Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.231
Filter
3.
Elife ; 132024 May 30.
Article in English | MEDLINE | ID: mdl-38813868

ABSTRACT

Germline epigenetic programming, including genomic imprinting, substantially influences offspring development. Polycomb Repressive Complex 2 (PRC2) plays an important role in Histone 3 Lysine 27 trimethylation (H3K27me3)-dependent imprinting, loss of which leads to growth and developmental changes in mouse offspring. In this study, we show that offspring from mouse oocytes lacking the PRC2 protein Embryonic Ectoderm Development (EED) were initially developmentally delayed, characterised by low blastocyst cell counts and substantial growth delay in mid-gestation embryos. This initial developmental delay was resolved as offspring underwent accelerated fetal development and growth in late gestation resulting in offspring that were similar stage and weight to controls at birth. The accelerated development and growth in offspring from Eed-null oocytes was associated with remodelling of the placenta, which involved an increase in fetal and maternal tissue size, conspicuous expansion of the glycogen-enriched cell population, and delayed parturition. Despite placental remodelling and accelerated offspring fetal growth and development, placental efficiency, and fetal blood glucose levels were low, and the fetal blood metabolome was unchanged. Moreover, while expression of the H3K27me3-imprinted gene and amino acid transporter Slc38a4 was increased, fetal blood levels of individual amino acids were similar to controls, indicating that placental amino acid transport was not enhanced. Genome-wide analyses identified extensive transcriptional dysregulation and DNA methylation changes in affected placentas, including a range of imprinted and non-imprinted genes. Together, while deletion of Eed in growing oocytes resulted in fetal growth and developmental delay and placental hyperplasia, our data indicate a remarkable capacity for offspring fetal growth to be normalised despite inefficient placental function and the loss of H3K27me3-dependent genomic imprinting.


Subject(s)
Genomic Imprinting , Animals , Female , Pregnancy , Mice , Polycomb Repressive Complex 2/metabolism , Polycomb Repressive Complex 2/genetics , Fetal Development/genetics , Placenta/metabolism , Oocytes/metabolism , Oocytes/growth & development , Amino Acid Transport System A
4.
BMC Biol ; 22(1): 127, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816743

ABSTRACT

BACKGROUND: Optimal size at birth dictates perinatal survival and long-term risk of developing common disorders such as obesity, type 2 diabetes and cardiovascular disease. The imprinted Grb10 gene encodes a signalling adaptor protein capable of inhibiting receptor tyrosine kinases, including the insulin receptor (Insr) and insulin-like growth factor type 1 receptor (Igf1r). Grb10 restricts fetal growth such that Grb10 knockout (KO) mice are at birth some 25-35% larger than wild type. Using a mouse genetic approach, we test the widely held assumption that Grb10 influences growth through interaction with Igf1r, which has a highly conserved growth promoting role. RESULTS: Should Grb10 interact with Igf1r to regulate growth Grb10:Igf1r double mutant mice should be indistinguishable from Igf1r KO single mutants, which are around half normal size at birth. Instead, Grb10:Igf1r double mutants were intermediate in size between Grb10 KO and Igf1r KO single mutants, indicating additive effects of the two signalling proteins having opposite actions in separate pathways. Some organs examined followed a similar pattern, though Grb10 KO neonates exhibited sparing of the brain and kidneys, whereas the influence of Igf1r extended to all organs. An interaction between Grb10 and Insr was similarly investigated. While there was no general evidence for a major interaction for fetal growth regulation, the liver was an exception. The liver in Grb10 KO mutants was disproportionately overgrown with evidence of excess lipid storage in hepatocytes, whereas Grb10:Insr double mutants were indistinguishable from Insr single mutants or wild types. CONCLUSIONS: Grb10 acts largely independently of Igf1r or Insr to control fetal growth and has a more variable influence on individual organs. Only the disproportionate overgrowth and excess lipid storage seen in the Grb10 KO neonatal liver can be explained through an interaction between Grb10 and the Insr. Our findings are important for understanding how positive and negative influences on fetal growth dictate size and tissue proportions at birth.


