Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters











Publication year range
1.
Biochem Soc Trans ; 52(2): 639-650, 2024 04 24.
Article in English | MEDLINE | ID: mdl-38506536

ABSTRACT

Pluripotent stem cells (PSCs), comprising embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), offer immense potential for regenerative medicine due to their ability to differentiate into all cell types of the adult body. A critical aspect of harnessing this potential is understanding their metabolic requirements during derivation, maintenance, and differentiation in vitro. Traditional culture methods using fetal bovine serum often lead to issues such as heterogeneous cell populations and diminished pluripotency. Although the chemically-defined 2i/LIF medium has provided solutions to some of these challenges, prolonged culturing of these cells, especially female ESCs, raises concerns related to genome integrity. This review discusses the pivotal role of lipids in genome stability and pluripotency of stem cells. Notably, the introduction of lipid-rich albumin, AlbuMAX, into the 2i/LIF culture medium offers a promising avenue for enhancing the genomic stability and pluripotency of cultured ESCs. We further explore the unique characteristics of lipid-induced pluripotent stem cells (LIP-ESCs), emphasizing their potential in regenerative medicine and pluripotency research.


Subject(s)
Genomic Instability , Lipids , Humans , Animals , Lipids/chemistry , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Cell Differentiation , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Lipid Metabolism
2.
bioRxiv ; 2023 Sep 29.
Article in English | MEDLINE | ID: mdl-37808786

ABSTRACT

Chromatin is a crucial regulator of gene expression and tightly controls development across species. Mutations in only one copy of multiple histone genes were identified in children with developmental disorders characterized by microcephaly, but their mechanistic roles in development remain unclear. Here we focus on dominant mutations affecting histone H4 lysine 91. These H4K91 mutants form aberrant nuclear puncta at specific heterochromatin regions. Mechanistically, H4K91 mutants demonstrate enhanced binding to the histone variant H3.3, and ablation of H3.3 or the H3.3-specific chaperone DAXX diminishes the mutant localization to chromatin. Our functional studies demonstrate that H4K91 mutant expression increases chromatin accessibility, alters developmental gene expression through accelerating pro-neural differentiation, and causes reduced mouse brain size in vivo, reminiscent of the microcephaly phenotypes of patients. Together, our studies unveil a distinct molecular pathogenic mechanism from other known histone mutants, where H4K91 mutants misregulate cell fate during development through abnormal genomic localization.

3.
Protein Cell ; 14(8): 591-602, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37029701

ABSTRACT

While Mek1/2 and Gsk3ß inhibition ("2i") supports the maintenance of murine embryonic stem cells (ESCs) in a homogenous naïve state, prolonged culture in 2i results in aneuploidy and DNA hypomethylation that impairs developmental potential. Additionally, 2i fails to support derivation and culture of fully potent female ESCs. Here we find that mouse ESCs cultured in 2i/LIF supplemented with lipid-rich albumin (AlbuMAX) undergo pluripotency transition yet maintain genomic stability and full potency over long-term culture. Mechanistically, lipids in AlbuMAX impact intracellular metabolism including nucleotide biosynthesis, lipid biogenesis, and TCA cycle intermediates, with enhanced expression of DNMT3s that prevent DNA hypomethylation. Lipids induce a formative-like pluripotent state through direct stimulation of Erk2 phosphorylation, which also alleviates X chromosome loss in female ESCs. Importantly, both male and female "all-ESC" mice can be generated from de novo derived ESCs using AlbuMAX-based media. Our findings underscore the importance of lipids to pluripotency and link nutrient cues to genome integrity in early development.


Subject(s)
Embryonic Stem Cells , Mouse Embryonic Stem Cells , Male , Animals , Female , Mice , Genomic Instability , Lipids , DNA/metabolism , Cell Differentiation
4.
Methods Cell Biol ; 170: 203-210, 2022.
Article in English | MEDLINE | ID: mdl-35811100

ABSTRACT

Mice derived entirely from embryonic stem (ES) cells can be generated through tetraploid complementation. Although XY male ES cell lines are commonly used in this system, occasionally, monosomic XO female mice are produced through spontaneous Y chromosome loss. Here, we describe an efficient method to obtain monosomic XO ES cells by CRISPR/Cas9-mediated deletion of the Y chromosome allowing generation of female clonal mice by tetraploid complementation. The monosomic XO female mice are viable and able to produce normal male and female offspring. Direct generation of clonal mice in both sexes can significantly accelerate the production of complex genetically modified mouse models.


