicating that the DNA methylation profile correlates with efficiency of ICM contribution. Furthermore, factors known to enhance iPSC quality (serum-free medium with PD0325901 and CHIR99021) improved the methylation status at the 36 genes. Thus, the DNA methylation profile of these 36 genes is a viable index for evaluation of porcine iPSCs.


Subject(s)
DNA Methylation , Embryonic Stem Cells/metabolism , Genetic Loci , Induced Pluripotent Stem Cells/metabolism , Swine, Miniature/embryology , Swine/embryology , Animals , Blastocyst Inner Cell Mass/metabolism , Cell Line , Chimera , Embryo, Mammalian , Gene Expression , Genes , Mice , Translational Research, Biomedical
15.
Mech Ageing Dev ; 134(7-8): 331-7, 2013.
Article in English | MEDLINE | ID: mdl-23707638

ABSTRACT

In mammals, differences in liver function and aging have been observed between sexes; however, the epigenetic mechanisms underlying such differences remain largely unexplored. In this study, we investigated sex- and age-dependent DNA methylation status in the mouse liver. We analyzed 90 known sex-differentially expressed genes, and identified sex-dependent methylation in Zfp809, Hsd3b5, Treh, Cxcl11, Cyp17a1, and Nnmt genes. After 4 weeks of age, we noted the gradual establishment of sex-dependent hypomethylation in each of these genes in either males or females. The exposure of male mice to female-like growth hormone (GH) profile repressed male-predominant hypomethylation and promoted female-predominant hypomethylation. The occurrence of age-dependent hypomethylation, including at loci for which we also observed sex-dependent changes in DNA methylation, was accompanied by the downregulation of DNMT3A/B. In addition, we found that age-dependent hypomethylation was promoted through liver regeneration induced by partial hepatectomy, suggesting that DNMT activities were not enough to retain methylation levels. In conclusion, our results demonstrate that sex-dependent GH profiles influence the age-progressive hypomethylation under decreased DNMT3A/B levels in certain regions of the genome.


Subject(s)
Aging/metabolism , DNA Methylation/drug effects , Gene Expression Regulation/drug effects , Growth Hormone/pharmacology , Liver Regeneration/drug effects , Liver/metabolism , Sex Characteristics , Aging/drug effects , Animals , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Female , Genetic Loci/physiology , Male , Mice , DNA Methyltransferase 3B
16.
J Biol Chem ; 288(24): 17099-110, 2013 Jun 14.
Article in English | MEDLINE | ID: mdl-23625921

ABSTRACT

The orexin system plays a central role in the integration of sleep/wake and feeding behaviors in a broad spectrum of neural-metabolic physiology. Orexin-A and orexin-B are produced by the cleavage of prepro-orexin, which is encoded on the Hcrt gene. To date, methods for generating other peptide neurons could not induce orexin neurons from pluripotent stem cells. Considering that the metabolic status affects orexin expression, we supplemented the culture medium with a nutrient factor, ManNAc, and succeeded in generating functional orexin neurons from mouse ES cells. Because DNA methylation inhibitors and histone deacetylase inhibitors could induce Hcrt expression in mouse ES cells, the epigenetic mechanism may be involved in this orexin neurogenesis. DNA methylation analysis showed the presence of a tissue-dependent differentially methylated region (T-DMR) around the transcription start site of the Hcrt gene. In the orexin neurons induced by supplementation of ManNAc, the T-DMR of the Hcrt gene was hypomethylated in association with higher H3/H4 acetylation. Concomitantly, the histone acetyltransferases p300, CREB-binding protein (CBP), and Mgea5 (also called O-GlcNAcase) were localized to the T-DMR in the orexin neurons. In non-orexin-expressing cells, H3/H4 hypoacetylation and hyper-O-GlcNAc modification were observed at the T-DMRs occupied by O-GlcNAc transferase and Sirt1. Therefore, the results of the present study suggest that the glucose metabolite, ManNAc, induces switching from the inactive state by Ogt-Sirt1 to the active state by Mgea5, p300, and CBP at the Hcrt gene locus.