Subject(s)
Fetal Development , GRB10 Adaptor Protein , Mice, Knockout , Receptor, IGF Type 1 , Receptor, Insulin , Animals , GRB10 Adaptor Protein/genetics , GRB10 Adaptor Protein/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Mice , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Fetal Development/genetics , Genomic Imprinting , Female , Male , Insulin-Like Peptides
5.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(6): 685-695, 2024 Jun 10.
Article in Chinese | MEDLINE | ID: mdl-38818552

ABSTRACT

Uniparental disomy (UPD)-related imprinting disorders are a group of congenital disorders which can lead to severe birth defects. Their molecular etiology is the occurrence of UPD in the genomic imprinting regions, which may cause disturbed expression of parent-of-origin imprinted genes. With the widespread applications of genetic testing techniques, the prenatal diagnosis of UPD-related imprinted diseases has gradually become clinical routines. However, due to the complicated pathogenesis of such disorders, currently there is still a lack of standards and norms for the understanding, diagnosis, management and genetic counseling. By referring to the relevant guidelines and consensus, the latest progress of research, and opinions from experts in the relevant fields, the writing group has formulated a consensus over the prenatal diagnosis and genetic counseling for UPD-related imprinting disorders, with an aim to provide a more accurate and rational evaluation in prenatal clinics.


Subject(s)
Genetic Counseling , Genomic Imprinting , Prenatal Diagnosis , Uniparental Disomy , Humans , Uniparental Disomy/genetics , Uniparental Disomy/diagnosis , Pregnancy , Female , Consensus , Genetic Testing/methods , Imprinting Disorders
6.
Mol Cell ; 84(10): 1842-1854.e7, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38759624

ABSTRACT

Genomic context critically modulates regulatory function but is difficult to manipulate systematically. The murine insulin-like growth factor 2 (Igf2)/H19 locus is a paradigmatic model of enhancer selectivity, whereby CTCF occupancy at an imprinting control region directs downstream enhancers to activate either H19 or Igf2. We used synthetic regulatory genomics to repeatedly replace the native locus with 157-kb payloads, and we systematically dissected its architecture. Enhancer deletion and ectopic delivery revealed previously uncharacterized long-range regulatory dependencies at the native locus. Exchanging the H19 enhancer cluster with the Sox2 locus control region (LCR) showed that the H19 enhancers relied on their native surroundings while the Sox2 LCR functioned autonomously. Analysis of regulatory DNA actuation across cell types revealed that these enhancer clusters typify broader classes of context sensitivity genome wide. These results show that unexpected dependencies influence even well-studied loci, and our approach permits large-scale manipulation of complete loci to investigate the relationship between regulatory architecture and function.


Subject(s)
CCCTC-Binding Factor , Enhancer Elements, Genetic , Insulin-Like Growth Factor II , RNA, Long Noncoding , SOXB1 Transcription Factors , Animals , Mice , CCCTC-Binding Factor/metabolism , CCCTC-Binding Factor/genetics , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Locus Control Region/genetics , Genomic Imprinting , Genomics/methods
7.
Genes (Basel) ; 15(5)2024 May 18.
Article in English | MEDLINE | ID: mdl-38790270

ABSTRACT

BACKGROUND: Diagnosing imprinting defects in neonates and young children presents challenges, often necessitating molecular analysis for a conclusive diagnosis. The isolation of genetic material from oral swabs becomes crucial, especially in settings where blood sample collection is impractical or for vulnerable populations like newborns, who possess limited blood volumes and are often too fragile for invasive procedures. Oral swab samples emerge as an excellent source of DNA, effectively overcoming obstacles associated with rare diseases. METHODS: In our study, we specifically addressed the determination of the quality and quantity of DNA extracted from oral swab samples using NaCl procedures. RESULTS: We compared these results with extractions performed using a commercial kit. Subsequently, the obtained material underwent MS-HRM analysis for loci associated with imprinting diseases such as Prader-Willi and Angelman syndromes. CONCLUSIONS: Our study emphasizes the significance of oral swab samples as a reliable source for obtaining DNA for MS-HRM analysis. NaCl extraction stands out as a practical and cost-effective method for genetic studies, contributing to a molecular diagnosis that proves particularly beneficial for patients facing delays in characterization, ultimately influencing their treatment.