Subject(s)
CRISPR-Cas Systems , Tetraploidy , Animals , CRISPR-Cas Systems/genetics , Chromosome Deletion , Chromosomes, Human, Y , Embryonic Stem Cells , Female , Infertility, Male , Male , Mice , Sex Chromosome Aberrations , Sex Chromosome Disorders of Sex Development
6.
Nat Cell Biol ; 24(1): 99-111, 2022 01.
Article in English | MEDLINE | ID: mdl-34961794

ABSTRACT

Histone variants and the associated post-translational modifications that govern the stemness of haematopoietic stem cells (HSCs) and differentiation thereof into progenitors (HSPCs) have not been well defined. H3.3 is a replication-independent H3 histone variant in mammalian systems that is enriched at both H3K4me3- and H3K27me3-marked bivalent genes as well as H3K9me3-marked endogenous retroviral repeats. Here we show that H3.3, but not its chaperone Hira, prevents premature HSC exhaustion and differentiation into granulocyte-macrophage progenitors. H3.3-null HSPCs display reduced expression of stemness and lineage-specific genes with a predominant gain of H3K27me3 marks at their promoter regions. Concomitantly, loss of H3.3 leads to a reduction of H3K9me3 marks at endogenous retroviral repeats, opening up binding sites for the interferon regulatory factor family of transcription factors, allowing the survival of rare, persisting H3.3-null HSCs. We propose a model whereby H3.3 maintains adult HSC stemness by safeguarding the delicate interplay between H3K27me3 and H3K9me3 marks, enforcing chromatin adaptability.


Subject(s)
Chromatin/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Histones/metabolism , Myelopoiesis/physiology , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Cycle Proteins , Cell Line , Granulocytes/cytology , Hematopoiesis/physiology , Histone Chaperones , Human Umbilical Vein Endothelial Cells , Humans , Macrophages/cytology , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic/genetics , Protein Processing, Post-Translational/physiology , Transcription Factors
7.
Cell Regen ; 10(1): 18, 2021 May 05.
Article in English | MEDLINE | ID: mdl-33948765

ABSTRACT

On January 28 in Nature online, Zheng et al. (Nature, 2021 (Online ahead of print)) reported that they developed an ingenious method of interspecies PSC co-culture system in vitro which unfolded interspecific cell competition. This study paves the way for discovering the mechanism of interspecific chimera and for further interspecific organogenesis between evolutionarily distant species.

8.
CRISPR J ; 4(1): 147-154, 2021 02.
Article in English | MEDLINE | ID: mdl-33567216

ABSTRACT

Mice derived entirely from embryonic stem (ES) cells can be generated through tetraploid complementation. Although XY male ES cell lines are commonly used in this system, occasionally, monosomic XO female mice are produced through spontaneous Y chromosome loss. Here, we describe an efficient method to obtain monosomic XO ES cells by CRISPR-Cas9-mediated deletion of the Y chromosome, allowing generation of female clonal mice by tetraploid complementation. The monosomic XO female mice are viable and able to produce normal male and female offspring. Direct generation of clonal mice in both sexes can significantly accelerate the production of complex genetically modified mouse models.


Subject(s)
CRISPR-Cas Systems , Chromosome Deletion , Embryonic Stem Cells , Infertility, Male , Sex Chromosome Aberrations , Sex Chromosome Disorders of Sex Development , Y Chromosome , Animals , Cell Line , Chromosomes, Human, Y , Clustered Regularly Interspaced Short Palindromic Repeats , Embryo, Mammalian , Female , Genotyping Techniques , Karyotyping , Male , Mice
9.
F S Sci ; 2(3): 237-247, 2021 08.
Article in English | MEDLINE | ID: mdl-35560275

