Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 151
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 186(15): 3166-3181.e18, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37413989

ABSTRACT

Proper preimplantation development is essential to assemble a blastocyst capable of implantation. Live imaging has uncovered major events driving early development in mouse embryos; yet, studies in humans have been limited by restrictions on genetic manipulation and lack of imaging approaches. We have overcome this barrier by combining fluorescent dyes with live imaging to reveal the dynamics of chromosome segregation, compaction, polarization, blastocyst formation, and hatching in the human embryo. We also show that blastocyst expansion mechanically constrains trophectoderm cells, causing nuclear budding and DNA shedding into the cytoplasm. Furthermore, cells with lower perinuclear keratin levels are more prone to undergo DNA loss. Moreover, applying trophectoderm biopsy, a mechanical procedure performed clinically for genetic testing, increases DNA shedding. Thus, our work reveals distinct processes underlying human development compared with mouse and suggests that aneuploidies in human embryos may not only originate from chromosome segregation errors during mitosis but also from nuclear DNA shedding.


Subject(s)
Preimplantation Diagnosis , Pregnancy , Female , Humans , Animals , Mice , Preimplantation Diagnosis/methods , Blastocyst , Embryo Implantation , Genetic Testing/methods , Aneuploidy , Biopsy/methods
2.
Mol Cell ; 82(19): 3613-3631.e7, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36108632

ABSTRACT

Allele-specific expression of imprinted gene clusters is governed by gametic DNA methylation at master regulators called imprinting control regions (ICRs). Non-gametic or secondary differentially methylated regions (DMRs) at promoters and exonic regions reinforce monoallelic expression but do not control an entire cluster. Here, we unveil an unconventional secondary DMR that is indispensable for tissue-specific imprinting of two previously unlinked genes, Grb10 and Ddc. Using polymorphic mice, we mapped an intronic secondary DMR at Grb10 with paternal-specific CTCF binding (CBR2.3) that forms contacts with Ddc. Deletion of paternal CBR2.3 removed a critical insulator, resulting in substantial shifting of chromatin looping and ectopic enhancer-promoter contacts. Destabilized gene architecture precipitated abnormal Grb10-Ddc expression with developmental consequences in the heart and muscle. Thus, we redefine the Grb10-Ddc imprinting domain by uncovering an unconventional intronic secondary DMR that functions as an insulator to instruct the tissue-specific, monoallelic expression of multiple genes-a feature previously ICR exclusive.


Subject(s)
Genomic Imprinting , RNA, Long Noncoding , Alleles , Animals , Chromatin/genetics , DNA Methylation , GRB10 Adaptor Protein/genetics , Heart , Mice
3.
Genes Dev ; 36(11-12): 752-763, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35738678

ABSTRACT

Self-renewal of spermatogonial stem cells is vital to lifelong production of male gametes and thus fertility. However, the underlying mechanisms remain enigmatic. Here, we show that DOT1L, the sole H3K79 methyltransferase, is required for spermatogonial stem cell self-renewal. Mice lacking DOT1L fail to maintain spermatogonial stem cells, characterized by a sequential loss of germ cells from spermatogonia to spermatids and ultimately a Sertoli cell only syndrome. Inhibition of DOT1L reduces the stem cell activity after transplantation. DOT1L promotes expression of the fate-determining HoxC transcription factors in spermatogonial stem cells. Furthermore, H3K79me2 accumulates at HoxC9 and HoxC10 genes. Our findings identify an essential function for DOT1L in adult stem cells and provide an epigenetic paradigm for regulation of spermatogonial stem cells.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Spermatogonia , Stem Cells , Animals , Cell Differentiation , Male , Mice , Spermatogonia/cytology , Spermatogonia/metabolism , Stem Cells/cytology , Stem Cells/metabolism
4.
Mol Cell ; 81(4): 859-869.e8, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33352108