Subject(s)
Angelman Syndrome , DNA , Genomic Imprinting , Mouth Mucosa , Prader-Willi Syndrome , Humans , Mouth Mucosa/cytology , Mouth Mucosa/pathology , Angelman Syndrome/genetics , Angelman Syndrome/diagnosis , Prader-Willi Syndrome/genetics , Prader-Willi Syndrome/diagnosis , DNA/genetics , DNA/isolation & purification , Sodium Chloride , Infant, Newborn , Male , Imprinting Disorders
8.
Clin Epigenetics ; 16(1): 62, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715103

ABSTRACT

BACKGROUND: Temple syndrome (TS14) is a rare imprinting disorder caused by maternal UPD14, imprinting defects or paternal microdeletions which lead to an increase in the maternal expressed genes and a silencing the paternally expressed genes in the 14q32 imprinted domain. Classical TS14 phenotypic features include pre- and postnatal short stature, small hands and feet, muscular hypotonia, motor delay, feeding difficulties, weight gain, premature puberty along and precocious puberty. METHODS: An exon array comparative genomic hybridization was performed on a patient affected by psychomotor and language delay, muscular hypotonia, relative macrocephaly, and small hand and feet at two years old. At 6 years of age, the proband presented with precocious thelarche. Genes dosage and methylation within the 14q32 region were analyzed by MS-MLPA. Bisulfite PCR and pyrosequencing were employed to quantification methylation at the four known imprinted differentially methylated regions (DMR) within the 14q32 domain: DLK1 DMR, IG-DMR, MEG3 DMR and MEG8 DMR. RESULTS: The patient had inherited a 69 Kb deletion, encompassing the entire DLK1 gene, on the paternal allele. Relative hypermethylation of the two maternally methylated intervals, DLK1 and MEG8 DMRs, was observed along with normal methylation level at IG-DMR and MEG3 DMR, resulting in a phenotype consistent with TS14. Additional family members with the deletion showed modest methylation changes at both the DLK1 and MEG8 DMRs consistent with parental transmission. CONCLUSION: We describe a girl with clinical presentation suggestive of Temple syndrome resulting from a small paternal 14q32 deletion that led to DLK1 whole-gene deletion, as well as hypermethylation of the maternally methylated DLK1-DMR.


Subject(s)
Calcium-Binding Proteins , Chromosomes, Human, Pair 14 , DNA Methylation , Genomic Imprinting , Intercellular Signaling Peptides and Proteins , Humans , Calcium-Binding Proteins/genetics , DNA Methylation/genetics , Chromosomes, Human, Pair 14/genetics , Intercellular Signaling Peptides and Proteins/genetics , Genomic Imprinting/genetics , Membrane Proteins/genetics , Child , Male , Comparative Genomic Hybridization/methods , Female , Chromosome Deletion , Child, Preschool , Phenotype , Abnormalities, Multiple/genetics , Imprinting Disorders , Muscle Hypotonia , Facies
9.
Genome Biol ; 25(1): 117, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715110

ABSTRACT

BACKGROUND: Preeclampsia, one of the most lethal pregnancy-related diseases, is associated with the disruption of uterine spiral artery remodeling during placentation. However, the early molecular events leading to preeclampsia remain unknown. RESULTS: By analyzing placentas from preeclampsia, non-preeclampsia, and twin pregnancies with selective intrauterine growth restriction, we show that the pathogenesis of preeclampsia is attributed to immature trophoblast and maldeveloped endothelial cells. Delayed epigenetic reprogramming during early extraembryonic tissue development leads to generation of excessive immature trophoblast cells. We find reduction of de novo DNA methylation in these trophoblast cells results in selective overexpression of maternally imprinted genes, including the endoretrovirus-derived gene PEG10 (paternally expressed gene 10). PEG10 forms virus-like particles, which are transferred from the trophoblast to the closely proximate endothelial cells. In normal pregnancy, only a low amount of PEG10 is transferred to maternal cells; however, in preeclampsia, excessive PEG10 disrupts maternal vascular development by inhibiting TGF-beta signaling. CONCLUSIONS: Our study reveals the intricate epigenetic mechanisms that regulate trans-generational genetic conflict and ultimately ensure proper maternal-fetal interface formation.