ABSTRACT

OBJECTIVE: To measure the influence of exogenous insulin-like growth factor 1 (IGF1) on follicle growth and maturation in human ovarian cortical xenografts. DESIGN: Xenotransplantation model. SETTING: University-based research laboratory. PATIENTS/ANIMALS: Ovarian tissue was donated with consent and institutional review board approval by brain-dead organ donors or patients undergoing ovarian tissue cryopreservation for fertility preservation. Cortical fragments were transplanted into immunocompromised mice. INTERVENTIONS: Cryopreserved ovarian cortical fragments from four women (aged 19, 25, 33, and 46 years) were transplanted into the gluteus muscle of immunocompromised mice in a fibrin matrix containing endothelial cells that were transduced with lentiviral particles encoding secreted IGF1. Xenografts were recovered after 3, 8, and 14 weeks. In addition, C57/Bl6 mice underwent intraovarian injection of saline or recombinant IGF1 (60 µg), followed by superovulation, analysis of ethynyl-deoxyuridine incorporation, and ribonucleic acid sequencing of the whole ovaries. MAIN OUTCOME MEASURES: For xenografts: follicle count and distribution; antral follicle count; and corpora lutea/albicans count. For mice: follicle count and distribution; oocyte yield, ethynyl-deoxyuridine incorporation (granulosa cell proliferation); and ovarian transcriptomic signature. RESULTS: At 3 weeks, xenografts in the IGF1 condition revealed a decreased percentage of primary follicles and increased percentage of secondary follicles that were concentrated in the preantral subtype; at 8 weeks, an increase in secondary follicles was concentrated in the simple subtype; after 14 weeks, primordial follicles were reduced, and while the number of advanced follicles did not power the experiment to demonstrate significance, antral follicles reduced and corpora lutea increased. Supporting experiments in mice revealed an increase in normal oocytes following intraovarian injection of recombinant IGF1 (60 µg) as well as increased proliferative index among follicles of secondary and preantral stages. Ribonucleic acid sequencing analysis of the whole ovaries following injection of recombinant IGF1 (25 µg) revealed an acute (24 hours) upregulation of transcripts related to steroidogenesis and luteinization. CONCLUSIONS: Exogenous IGF1 advances the pace of growth among primordial, primary, and secondary stage follicles but results in near absence of antral stage follicles in long-term (14 weeks) xenografts. In mice, acute administration of IGF1 promotes follicle advance and increased oocyte yield. The results suggest that while superphysiological IGF1 alone advances the pace of growth among early/preantral follicles, a sustained and/or later-stage influence undermines antral follicle growth/survival or promotes premature luteinization. These findings provide a temporal framework for interpreting follicle growth/mobilization and may be useful in understanding the clinical application of human growth hormone in the context of assisted reproduction.


Subject(s)
Insulin-Like Growth Factor I , Ovary , Animals , Deoxyuridine , Endothelial Cells , Female , Heterografts , Humans , Mice , Ovary/physiology , RNA , Transplantation, Heterologous
10.
Stem Cells ; 38(5): 613-623, 2020 05.
Article in English | MEDLINE | ID: mdl-31916656

ABSTRACT

Sphingosine-1-phosphate (S1P) is a bioactive lipid molecule regulating organogenesis, angiogenesis, cell proliferation, and apoptosis. S1P is generated by sphingosine kinases (SPHK1 and SPHK2) through the phosphorylation of ceramide-derived sphingosine. Phenotypes caused by manipulating S1P metabolic enzymes and receptors suggested several possible functions for S1P in embryonic stem cells (ESCs), yet the mechanisms by which S1P and related sphingolipids act in ESCs are controversial. We designed a rigorous test to evaluate the requirement of S1P in murine ESCs by knocking out both Sphk1 and Sphk2 to create cells incapable of generating S1P. To accomplish this, we created lines mutant for Sphk2 and conditionally mutant (floxed) for Sphk1, allowing evaluation of ESCs that transition to double-null state. The Sphk1/2-null ESCs lack S1P and accumulate the precursor sphingosine. The double-mutant cells fail to grow due to a marked cell cycle arrest at G2/M. Mutant cells activate expression of telomere elongation factor genes Zscan4, Tcstv1, and Tcstv3 and display longer telomeric repeats. Adding exogenous S1P to the medium had no impact, but the cell cycle arrest is partially alleviated by the expression of a ceramide synthase 2, which converts excess sphingosine into ceramide. The results indicate that sphingosine kinase activity is essential in mouse ESCs for limiting the accumulation of sphingosine that otherwise drives cell cycle arrest.