ABSTRACT

Active DNA demethylation via ten-eleven translocation (TET) family enzymes is essential for epigenetic reprogramming in cell state transitions. TET enzymes catalyze up to three successive oxidations of 5-methylcytosine (5mC), generating 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), or 5-carboxycytosine (5caC). Although these bases are known to contribute to distinct demethylation pathways, the lack of tools to uncouple these sequential oxidative events has constrained our mechanistic understanding of the role of TETs in chromatin reprogramming. Here, we describe the first application of biochemically engineered TET mutants that unlink 5mC oxidation steps, examining their effects on somatic cell reprogramming. We show that only TET enzymes proficient for oxidation to 5fC/5caC can rescue the reprogramming potential of Tet2-deficient mouse embryonic fibroblasts. This effect correlated with rapid DNA demethylation at reprogramming enhancers and increased chromatin accessibility later in reprogramming. These experiments demonstrate that DNA demethylation through 5fC/5caC has roles distinct from 5hmC in somatic reprogramming to pluripotency.


Subject(s)
5-Methylcytosine/metabolism , Cellular Reprogramming , DNA-Binding Proteins/metabolism , Embryo, Mammalian/metabolism , Enhancer Elements, Genetic , Epigenesis, Genetic , Fibroblasts/metabolism , Proto-Oncogene Proteins/metabolism , Animals , DNA-Binding Proteins/genetics , Dioxygenases , Embryo, Mammalian/cytology , Fibroblasts/cytology , HEK293 Cells , Humans , Mice , Mice, Knockout , Mutation , NIH 3T3 Cells , Proto-Oncogene Proteins/genetics
5.
Cell ; 152(6): 1308-23, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23498939

ABSTRACT

X chromosome inactivation and genomic imprinting are classic epigenetic processes that cause disease when not appropriately regulated in mammals. Whereas X chromosome inactivation evolved to solve the problem of gene dosage, the purpose of genomic imprinting remains controversial. Nevertheless, the two phenomena are united by allelic control of large gene clusters, such that only one copy of a gene is expressed in every cell. Allelic regulation poses significant challenges because it requires coordinated long-range control in cis and stable propagation over time. Long noncoding RNAs have emerged as a common theme, and their contributions to diseases of imprinting and the X chromosome have become apparent. Here, we review recent advances in basic biology, the connections to disease, and preview potential therapeutic strategies for future development.


Subject(s)
Disease/genetics , Genomic Imprinting , RNA, Long Noncoding/metabolism , X Chromosome Inactivation , Animals , Humans
6.
Genes Dev ; 34(11-12): 745-750, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32381626

ABSTRACT

DNA methylation is a major silencing mechanism of transposable elements (TEs). Here we report that TEX15, a testis-specific protein, is required for TE silencing. TEX15 is expressed in embryonic germ cells and functions during genome-wide epigenetic reprogramming. The Tex15 mutant exhibits DNA hypomethylation in TEs at a level similar to Mili and Dnmt3c but not Miwi2 mutants. TEX15 is associated with MILI in testis. As loss of Tex15 causes TE desilencing with intact piRNA production, our results identify TEX15 as a new essential epigenetic regulator that may function as a nuclear effector of MILI to silence TEs by DNA methylation.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA Transposable Elements/genetics , Gene Silencing/physiology , Germ Cells/metabolism , Animals , DNA Methylation , Embryonic Germ Cells/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Developmental/genetics , Male , Mice , Mutation
7.
Genes Dev ; 33(1-2): 1-3, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30602436

ABSTRACT

The monoallelic parent of origin-specific expression of imprinted genes in mammals is regulated by differentially DNA methylated imprinting control regions (ICRs). In contrast to most of the genome, ICRs must maintain their DNA methylation and parental identity despite extensive epigenetic reprogramming that takes place after fertilization. Previous work demonstrated that the Krüppel-associated box (KRAB)-containing zinc finger protein (KZFP) ZFP57 protects select ICRs from demethylation and preserves parental identity. However, some loci are unaffected in Zfp57-null mice. Thus, it has been speculated that other KZFPs must be involved in this process. Takahashi and colleagues (pp. 49-54) now report one such KZFP: ZFP445.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genomic Imprinting/genetics , Zinc Fingers , Animals , DNA Methylation , Epigenomics , Humans , Mice
8.
Cell ; 146(1): 11-3, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21729777

ABSTRACT

Xist RNA inactivates one mammalian X chromosome (the Xi) by associating with it in cis. The mechanism of this interaction is unresolved. Jeon and Lee (2011) now show that YY1 binds both Xist RNA and DNA, thereby providing a mechanism to anchor Xist to the Xi and facilitate X chromosome inactivation.