Subject(s)
Pre-Eclampsia , Trophoblasts , Vascular Remodeling , Pre-Eclampsia/genetics , Pregnancy , Female , Humans , Trophoblasts/metabolism , Vascular Remodeling/genetics , Placenta/metabolism , DNA Methylation , Epigenesis, Genetic , Endothelial Cells/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genomic Imprinting , Transforming Growth Factor beta/metabolism , Fetal Growth Retardation/genetics , Placentation/genetics , RNA-Binding Proteins , Apoptosis Regulatory Proteins
10.
Clin Epigenetics ; 16(1): 58, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38658973

ABSTRACT

Alzheimer's disease (AD) prevalence is twice as high in non-Hispanic Blacks (NHBs) as in non-Hispanic Whites (NHWs). The objective of this study was to determine whether aberrant methylation at imprint control regions (ICRs) is associated with AD. Differentially methylated regions (DMRs) were bioinformatically identified from whole-genome bisulfite sequenced DNA derived from brain tissue of 9 AD (5 NHBs and 4 NHWs) and 8 controls (4 NHBs and 4 NHWs). We identified DMRs located within 120 regions defined as candidate ICRs in the human imprintome ( https://genome.ucsc.edu/s/imprintome/hg38.AD.Brain_track ). Eighty-one ICRs were differentially methylated in NHB-AD, and 27 ICRs were differentially methylated in NHW-AD, with two regions common to both populations that are proximal to the inflammasome gene, NLRP1, and a known imprinted gene, MEST/MESTIT1. These findings indicate that early developmental alterations in DNA methylation of regions regulating genomic imprinting may contribute to AD risk and that this epigenetic risk differs between NHBs and NHWs.


Subject(s)
Alzheimer Disease , DNA Methylation , Aged , Aged, 80 and over , Female , Humans , Male , Alzheimer Disease/genetics , Alzheimer Disease/ethnology , Black or African American/genetics , Case-Control Studies , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Genomic Imprinting/genetics , NLR Proteins/genetics , White/genetics
11.
Dev Cell ; 59(8): 1010-1027.e8, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38569549

ABSTRACT

Ten-eleven translocation (TET) enzymes iteratively oxidize 5-methylcytosine (5mC) to generate 5-hydroxymethylcytosine (5hmC), 5-formylcytosine, and 5-carboxylcytosine to facilitate active genome demethylation. Whether these bases are required to promote replication-coupled dilution or activate base excision repair during mammalian germline reprogramming remains unresolved due to the inability to decouple TET activities. Here, we generated two mouse lines expressing catalytically inactive TET1 (Tet1-HxD) and TET1 that stalls oxidation at 5hmC (Tet1-V). Tet1 knockout and catalytic mutant primordial germ cells (PGCs) fail to erase methylation at select imprinting control regions and promoters of meiosis-associated genes, validating the requirement for the iterative oxidation of 5mC for complete germline reprogramming. TET1V and TET1HxD rescue most hypermethylation of Tet1-/- sperm, suggesting the role of TET1 beyond its oxidative capability. We additionally identify a broader class of hypermethylated regions in Tet1 mutant mouse sperm that depend on TET oxidation for reprogramming. Our study demonstrates the link between TET1-mediated germline reprogramming and sperm methylome patterning.


Subject(s)
5-Methylcytosine , 5-Methylcytosine/analogs & derivatives , DNA Methylation , DNA-Binding Proteins , Genomic Imprinting , Oxidation-Reduction , Proto-Oncogene Proteins , Spermatozoa , Animals , Male , Mice , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Spermatozoa/metabolism , 5-Methylcytosine/metabolism , Cellular Reprogramming/genetics , Mice, Knockout , Mice, Inbred C57BL
12.
Chin Clin Oncol ; 13(2): 21, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38644545