Subject(s)
Cell Cycle Checkpoints/drug effects , Embryonic Stem Cells/metabolism , Phosphotransferases (Alcohol Group Acceptor)/adverse effects , Animals , Cell Proliferation , Female , Humans , Mice , Mice, Knockout
11.
J Biol Chem ; 294(47): 17863-17874, 2019 11 22.
Article in English | MEDLINE | ID: mdl-31624145

ABSTRACT

Long noncoding RNAs (lncRNAs) modulate gene expression as competing endogenous RNAs (ceRNAs) that sponge regulatory microRNAs (miRNAs). During cellular reprogramming, genes associated with pluripotency establishment need to be up-regulated, and developmental genes need to be silenced. However, how ceRNAs control cellular reprogramming still awaits full elucidation. Here, we used doxycycline-inducible expression of the four transcription factors octamer-binding protein 4 (OCT4), SRY-box 2 (SOX2), Krüppel-like factor 4 (KLF4), and proto-oncogene c-Myc (c-Myc) to generate induced pluripotent stem cells (iPSCs) from mouse embryonic fibroblasts (MEFs). Using RNA-Seq and bioinformatics approaches, we found that the expression levels of miRNAs from MEFs remain high from day 0 to 6 after the doxycycline induction. Many genes targeted by these miRNAs were up-regulated, and long intergenic noncoding RNAs (lincRNAs) and circular RNAs (circRNAs), which have complementary binding sites to these miRNAs, were highly expressed, indicating lincRNAs and circRNAs may function as ceRNAs. Intriguingly, knockdown of the linc/circRNAs that sponge the miRNAs, which target OCT4 down-regulated exogenous OCT4, decreased reprogramming efficiency, and resulted in low-grade iPSCs. Our results suggest that the ceRNA network plays an important role in cellular reprogramming.


Subject(s)
Cellular Reprogramming/genetics , Gene Expression Regulation , MicroRNAs/metabolism , Octamer Transcription Factor-3/genetics , RNA, Long Noncoding/metabolism , Animals , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Mice, Inbred C57BL , Models, Biological , Octamer Transcription Factor-3/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , RNA, Long Noncoding/genetics
12.
Nat Cell Biol ; 20(5): 565-574, 2018 05.
Article in English | MEDLINE | ID: mdl-29662175

ABSTRACT

A robust network of transcription factors and an open chromatin landscape are hallmarks of the naive pluripotent state. Recently, the acetyllysine reader Brd4 has been implicated in stem cell maintenance, but the relative contribution of Brd4 to pluripotency remains unclear. Here, we show that Brd4 is dispensable for self-renewal and pluripotency of embryonic stem cells (ESCs). When maintained in their ground state, ESCs retain transcription factor binding and chromatin accessibility independent of Brd4 function or expression. In metastable ESCs, Brd4 independence can be achieved by increased expression of pluripotency transcription factors, including STAT3, Nanog or Klf4, so long as the DNA methylcytosine oxidases Tet1 and Tet2 are present. These data reveal that Brd4 is not essential for ESC self-renewal. Rather, the levels of pluripotency transcription factor abundance and Tet1/2 function determine the extent to which bromodomain recognition of protein acetylation contributes to the maintenance of gene expression and cell identity.


Subject(s)
Cell Differentiation , Cell Lineage , Cell Self Renewal , DNA-Binding Proteins/metabolism , Mouse Embryonic Stem Cells/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Acetylation , Animals , Binding Sites , Cell Line , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly , DNA-Binding Proteins/genetics , Dioxygenases , Female , Gene Expression Regulation, Developmental , Histones/genetics , Histones/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred ICR , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Nuclear Proteins/genetics , Phenotype , Protein Binding , Protein Processing, Post-Translational , Proto-Oncogene Proteins/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Transcription Factors/genetics
13.
J Biol Chem ; 293(10): 3829-3838, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29358330