9.
Cell ; 146(1): 67-79, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21722948

ABSTRACT

DNA methylation is a major epigenetic mechanism for gene silencing. Whereas methyltransferases mediate cytosine methylation, it is less clear how unmethylated regions in mammalian genomes are protected from de novo methylation and whether an active demethylating activity is involved. Here, we show that either knockout or catalytic inactivation of the DNA repair enzyme thymine DNA glycosylase (TDG) leads to embryonic lethality in mice. TDG is necessary for recruiting p300 to retinoic acid (RA)-regulated promoters, protection of CpG islands from hypermethylation, and active demethylation of tissue-specific developmentally and hormonally regulated promoters and enhancers. TDG interacts with the deaminase AID and the damage response protein GADD45a. These findings highlight a dual role for TDG in promoting proper epigenetic states during development and suggest a two-step mechanism for DNA demethylation in mammals, whereby 5-methylcytosine and 5-hydroxymethylcytosine are first deaminated by AID to thymine and 5-hydroxymethyluracil, respectively, followed by TDG-mediated thymine and 5-hydroxymethyluracil excision repair.


Subject(s)
DNA Methylation , Embryonic Development , Gene Expression Regulation, Developmental , Thymine DNA Glycosylase/metabolism , 5-Methylcytosine/metabolism , Animals , Cell Cycle Proteins/metabolism , Cytidine Deaminase/metabolism , Cytosine/analogs & derivatives , Cytosine/metabolism , Female , Gene Knock-In Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Thymine DNA Glycosylase/genetics , Transcription, Genetic
10.
Nucleic Acids Res ; 52(7): e38, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38407446

ABSTRACT

The Infinium BeadChip is the most widely used DNA methylome assay technology for population-scale epigenome profiling. However, the standard workflow requires over 200 ng of input DNA, hindering its application to small cell-number samples, such as primordial germ cells. We developed experimental and analysis workflows to extend this technology to suboptimal input DNA conditions, including ultra-low input down to single cells. DNA preamplification significantly enhanced detection rates to over 50% in five-cell samples and ∼25% in single cells. Enzymatic conversion also substantially improved data quality. Computationally, we developed a method to model the background signal's influence on the DNA methylation level readings. The modified detection P-value calculation achieved higher sensitivities for low-input datasets and was validated in over 100 000 public diverse methylome profiles. We employed the optimized workflow to query the demethylation dynamics in mouse primordial germ cells available at low cell numbers. Our data revealed nuanced chromatin states, sex disparities, and the role of DNA methylation in transposable element regulation during germ cell development. Collectively, we present comprehensive experimental and computational solutions to extend this widely used methylation assay technology to applications with limited DNA.


Subject(s)
DNA Methylation , Single-Cell Analysis , Animals , Female , Humans , Male , Mice , CpG Islands , DNA/genetics , DNA/metabolism , Epigenomics/methods , Germ Cells/metabolism , Oligonucleotide Array Sequence Analysis/methods , Single-Cell Analysis/methods
12.
Hum Mol Genet ; 31(22): 3855-3872, 2022 11 10.
Article in English | MEDLINE | ID: mdl-35717573