ABSTRACT

BACKGROUND: The current preoperative malignancy risk evaluation for thyroid nodules involves stepwise diagnostic modalities including ultrasonography, thyroid function serology and fine-needle aspiration (FNA) cytopathology, respectively. We aimed to substantiate the stepwise contributions of each diagnostic step and additionally investigate the diagnostic significance of quantitative chromogenic imprinted gene in-situ hybridization (QCIGISH)-an adjunctive molecular test based on epigenetic imprinting alterations. METHODS: A total of 114 cytopathologically-diagnosed and histopathologically-confirmed thyroid nodules with complete ultrasonographic and serological examination records were evaluated using QCIGISH in the study. Logistic regression models for thyroid malignancy prediction were developed with the stepwise addition of each diagnostic modality and the contribution of each step evaluated in terms of discrimination performance and goodness-of-fit. RESULTS: From the baseline model using ultrasonography [area under the receiver operating characteristics curve (AUROC): 0.79; 95% confidence interval (CI): 0.71-0.86], significant improvements in thyroid malignancy discrimination were observed with the stepwise addition of thyroid function serology (AUROC: 0.82; 95% CI: 0.74-0.90; P=0.23) and FNA cytopathology (AUROC: 0.88; 95% CI: 0.81-0.94; P=0.02), respectively. The inclusion of QCIGISH as an adjunctive molecular test further advanced the preceding model's diagnostic performance (AUROC: 0.95; 95% CI: 0.91-1.00, P=0.007). CONCLUSIONS: Our study demonstrated the significant stepwise diagnostic contributions of standard clinical assessments in the malignancy risk stratification of thyroid nodules. However, the addition of molecular imprinting detection further enabled a more accurate and definitive preoperative evaluation especially for morphologically indeterminate thyroid nodules and cases with potentially discordant results among standard modalities.


Subject(s)
Genomic Imprinting , Humans , Female , Male , Middle Aged , Adult , Thyroid Neoplasms/genetics , Biopsy, Fine-Needle/methods , Thyroid Nodule/genetics , Aged , Thyroid Gland/pathology
13.
Anim Genet ; 55(3): 452-456, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38594908

ABSTRACT

Genomic imprinting is an epigenetic regulation in mammals in which a small subset of genes is monoallelically expressed dependent on their parental origin. A large imprinted domain, SGCE/PEG10 locus, is located on human chromosome 7q21s and mouse proximal chromosome 6. However, genomic imprinting of bovine SGCE/PEG10 cluster has not been systematically studied. In this study, we investigated allele expression of 14 genes of the SGCE/PEG10 locus in bovine somatic tissues and term placenta using a single nucleotide polymorphism (SNP)-based sequencing method. In addition to SGCE and PEG10, two conserved paternally expressed genes in human and mice, five other genes (TFPI2, GNG11, ASB4, PON1, and PON3) were paternally expressed. Three genes, BET1, COL1A2, and CASD1, exhibited tissue-specific monoallelic expression. CALCR showed monoallelic expression in tissues but biallelic expression in the placenta. Three genes, GNGT1, PPP1R9A, and PON2, showed biallelic expression in cattle. Five differentially methylated regions (DMRs) were found to be associated with the allelic expression of TFPI2, COL1A2, SGCE/PEG10, PON3, and ASB4 genes, respectively. The SGCE/PEG10 DMR is a maternally hypermethylated germline DMR, but TFPI2, COL1A2, PON3, and ASB4 DMRs are secondary DMRs. In summary, we identified five novel bovine imprinted genes (GNG11, BET1, COL1A2, CASD1, and PON1) and four secondary DMRs at the SGCE/PEG10 locus.


Subject(s)
Alleles , DNA Methylation , Genomic Imprinting , Animals , Cattle/genetics , Placenta/metabolism , Female , Polymorphism, Single Nucleotide , Pregnancy
14.
Anim Reprod Sci ; 265: 107470, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38657462

ABSTRACT

The global population as well as the demand for human food is rapidly growing worldwide, which necessitates improvement of efficiency in livestock operations. In this context, environmental factors during fetal and/or neonatal life have been observed to influence normal physical and physiological function of an individual during adulthood, and this phenomenon is called fetal or developmental programming. While numerous studies have reported the impact of maternal factors on development of the female progeny, limited information is available on the potential effects of fetal programming on reproductive function of the male offspring. Therefore, the objective for this review article was to focus on available literature regarding the impact of maternal factors, particularly maternal nutrition, on reproductive system of the male offspring. To this end, we highlighted developmental programming of the male offspring in domestic species (i.e., pig, cow and sheep) as well as laboratory species (i.e., mice and rat) during pregnancy and lactation. In this sense, we pointed out the effects of maternal nutrition on various functions of the male offspring including hypothalamic-pituitary axis, hormonal levels, testicular tissue and semen parameters.