Subject(s)
Embryonic Stem Cells/physiology , Epigenesis, Genetic/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Acetylation , Animals , Cell Differentiation , Cytidine/analogs & derivatives , Cytidine/pharmacology , DNA Methylation , DNA-Cytosine Methylases/antagonists & inhibitors , DNA-Cytosine Methylases/metabolism , Deoxycytidine/pharmacology , Embryonic Stem Cells/drug effects , Female , Glycosylation , Hexosamines/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Hydroxamic Acids/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Neuropeptides/genetics , Orexins , Protein Processing, Post-Translational , Sequence Analysis, DNA , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/metabolism , Spheroids, Cellular , Transcription Initiation Site
17.
BMC Genomics ; 14: 82, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23387509

ABSTRACT

BACKGROUND: Tissues and their component cells have unique DNA methylation profiles comprising DNA methylation patterns of tissue-dependent and differentially methylated regions (T-DMRs). Previous studies reported that DNA methylation plays crucial roles in cell differentiation and development. Here, we investigated the genome-wide DNA methylation profiles of mouse neural progenitors derived from different developmental stages using HpyCH4IV, a methylation-sensitive restriction enzyme that recognizes ACGT residues, which are uniformly distributed across the genome. RESULTS: Using a microarray-based genome-wide DNA methylation analysis system focusing on 8.5-kb regions around transcription start sites (TSSs), we analyzed the DNA methylation profiles of mouse neurospheres derived from telencephalons at embryonic days 11.5 (E11.5NSph) and 14.5 (E14.5NSph) and the adult brain (AdBr). We identified T-DMRs with different DNA methylation statuses between E11.5NSph and E14.5NSph at genes involved in neural development and/or associated with neurological disorders in humans, such as Dclk1, Nrcam, Nfia, and Ntng1. These T-DMRs were located not only within 2 kb but also distal (several kbs) from the TSSs, and those hypomethylated in E11.5NSph tended to be in CpG island (CGI-) associated genes. Most T-DMRs that were hypomethylated in neurospheres were also hypomethylated in the AdBr. Interestingly, among the T-DMRs hypomethylated in the progenitors, there were T-DMRs that were hypermethylated in the AdBr. Although certain genes, including Ntng1, had hypermethylated T-DMRs 5' upstream, we identified hypomethylated T-DMRs in the AdBr, 3' downstream from their TSSs. This observation could explain why Ntng1 was highly expressed in the AdBr despite upstream hypermethylation. CONCLUSION: Mouse adult brain DNA methylation and gene expression profiles could be attributed to developmental dynamics of T-DMRs in neural-related genes.


Subject(s)
Brain/growth & development , Cell Differentiation/genetics , DNA Methylation/genetics , Gene Expression Regulation, Developmental , Animals , CpG Islands/genetics , Embryonic Stem Cells/cytology , Genome , Mice , Oligonucleotide Array Sequence Analysis , Organ Specificity , Transcription Initiation Site
18.
Mol Reprod Dev ; 80(3): 212-22, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23325669

ABSTRACT

DNA methyltransferase1o (Dnmt1o), which is specific to oocyte and preimplantation embryo, plays a role in maintaining DNA methylation in mammalian cells. Here, we investigated the methylation status of CpGs sites in the Dnmt1o 5'-flanking region in germ cells at different stages of oogenesis or spermatogenesis. The methylation levels of the CpG sites at the 5'-flanking regions were hypermethylated in growing oocytes of all follicular stages, while the oocytes in meiotic metaphase II (MII) were demethylated. The methylation pattern within the CpGs sites in the 5'-flanking region, however, was dramatically changed during spermatogenesis. We observed that there was significant non-CpG methylation both in MII oocytes and spermatocytes. Although a low methylation level in non-CpG sites was observed in primary and secondary oocytes, the CpA site of position 25 and CpT site of position 29 within the no-CpG region in the 5'-flanking region of Dnmt1o was highly methylated in MII oocytes. During spermatogenesis, the low degree of methylation at CpG sites in spermatocytes increased to a higher degree in sperm, while the high ratio of methylation in non-CpG sites in spermatocytes decreased. Together, germ cells showed inverted methylation patterns between CpG and non-CpG sites in the Dnmt1o 5'-upstream region, and the methylation pattern during oogenesis did not drastically change, remaining generally hypomethylated at the MII stage.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , Oogenesis/genetics , Spermatogenesis/genetics , Animals , CpG Islands , DNA (Cytosine-5-)-Methyltransferase 1 , Female , Histocytochemistry , Male , Mice , Mice, Inbred C57BL , Ovarian Follicle/cytology , Ovary/cytology , Photomicrography , Testis/cytology
19.
Biosci Biotechnol Biochem ; 76(12): 2249-54, 2012.
Article in English | MEDLINE | ID: mdl-23221698