ABSTRACT

In vitro fertilization (IVF) is associated with DNA methylation abnormalities and a higher incidence of adverse pregnancy outcomes. However, which exposure(s), among the many IVF interventions, contributes to these outcomes remains unknown. Frozen embryo transfer (ET) is increasingly utilized as an alternative to fresh ET, but reports suggest a higher incidence of pre-eclampsia and large for gestational age infants. This study examines DNA methylation in human placentas using the 850K Infinium MethylationEPIC BeadChip array obtained after 65 programmed frozen ET cycles, 82 fresh ET cycles and 45 unassisted conceptions. Nine patients provided placentas following frozen and fresh ET from consecutive pregnancies for a paired subgroup analysis. In parallel, eight mouse placentas from fresh and frozen ET were analyzed using the Infinium Mouse Methylation BeadChip array. Human and mouse placentas were significantly hypermethylated after frozen ET compared with fresh. Paired analysis showed similar trends. Sex-specific analysis revealed that these changes were driven by male placentas in humans and mice. Frozen and fresh ET placentas were significantly different from controls, with frozen samples hypermethylated compared with controls driven by males and fresh samples being hypomethylated compared with controls, driven by females. Sexually dimorphic epigenetic changes could indicate differential susceptibility to IVF-associated perturbations, which highlights the importance of sex-specific evaluation of adverse outcomes. Similarities between changes in mice and humans underscore the suitability of the mouse model in evaluating how IVF impacts the epigenetic landscape, which is valuable given limited access to human tissue and the ability to isolate specific interventions in mice.


Subject(s)
DNA Methylation , Embryo Transfer , Pregnancy , Female , Humans , Male , Mice , Animals , DNA Methylation/genetics , Embryo Transfer/adverse effects , Cryopreservation , Fertilization in Vitro/adverse effects , Placenta , Retrospective Studies
13.
Hum Reprod ; 39(1): 154-176, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37994669

ABSTRACT

STUDY QUESTION: Does trophectoderm biopsy (TEBx) of blastocysts for preimplantation genetic testing in the clinic affect normal placental and embryo development and offspring metabolic outcomes in a mouse model? SUMMARY ANSWER: TEBx impacts placental and embryonic health during early development, with some alterations resolving and others worsening later in development and triggering metabolic changes in adult offspring. WHAT IS KNOWN ALREADY: Previous studies have not assessed the epigenetic and morphological impacts of TEBx either in human populations or in animal models. STUDY DESIGN, SIZE, DURATION: We employed a mouse model to identify the effects of TEBx during IVF. Three groups were assessed: naturally conceived (Naturals), IVF, and IVF + TEBx, at two developmental timepoints: embryonic day (E)12.5 (n = 40/Naturals, n = 36/IVF, and n = 36/IVF + TEBx) and E18.5 (n = 42/Naturals, n = 30/IVF, and n = 35/IVF + TEBx). Additionally, to mimic clinical practice, we assessed a fourth group: IVF + TEBx + Vitrification (Vit) at E12.5 (n = 29) that combines TEBx and vitrification. To assess the effect of TEBx in offspring health, we characterized a 12-week-old cohort (n = 24/Naturals, n = 25/IVF and n = 25/IVF + TEBx). PARTICIPANTS/MATERIALS, SETTING, METHODS: Our mouse model used CF-1 females as egg donors and SJL/B6 males as sperm donors. IVF, TEBx, and vitrification were performed using standardized methods. Placenta morphology was evaluated by hematoxylin-eosin staining, in situ hybridization using Tpbpa as a junctional zone marker and immunohistochemistry using CD34 fetal endothelial cell markers. For molecular analysis of placentas and embryos, DNA methylation was analyzed using pyrosequencing, luminometric methylation assay, and chip array technology. Expression patterns were ascertained by RNA sequencing. Triglycerides, total cholesterol, high-, low-, and very low-density lipoprotein, insulin, and glucose were determined in the 12-week-old cohort using commercially available kits. MAIN RESULTS AND THE ROLE OF CHANCE: We observed that at E12.5, IVF + TEBx had a worse outcome in terms of changes in DNA methylation and differential gene expression in placentas and whole embryos compared with IVF alone and compared with Naturals. These changes were reflected in alterations in placental morphology and blood vessel density. At E18.5, early molecular changes in fetuses were maintained or exacerbated. With respect to placentas, the molecular and morphological changes, although different compared to Naturals, were equivalent to the IVF group, except for changes in blood vessel density, which persisted. Of note is that most differences were sex specific. We conclude that TEBx has more detrimental effects in mid-gestation placental and embryonic tissues, with alterations in embryonic tissues persisting or worsening in later developmental stages compared to IVF alone, and the addition of vitrification after TEBx results in more pronounced and potentially detrimental epigenetic effects: these changes are significantly different compared to Naturals. Finally, we observed that 12-week IVF + TEBx offspring, regardless of sex, showed higher glucose, insulin, triglycerides, lower total cholesterol, and lower high-density lipoprotein compared to IVF and Naturals, with only males having higher body weight compared to IVF and Naturals. Our findings in a mouse model additionally support the need for more studies to assess the impact of new procedures in ART to ensure healthy pregnancies and offspring outcomes. LARGE SCALE DATA: Data reported in this work have been deposited in the NCBI Gene Expression Omnibus under accession number GSE225318. LIMITATIONS, REASONS FOR CAUTION: This study was performed using a mouse model that mimics many clinical IVF procedures and outcomes observed in humans, where studies on early embryos are not possible. WIDER IMPLICATIONS OF THE FINDINGS: This study highlights the importance of assaying new procedures used in ART to assess their impact on placenta and embryo development, and offspring metabolic outcomes. STUDY FUNDING/COMPETING INTEREST(S): This work was funded by a National Centers for Translational Research in Reproduction and Infertility grant P50 HD068157-06A1 (M.S.B., C.C., M.M.), Ruth L. Kirschstein National Service Award Individual Postdoctoral Fellowship F32 HD107914 (E.A.R.-C.) and F32 HD089623 (L.A.V.), and National Institutes of Health Training program in Cell and Molecular Biology T32 GM007229 (C.N.H.). No conflict of interest.