Subject(s)
Maternal Nutritional Physiological Phenomena , Animals , Male , Female , Pregnancy , Fetal Development/physiology , Genomic Imprinting
15.
Nature ; 628(8006): 122-129, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38448590

ABSTRACT

Genomic imprinting-the non-equivalence of maternal and paternal genomes-is a critical process that has evolved independently in many plant and mammalian species1,2. According to kinship theory, imprinting is the inevitable consequence of conflictive selective forces acting on differentially expressed parental alleles3,4. Yet, how these epigenetic differences evolve in the first place is poorly understood3,5,6. Here we report the identification and molecular dissection of a parent-of-origin effect on gene expression that might help to clarify this fundamental question. Toxin-antidote elements (TAs) are selfish elements that spread in populations by poisoning non-carrier individuals7-9. In reciprocal crosses between two Caenorhabditis tropicalis wild isolates, we found that the slow-1/grow-1 TA is specifically inactive when paternally inherited. This parent-of-origin effect stems from transcriptional repression of the slow-1 toxin by the PIWI-interacting RNA (piRNA) host defence pathway. The repression requires PIWI Argonaute and SET-32 histone methyltransferase activities and is transgenerationally inherited via small RNAs. Remarkably, when slow-1/grow-1 is maternally inherited, slow-1 repression is halted by a translation-independent role of its maternal mRNA. That is, slow-1 transcripts loaded into eggs-but not SLOW-1 protein-are necessary and sufficient to counteract piRNA-mediated repression. Our findings show that parent-of-origin effects can evolve by co-option of the piRNA pathway and hinder the spread of selfish genes that require sex for their propagation.


Subject(s)
Caenorhabditis , Genomic Imprinting , Piwi-Interacting RNA , Repetitive Sequences, Nucleic Acid , Animals , Female , Male , Alleles , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Caenorhabditis/genetics , Caenorhabditis/metabolism , Crosses, Genetic , Fathers , Genome/genetics , Genomic Imprinting/genetics , Hermaphroditic Organisms/genetics , Histone Methyltransferases/genetics , Histone Methyltransferases/metabolism , Mothers , Oocytes/metabolism , Piwi-Interacting RNA/genetics , Protein Biosynthesis , Repetitive Sequences, Nucleic Acid/genetics , RNA, Messenger/genetics , Toxins, Biological/genetics , Transcription, Genetic
16.
Genes Dev ; 38(3-4): 131-150, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38453481

ABSTRACT

Maternal inactivation of genes encoding components of the subcortical maternal complex (SCMC) and its associated member, PADI6, generally results in early embryo lethality. In humans, SCMC gene variants were found in the healthy mothers of children affected by multilocus imprinting disturbances (MLID). However, how the SCMC controls the DNA methylation required to regulate imprinting remains poorly defined. We generated a mouse line carrying a Padi6 missense variant that was identified in a family with Beckwith-Wiedemann syndrome and MLID. If homozygous in female mice, this variant resulted in interruption of embryo development at the two-cell stage. Single-cell multiomic analyses demonstrated defective maturation of Padi6 mutant oocytes and incomplete DNA demethylation, down-regulation of zygotic genome activation (ZGA) genes, up-regulation of maternal decay genes, and developmental delay in two-cell embryos developing from Padi6 mutant oocytes but little effect on genomic imprinting. Western blotting and immunofluorescence analyses showed reduced levels of UHRF1 in oocytes and abnormal localization of DNMT1 and UHRF1 in both oocytes and zygotes. Treatment with 5-azacytidine reverted DNA hypermethylation but did not rescue the developmental arrest of mutant embryos. Taken together, this study demonstrates that PADI6 controls both nuclear and cytoplasmic oocyte processes that are necessary for preimplantation epigenetic reprogramming and ZGA.


Subject(s)
Oocytes , Zygote , Animals , Child , Female , Humans , Mice , CCAAT-Enhancer-Binding Proteins/genetics , Cytoplasm/genetics , Cytoplasm/metabolism , DNA Methylation/genetics , Embryonic Development/genetics , Genomic Imprinting/genetics , Ubiquitin-Protein Ligases/metabolism
17.
Mol Cell ; 84(8): 1442-1459.e7, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38458200

ABSTRACT

In mammals, dosage compensation involves two parallel processes: (1) X inactivation, which equalizes X chromosome dosage between males and females, and (2) X hyperactivation, which upregulates the active X for X-autosome balance. The field currently favors models whereby dosage compensation initiates "de novo" during mouse development. Here, we develop "So-Smart-seq" to revisit the question and interrogate a comprehensive transcriptome including noncoding genes and repeats in mice. Intriguingly, de novo silencing pertains only to a subset of Xp genes. Evolutionarily older genes and repetitive elements demonstrate constitutive Xp silencing, adopt distinct signatures, and do not require Xist to initiate silencing. We trace Xp silencing backward in developmental time to meiotic sex chromosome inactivation in the male germ line and observe that Xm hyperactivation is timed to Xp silencing on a gene-by-gene basis. Thus, during the gamete-to-embryo transition, older Xp genes are transmitted in a "pre-inactivated" state. These findings have implications for the evolution of imprinting.