ABSTRACT

Sialic acids may modulate cell proliferation and gene expression, particularly in neural cells in vitro. However, the function of sialic acids in the central nervous system has not previously been examined. We examined whether N-acetylmannosamine (ManNAc) could improve object recognition and hippocampal cell proliferations in middle-aged mice. C56BL/6J mice aged 52 weeks were treated with ManNAc for 4 weeks. Their cognitive-ability was assessed with a place and object recognition test. ManNAc, but not N-acetylglucosamine or N-acetylneuraminic acid, improved the index score in the place recognition task at a dosage of 5.0 mg/mL in drinking water. Additionally, ManNAc enhanced the hippocampal cell proliferation, which was evaluated by a bromodeoxyuridine assay and the number of Ki67-immunoreactive cells. We could demonstrate that ManNAc had positive effects on the age-related brain dysfunction. These findings suggest that the use of ManNAc or related compounds may be a new approach for the treatment of human dementia.


Subject(s)
Hexosamines/pharmacology , Hippocampus/cytology , Hippocampus/physiology , Recognition, Psychology/drug effects , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Cell Proliferation/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Neurogenesis/drug effects
20.
BMC Immunol ; 13: 58, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-23116172

ABSTRACT

BACKGROUND: Medullary thymic epithelial cells (mTECs) are characterized by ectopic expression of self-antigens during the establishment of central tolerance. The autoimmune regulator (Aire), which is specifically expressed in mTECs, is responsible for the expression of a large repertoire of tissue-restricted antigens (TRAs) and plays a role in the development of mTECs. However, Aire-deficient mTECs still express TRAs. Moreover, a subset of mTECs, which are considered to be at a stage of terminal differentiation, exists in the Aire-deficient thymus. The phenotype of a specific cell type in a multicellular organism is governed by the epigenetic regulation system. DNA methylation modification is an important component of this system. Every cell or tissue type displays a DNA methylation profile, consisting of tissue-dependent and differentially methylated regions (T-DMRs), and this profile is involved in cell-type-specific genome usage. The aim of this study was to examine the DNA methylation profile of mTECs by using Aire-deficient mTECs as a model. RESULTS: We identified the T-DMRs of mTECs (mTEC-T-DMRs) via genome-wide DNA methylation analysis of Aire(-/-) mTECs by comparison with the liver, brain, thymus, and embryonic stem cells. The hypomethylated mTEC-T-DMRs in Aire(-/-) mTECs were associated with mTEC-specific genes, including Aire, CD80, and Trp63, as well as other genes involved in the RANK signaling pathway. While these mTEC-T-DMRs were also hypomethylated in Aire(+/+) mTECs, they were hypermethylated in control thymic stromal cells. We compared the pattern of DNA methylation levels at a total of 55 mTEC-T-DMRs and adjacent regions and found that the DNA methylation status was similar for Aire(+/+) and Aire(-/-) mTECs but distinct from that of athymic cells and tissues. CONCLUSIONS: These results indicate a unique DNA methylation profile that is independent of Aire in mTECs. This profile is distinct from other cell types in the thymic microenvironment and is indicated to be involved in the differentiation of the mTEC lineage.


Subject(s)
DNA Methylation/genetics , Epithelial Cells/metabolism , Thymus Gland/cytology , Transcription Factors/deficiency , Animals , Biomarkers/metabolism , Cell Separation , Gene Expression Profiling , Gene Expression Regulation , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Organ Specificity/genetics , Stromal Cells/metabolism , Transcription Factors/metabolism , Transcription Initiation Site , AIRE Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...