Subject(s)
Insulins , Placenta , Adult , Animals , Pregnancy , Humans , Male , Female , Placenta/metabolism , Semen/metabolism , Blastocyst/metabolism , Fertilization in Vitro , Epigenesis, Genetic , Biopsy , Glucose , Triglycerides , Cholesterol , Insulins/metabolism
14.
Genes Dev ; 30(5): 567-78, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26944681

ABSTRACT

Imprinting is a classic mammalian epigenetic phenomenon that results in expression from a single parental allele. Imprinting defects can lead to inappropriate expression from the normally silenced allele, but it remains unclear whether every cell in a mutant organism follows the population average, which would have profound implications for human imprinting disorders. Here, we apply a new fluorescence in situ hybridization method that measures allele-specific expression in single cells to address this question in mutants exhibiting aberrant H19/Igf2 (insulin-like growth factor 2) imprinting. We show that mutant primary embryonic mouse fibroblasts are comprised of two subpopulations: one expressing both H19 alleles and another expressing only the maternal copy. Only in the latter cell population is Igf2 expression detected. Furthermore, the two subpopulations are stable in that cells do not interconvert between the two expression patterns. Combined small input methylation analysis and transcriptional imaging revealed that these two mutant subpopulations exhibit distinct methylation patterns at their imprinting control regions. Consistently, pharmacological inhibition of DNA methylation reduced the proportion of monoallelic cells. Importantly, we observed that the same two subpopulations are also present in vivo within murine cardiac tissue. Our results establish that imprinting disorders can display striking single-cell heterogeneity in their molecular phenotypes and suggest that such heterogeneity may underlie epigenetic mosaicism in human imprinting disorders.


Subject(s)
Alleles , Epigenomics , Gene Expression Regulation , Genomic Imprinting/genetics , Insulin-Like Growth Factor II/genetics , Mosaicism , RNA, Long Noncoding/genetics , Animals , Cells, Cultured , DNA Methylation , In Situ Hybridization, Fluorescence , Mice , Mice, Inbred C57BL , Mutation , Single-Cell Analysis
15.
Hum Mol Genet ; 30(16): 1509-1520, 2021 07 28.
Article in English | MEDLINE | ID: mdl-34132339