Subject(s)
RNA, Long Noncoding , X Chromosome Inactivation , Female , Mice , Male , Animals , X Chromosome Inactivation/genetics , Genomic Imprinting , Germ Cells , Epigenesis, Genetic , Embryo, Mammalian , RNA, Long Noncoding/genetics , X Chromosome/genetics , Mammals/genetics
18.
Mol Genet Genomics ; 299(1): 40, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38546894

ABSTRACT

Genomic imprinting is an epigenetic regulation mechanism in mammals resulting in the parentally dependent monoallelic expression of genes. Imprinting disorders in humans are associated with several congenital syndromes and cancers and remain the focus of many medical studies. Cattle is a better model organism for investigating human embryo development than mice. Imprinted genes usually cluster on chromosomes and are regulated by different methylation regions (DMRs) located in imprinting control regions that control gene expression in cis. There is an imprinted locus on human chromosome 16q24.1 associated with congenital lethal developmental lung disease in newborns. However, genomic imprinting on bovine chromosome 18, which is homologous with human chromosome 16 has not been systematically studied. The aim of this study was to analyze the allelic expressions of eight genes (CDH13, ATP2C2, TLDC1, COTL1, CRISPLD2, ZDHHC7, KIAA0513, and GSE1) on bovine chromosome 18 and to search the DMRs associated gene allelic expression. Three transcript variants of the ZDHHC7 gene (X1, X2, and X5) showed maternal imprinting in bovine placentas. In addition, the monoallelic expression of X2 and X5 was tissue-specific. Five transcripts of the KIAA0513 gene showed tissue- and isoform-specific monoallelic expression. The CDH13, ATP2C2, and TLDC1 genes exhibited tissue-specific imprinting, however, COTL1, CRISLPLD2, and GSE1 escaped imprinting. Four DMRs, established after fertilization, were found in this region. Two DMRs were located between the ZDHHC7 and KIAA0513 genes, and two were in exon 1 of the CDH13 and ATP2C2 genes, respectively. The results from this study support future studies on the molecular mechanism to regulate the imprinting of candidate genes on bovine chromosome 18.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Infant, Newborn , Pregnancy , Female , Humans , Cattle/genetics , Animals , Mice , DNA Methylation/genetics , Chromosomes, Human, Pair 18 , Genomic Imprinting/genetics , Chromosomes , Mammals/genetics , Nerve Tissue Proteins/genetics
19.
Nat Struct Mol Biol ; 31(3): 404-412, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38499830

ABSTRACT

Cytosine DNA methylation is a highly conserved epigenetic mark in eukaryotes. Although the role of DNA methylation at gene promoters and repetitive elements has been extensively studied, the function of DNA methylation in other genomic contexts remains less clear. In the nucleus of mammalian cells, the genome is spatially organized at different levels, and strongly influences myriad genomic processes. There are a number of factors that regulate the three-dimensional (3D) organization of the genome, with the CTCF insulator protein being among the most well-characterized. Pertinently, CTCF binding has been reported as being DNA methylation-sensitive in certain contexts, perhaps most notably in the process of genomic imprinting. Therefore, it stands to reason that DNA methylation may play a broader role in the regulation of chromatin architecture. Here we summarize the current understanding that is relevant to both the mammalian DNA methylation and chromatin architecture fields and attempt to assess the extent to which DNA methylation impacts the folding of the genome. The focus is in early embryonic development and cellular transitions when the epigenome is in flux, but we also describe insights from pathological contexts, such as cancer, in which the epigenome and 3D genome organization are misregulated.


Subject(s)
DNA Methylation , Repressor Proteins , Animals , Repressor Proteins/metabolism , CCCTC-Binding Factor/metabolism , Genomic Imprinting , Chromatin , Mammals/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...