ABSTRACT

Derepression of chromatin-mediated transcriptional repression of paternal and maternal genomes is considered the first major step that initiates zygotic gene expression after fertilization. The histone variant H3.3 is present in both male and female gametes and is thought to be important for remodeling the paternal and maternal genomes for activation during both fertilization and embryogenesis. However, the underlying mechanisms remain poorly understood. Using our H3.3B-HA-tagged mouse model, engineered to report H3.3 expression in live animals and to distinguish different sources of H3.3 protein in embryos, we show here that sperm-derived H3.3 (sH3.3) protein is removed from the sperm genome shortly after fertilization and extruded from the zygotes via the second polar bodies (PBII) during embryogenesis. We also found that the maternal H3.3 (mH3.3) protein is incorporated into the paternal genome as early as 2 h postfertilization and is detectable in the paternal genome until the morula stage. Knockdown of maternal H3.3 resulted in compromised embryonic development both of fertilized embryos and of androgenetic haploid embryos. Furthermore, we report that mH3.3 depletion in oocytes impairs both activation of the Oct4 pluripotency marker gene and global de novo transcription from the paternal genome important for early embryonic development. Our results suggest that H3.3-mediated paternal chromatin remodeling is essential for the development of preimplantation embryos and the activation of the paternal genome during embryogenesis.


Subject(s)
Blastocyst/metabolism , Chromatin Assembly and Disassembly , Histones/metabolism , Paternal Inheritance , Transcriptional Activation , Animals , Blastocyst/cytology , Blastomeres/cytology , Blastomeres/metabolism , Embryonic Development , Female , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Histones/antagonists & inhibitors , Histones/genetics , Male , Mice , Mice, Inbred ICR , Mice, Transgenic , Morula/cytology , Morula/metabolism , Octamer Transcription Factor-3/chemistry , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
14.
J Ovarian Res ; 10(1): 64, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28927438

ABSTRACT

BACKGROUND: New data suggests that endothelial cells (ECs) elaborate essential "angiocrine factors". The aim of this study is to investigate the role of activated ovarian endothelial cells in early in-vitro follicular development. METHODS: Mouse ovarian ECs were isolated using magnetic cell sorting or by FACS and cultured in serum free media. After a constitutive activation of the Akt pathway was initiated, early follicles (50-150 um) were mechanically isolated from 8-day-old mice and co-cultured with these activated ovarian endothelial cells (AOEC) (n = 32), gel (n = 24) or within matrigel (n = 27) in serum free media for 14 days. Follicular growth, survival and function were assessed. RESULTS: After 6 passages, flow cytometry showed 93% of cells grown in serum-free culture were VE-cadherin positive, CD-31 positive and CD 45 negative, matching the known EC profile. Beginning on day 4 of culture, we observed significantly higher follicular and oocyte growth rates in follicles co-cultured with AOECs compared with follicles on gel or matrigel. After 14 days of culture, 73% of primary follicles and 83% of secondary follicles co-cultured with AOEC survived, whereas the majority of follicles cultured on gel or matrigel underwent atresia. CONCLUSIONS: This is the first report of successful isolation and culture of ovarian ECs. We suggest that co-culture with activated ovarian ECs promotes early follicular development and survival. This model is a novel platform for the in vitro maturation of early follicles and for the future exploration of endothelial-follicular communication. CAPSULE: In vitro development of early follicles necessitates a complex interplay of growth factors and signals required for development. Endothelial cells (ECs) may elaborate essential "angiocrine factors" involved in organ regeneration. We demonstrate that co-culture with ovarian ECs enables culture of primary and early secondary mouse ovarian follicles.


Subject(s)
Cell Culture Techniques , Endothelial Cells/cytology , Ovarian Follicle/cytology , Animals , Apoptosis , Cell Communication , Cell Proliferation , Cells, Cultured , Estradiol/metabolism , Female , Fibroblasts/cytology , Granulosa Cells/cytology , Mice , Oocytes/cytology , Ovarian Follicle/metabolism
15.
Nat Commun ; 7: 11275, 2016 04 27.
Article in English | MEDLINE | ID: mdl-27117588

ABSTRACT

Spermatogonial stem and progenitor cells (SSCs) generate adult male gametes. During in vitro expansion, these unipotent murine cells spontaneously convert to multipotent adult spermatogonial-derived stem cells (MASCs). Here we investigate this conversion process through integrative transcriptomic and epigenomic analyses. We find in SSCs that promoters essential to maintenance and differentiation of embryonic stem cells (ESCs) are enriched with histone H3-lysine4 and -lysine 27 trimethylations. These bivalent modifications are maintained at most somatic promoters after conversion, bestowing MASCs an ESC-like promoter chromatin. At enhancers, the core pluripotency circuitry is activated partially in SSCs and completely in MASCs, concomitant with loss of germ cell-specific gene expression and initiation of embryonic-like programs. Furthermore, SSCs in vitro maintain the epigenomic characteristics of germ cells in vivo. Our observations suggest that SSCs encode innate plasticity through the epigenome and that both conversion of promoter chromatin states and activation of cell type-specific enhancers are prominent features of reprogramming.