ABSTRACT

The reciprocal parent of origin-specific expression of H19 and IGF2 is controlled by the H19/IGF2:IG-DMR (IC1), whose maternal allele is unmethylated and acts as a CTCF-dependent insulator. In humans, internal IC1 deletions are associated with Beckwith-Wiedemann syndrome (BWS) and Silver-Russell syndrome (SRS), depending on their parental origin. These genetic mutations result in aberrant DNA methylation, deregulation of IGF2/H19 and disease with incomplete penetrance. However, the mechanism linking the microdeletions to altered molecular and clinical phenotypes remains unclear. To address this issue, we have previously generated and characterized two knock-in mouse lines with the human wild-type (hIC1wt) or mutant (hIC1∆2.2) IC1 allele replacing the endogenous mouse IC1 (mIC1). Here, we report an additional knock-in line carrying a mutant hIC1 allele with an internal 1.8 kb deletion (hIC1∆1.8). The phenotype of these mice is different from that of the hIC1∆2.2-carrying mice, partially resembling hIC1wt animals. Indeed, proper H19 and Igf2 imprinting and normal growth phenotype were evident in the mice with maternal transmission of hIC1Δ1.8, while low DNA methylation and non-viable phenotype characterize its paternal transmission. In contrast to hIC1wt, E15.5 embryos that paternally inherit hIC1Δ1.8 displayed variegated hIC1 methylation. In addition, increased Igf2 expression, correlating with increased body weight, was found in one third of these mice. Chromatin immunoprecipitation experiments in mouse embryonic stem cells carrying the three different hIC1 alleles demonstrate that the number of CTCF target sites influences its binding to hIC1, indicating that in the mouse, CTCF binding is key to determining hIC1 methylation and Igf2 expression.


Subject(s)
Beckwith-Wiedemann Syndrome , RNA, Long Noncoding , Animals , Beckwith-Wiedemann Syndrome/genetics , Binding Sites , CCCTC-Binding Factor/genetics , DNA Methylation/genetics , Genomic Imprinting , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Mice , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
16.
Development ; 147(11)2020 05 29.
Article in English | MEDLINE | ID: mdl-32471820

ABSTRACT

Although widely used, assisted reproductive technologies (ARTs) are associated with adverse perinatal outcomes. To elucidate their underlying causes, we have conducted a longitudinal analysis of placental development and fetal growth using a mouse model to investigate the effects of individual ART procedures: hormone stimulation, in vitro fertilization (IVF), embryo culture and embryo transfer. We demonstrate that transfer of blastocysts naturally conceived without hormone stimulation and developed in vivo prior to transfer can impair early placentation and fetal growth, but this effect normalizes by term. In contrast, embryos cultured in vitro before transfer do not exhibit this compensation but rather display placental overgrowth, reduced fetal weight, reduced placental DNA methylation and increased levels of sFLT1, an anti-angiogenic protein implicated in causing the maternal symptoms of preeclampsia in humans. Increases in sFLT1 observed in this study suggest that IVF procedures could increase the risk for preeclampsia. Moreover, our results indicate that embryo culture is the major factor contributing to most placental abnormalities and should therefore be targeted for optimization.


Subject(s)
Placenta/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , DNA Methylation , Embryo Transfer , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Development , Female , Fertilization in Vitro , Male , Mice , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Pre-Eclampsia/veterinary , Pregnancy , Risk , Symporters/genetics , Symporters/metabolism , Trophoblasts/cytology , Trophoblasts/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics
17.
Hum Mol Genet ; 29(R1): R107-R116, 2020 09 30.
Article in English | MEDLINE | ID: mdl-32592473

ABSTRACT

Temple syndrome (TS) and Kagami-Ogata syndrome (KOS) are imprinting disorders caused by absence or overexpression of genes within a single imprinted cluster on human chromosome 14q32. TS most frequently arises from maternal UPD14 or epimutations/deletions on the paternal chromosome, whereas KOS most frequently arises from paternal UPD14 or epimutations/deletions on the maternal chromosome. In this review, we describe the clinical symptoms and genetic/epigenetic features of this imprinted region. The locus encompasses paternally expressed protein-coding genes (DLK1, RTL1 and DIO3) and maternally expressed lncRNAs (MEG3/GTL2, RTL1as and MEG8), as well as numerous miRNAs and snoRNAs. Control of expression is complex, with three differentially methylated regions regulating germline, placental and tissue-specific transcription. The strong conserved synteny between mouse chromosome 12aF1 and human chromosome 14q32 has enabled the use of mouse models to elucidate imprinting mechanisms and decipher the contribution of genes to the symptoms of TS and KOS. In this review, we describe relevant mouse models and highlight their value to better inform treatment options for long-term management of TS and KOS patients.