Subject(s)
Cell Differentiation/genetics , Cell Plasticity/genetics , Embryonic Stem Cells/metabolism , Epigenomics/methods , Multipotent Stem Cells/metabolism , Spermatogonia/metabolism , Animals , Cells, Cultured , Gene Expression Profiling/methods , Histones/metabolism , Lysine/metabolism , Male , Methylation , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Spermatogenesis/genetics , Spermatogonia/cytology
16.
Neuron ; 87(1): 77-94, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26139371

ABSTRACT

Turnover and exchange of nucleosomal histones and their variants, a process long believed to be static in post-replicative cells, remains largely unexplored in brain. Here, we describe a novel mechanistic role for HIRA (histone cell cycle regulator) and proteasomal degradation-associated histone dynamics in the regulation of activity-dependent transcription, synaptic connectivity, and behavior. We uncover a dramatic developmental profile of nucleosome occupancy across the lifespan of both rodents and humans, with the histone variant H3.3 accumulating to near-saturating levels throughout the neuronal genome by mid-adolescence. Despite such accumulation, H3.3-containing nucleosomes remain highly dynamic-in a modification-independent manner-to control neuronal- and glial-specific gene expression patterns throughout life. Manipulating H3.3 dynamics in both embryonic and adult neurons confirmed its essential role in neuronal plasticity and cognition. Our findings establish histone turnover as a critical and previously undocumented regulator of cell type-specific transcription and plasticity in mammalian brain.


Subject(s)
Brain/metabolism , Chromatin/metabolism , Gene Expression Regulation, Developmental , Histones/metabolism , Neuronal Plasticity/genetics , Neurons/metabolism , Nucleosomes/metabolism , Adolescent , Adult , Aged , Animals , Cerebellum/metabolism , Child , Child, Preschool , Epigenesis, Genetic , Female , Fetus , Frontal Lobe/metabolism , Hippocampus/metabolism , Humans , Male , Mice , Middle Aged , Transcription, Genetic , Young Adult
17.
Nucleus ; 5(5): 369-75, 2014.
Article in English | MEDLINE | ID: mdl-25482190

ABSTRACT

Transfer of a somatic nucleus into an enucleated oocyte is the most efficient approach for somatic cell reprogramming. While this process is known to involve extensive chromatin remodeling of the donor nucleus, the maternal factors responsible and the underlying chromatin-based mechanisms remain largely unknown. Here we discuss our recent findings demonstrating that the histone variant H3.3 plays an essential role in reprogramming and is required for reactivation of key pluripotency genes in somatic cell nuclear transfer (SCNT) embryos. Maternal-derived H3.3 replaces H3 in the donor nucleus shortly after oocyte activation, with the amount of replacement directly related to the differentiation status of the donor nucleus in SCNT embryos. We provide additional evidence to suggest that de novo synthesized H3.3 replaces histone H3 carrying repressive modifications in the donor nuclei of SCNT embryos, and hypothesize that replacement may occur at specific loci that must be reprogrammed for gene reactivation.


Subject(s)
Cellular Reprogramming/genetics , Chromatin Assembly and Disassembly/genetics , Epigenesis, Genetic , Histones/genetics , Animals , Cell Nucleus/genetics , Chromatin/genetics , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase , Mice , Nuclear Transfer Techniques , Oocytes/growth & development
18.
Genesis ; 52(12): 959-66, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25262655