Subject(s)
Abnormalities, Multiple , Chromosome Disorders/pathology , Chromosomes, Human, Pair 14/genetics , Disease Models, Animal , Genomic Imprinting , Hallux/abnormalities , Intellectual Disability/pathology , Nails, Malformed/pathology , Thumb/abnormalities , Uniparental Disomy/pathology , Animals , Chromosome Disorders/genetics , Hallux/pathology , Humans , Intellectual Disability/genetics , Mice , Nails, Malformed/genetics , Phenotype , Thumb/pathology , Uniparental Disomy/genetics
18.
FASEB J ; 35(4): e21523, 2021 04.
Article in English | MEDLINE | ID: mdl-33734487

ABSTRACT

Although in vitro fertilization (IVF) is associated with adverse perinatal outcomes, there is increasing concern about the long-term and sex-specific health implications. Augmenting our IVF mouse model to longitudinally investigate metabolic outcomes in offspring from optimal neonatal litter sizes, we found sex-specific metabolic outcomes in IVF offspring. IVF-conceived females had higher body weight and cholesterol levels compared to naturally conceived females, whereas IVF-conceived males had higher levels of triglycerides and insulin, and increased body fat composition. Through adult liver transcriptomics and proteomics, we identified sexually dimorphic dysregulation of the sterol regulatory element-binding protein (SREBP) pathways that are associated with the sex-specific phenotypes. We also found that global loss of DNA methylation in placenta was linked to higher cholesterol levels in IVF-conceived females. Our findings indicate that IVF procedures have long-lasting sex-specific effects on metabolic health of offspring and lay the foundation to utilize the placenta as a predictor of long-term outcomes.


Subject(s)
Fertilization/physiology , Proteome/metabolism , Sex Factors , Transcriptome/physiology , Animals , Body Composition/physiology , DNA Methylation/physiology , Female , Liver/metabolism , Mice , Placenta/metabolism , Pregnancy
19.
PLoS Genet ; 15(1): e1007874, 2019 01.
Article in English | MEDLINE | ID: mdl-30625149

ABSTRACT

Extensive cell-to-cell variation exists even among putatively identical cells, and there is great interest in understanding how the properties of transcription relate to this heterogeneity. Differential expression from the two gene copies in diploid cells could potentially contribute, yet our ability to measure from which gene copy individual RNAs originated remains limited, particularly in the context of tissues. Here, we demonstrate quantitative, single molecule allele-specific RNA FISH adapted for use on tissue sections, allowing us to determine the chromosome of origin of individual RNA molecules in formaldehyde-fixed tissues. We used this method to visualize the allele-specific expression of Xist and multiple autosomal genes in mouse kidney. By combining these data with mathematical modeling, we evaluated models for allele-specific heterogeneity, in particular demonstrating that apparent expression from only one of the alleles in single cells can arise as a consequence of low-level mRNA abundance and transcriptional bursting.


Subject(s)
Allelic Imbalance/genetics , In Situ Hybridization, Fluorescence/methods , Kidney/metabolism , RNA, Long Noncoding/genetics , Alleles , Animals , Gene Expression Regulation, Developmental/genetics , Mice , Organ Specificity , RNA, Long Noncoding/isolation & purification
20.
Development ; 145(5)2018 03 14.
Article in English | MEDLINE | ID: mdl-29540437

ABSTRACT

Anne Ferguson-Smith and Marisa Bartolomei look back at the life and science of Denise Barlow, a pioneer in genomic imprinting and epigenetics.


Subject(s)
Developmental Biology/history , Epigenomics/history , Genomic Imprinting/physiology , Laboratory Personnel , Animals , Austria , Epigenesis, Genetic , History, 20th Century , History, 21st Century , Humans , Laboratory Personnel/history , United Kingdom
SELECTION OF CITATIONS
SEARCH DETAIL