ABSTRACT

Chromatin remodeling via incorporation of histone variants plays a key role in the regulation of embryonic development. The histone variant H3.3 has been associated with a number of early events including formation of the paternal pronucleus upon fertilization. The small number of amino acid differences between H3.3 and its canonical counterparts (H3.1 and H3.2) has limited studies of the developmental significance of H3.3 deposition into chromatin due to difficulties in distinguishing the H3 isoforms. To this end, we used zinc-finger nuclease (ZFN) mediated gene editing to introduce a small C-terminal hemagglutinin (HA) tag to the endogenous H3.3B locus in mouse embryonic stem cells (ESCs), along with an internal ribosome entry site (IRES) and a separately translated fluorescent reporter of expression. This system will allow detection of expression driven by the reporter in cells, animals, and embryos, and will facilitate investigation of differential roles of paternal and maternal H3.3 protein during embryogenesis that would not be possible using variant-specific antibodies. Further, the ability to monitor endogenous H3.3 protein in various cell lineages will enhance our understanding of the dynamics of this histone variant over the course of development.


Subject(s)
Embryo, Mammalian/metabolism , Genetic Engineering/methods , Histones/genetics , Histones/metabolism , Animals , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly , Embryonic Development , Female , Gene Expression Regulation, Developmental , Genetic Loci , Genetic Variation , Male , Mice , Protein Isoforms/genetics , Protein Isoforms/metabolism
19.
Cell Stem Cell ; 15(3): 281-294, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25192463

ABSTRACT

For future application of induced pluripotent stem cell (iPSC) technology, the ability to assess the overall quality of iPSC clones will be an important issue. Here we show that the histone variant H2A.X is a functional marker that can distinguish the developmental potentials of mouse iPSC lines. We found that H2A.X is specifically targeted to and negatively regulates extraembryonic lineage gene expression in embryonic stem cells (ESCs) and prevents trophectoderm lineage differentiation. ESC-specific H2A.X deposition patterns are faithfully recapitulated in iPSCs that support the development of "all-iPS" animals via tetraploid complementation, the most stringent test available of iPSC quality. In contrast, iPSCs that fail to support all-iPS embryonic development show aberrant H2A.X deposition, upregulation of extraembryonic lineage genes, and a predisposition to extraembryonic differentiation. Thus, our work has highlighted an epigenetic mechanism for maintaining cell lineage commitment in ESCs and iPSCs that can be used to distinguish the quality of iPSC lines.


Subject(s)
Epigenesis, Genetic , Histones/metabolism , Induced Pluripotent Stem Cells/metabolism , Animals , Base Sequence , Cell Differentiation/genetics , Cell Line , Cell Lineage/genetics , Clone Cells , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Gene Expression Regulation, Developmental , Mice, Inbred ICR , Models, Biological , Molecular Sequence Data , Up-Regulation/genetics
20.
Proc Natl Acad Sci U S A ; 111(20): 7325-30, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24799717

ABSTRACT

Mature oocyte cytoplasm can reprogram somatic cell nuclei to the pluripotent state through a series of sequential events including protein exchange between the donor nucleus and ooplasm, chromatin remodeling, and pluripotency gene reactivation. Maternal factors that are responsible for this reprogramming process remain largely unidentified. Here, we demonstrate that knockdown of histone variant H3.3 in mouse oocytes results in compromised reprogramming and down-regulation of key pluripotency genes; and this compromised reprogramming for developmental potentials and transcription of pluripotency genes can be rescued by injecting exogenous H3.3 mRNA, but not H3.2 mRNA, into oocytes in somatic cell nuclear transfer embryos. We show that maternal H3.3, and not H3.3 in the donor nucleus, is essential for successful reprogramming of somatic cell nucleus into the pluripotent state. Furthermore, H3.3 is involved in this reprogramming process by remodeling the donor nuclear chromatin through replacement of donor nucleus-derived H3 with de novo synthesized maternal H3.3 protein. Our study shows that H3.3 is a crucial maternal factor for oocyte reprogramming and provides a practical model to directly dissect the oocyte for its reprogramming capacity.


Subject(s)
Cell Nucleus/metabolism , Cellular Reprogramming , Gene Expression Regulation, Developmental , Histones/chemistry , Oocytes/cytology , Animals , Chromatin/metabolism , Cytoplasm/metabolism , Female , Mice , Nuclear Transfer Techniques , Oocytes/metabolism , RNA, Small Interfering/metabolism , Sequence Analysis, RNA
SELECTION OF CITATIONS
SEARCH DETAIL