Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Epigenomics ; 16(2): 127-136, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38126127

ABSTRACT

The authors highlight an area of research that focuses on the establishment of genomic imprints: how the female and male germlines set up opposite instructions for imprinted genes in the maternally and paternally inherited chromosomes. Mouse genetics studies have solidified the role of transcription across the germline differentially methylated regions in the establishment of maternal genomic imprinting. One work now reveals that such transcription is also important in paternal imprinting establishment. This allows the authors to propose a unifying mechanism, in the form of transcription across germline differentially methylated regions, that specifies DNA methylation imprint establishment. Differences in the timing, genomic location and nature of such transcription events in the male versus female germlines in turn explain the difference between paternal and maternal imprints.


Subject(s)
DNA Methylation , Genomic Imprinting , Animals , Mice , Germ Cells
2.
Sci Adv ; 9(36): eadi2050, 2023 09 08.
Article in English | MEDLINE | ID: mdl-37672574

ABSTRACT

The insulator model explains the workings of the H19 and Igf2 imprinted domain in the soma, where insulation of the Igf2 promoter from its enhancers occurs by CTCF in the maternally inherited unmethylated chromosome but not the paternally inherited methylated allele. The molecular mechanism that targets paternal methylation imprint establishment to the imprinting control region (ICR) in the male germline is unknown. We tested the function of prospermatogonia-specific broad low-level transcription in this process using mouse genetics. Paternal imprint establishment was abnormal when transcription was stopped at the entry point to the ICR. The germline epimutation persisted into the paternal allele of the soma, resulting in reduced Igf2 in fetal organs and reduced fetal growth, consistent with the insulator model and insulin-like growth factor 2 (IGF2)'s role as fetal growth factor. These results collectively support the role of broad low-level transcription through the H19/Igf2 ICR in the establishment of its paternal methylation imprint in the male germ line, with implications for Silver-Russell syndrome.


Subject(s)
Fetal Development , Protein Processing, Post-Translational , Animals , Mice , Methylation , Alleles , Phosphorylation
4.
Biochem Soc Trans ; 50(6): 1875-1884, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36454621

ABSTRACT

We recently discovered a novel biological process, the scheduled remodeling of Z-DNA structures in the developing fetal mouse male germ cells [Nat. Cell Biol. 24, 1141-1153]. This process affects purine/pyrimidine dinucleotide repeat (PPR) rich sequences, which can form stable left-handed Z-DNA structures. The protein that carries out this function is identified as ZBTB43, member of a large family of ZBTB proteins. Z-DNA remodeling by ZBTB43 not only coincides with global remodeling of DNA methylation and chromatin events in the male germ line, but it also is a prerequisite for de novo DNA methylation. When ZBTB43 changes DNA structure from the left-handed zigzag shaped Z-DNA to the regular smooth right-handed B-DNA, it also generates a suitable substrate for the de novo DNA methyltransferase, DNMT3A. By instructing de novo DNA methylation at PPRs in prospermatogonia, ZBTB43 safeguards epigenomic integrity of the male gamete. PPRs are fragile sequences, sites of large deletions and rearrangements in mammalian cells, and this fragility is thought to be due to Z-DNA structure formation rather than the sequence itself. This idea is now supported by the in vivo finding that DNA double strand breaks accumulate in mutant prospermatogonia which lack ZBTB43-dependent Z-DNA remodeling. If unrepaired, double stranded DNA breaks can lead to germ line mutations. Therefore, by preventing such breaks ZBTB43 is critical for guarding genome stability between generations. Here, we discuss the significance and implications of these findings in more detail.


Subject(s)
DNA, Z-Form , Mice , Animals , Male , DNA, Z-Form/metabolism , Germ Cells/metabolism , Chromatin/metabolism , DNA Methylation , DNA/metabolism , Mammals/genetics , Mammals/metabolism
5.
Cell Genom ; 2(7)2022 Jul 13.
Article in English | MEDLINE | ID: mdl-35873672

ABSTRACT

We have developed a mouse DNA methylation array that contains 296,070 probes representing the diversity of mouse DNA methylation biology. We present a mouse methylation atlas as a rich reference resource of 1,239 DNA samples encompassing distinct tissues, strains, ages, sexes, and pathologies. We describe applications for comparative epigenomics, genomic imprinting, epigenetic inhibitors, patient-derived xenograft assessment, backcross tracing, and epigenetic clocks. We dissect DNA methylation processes associated with differentiation, aging, and tumorigenesis. Notably, we find that tissue-specific methylation signatures localize to binding sites for transcription factors controlling the corresponding tissue development. Age-associated hypermethylation is enriched at regions of Polycomb repression, while hypomethylation is enhanced at regions bound by cohesin complex members. Apc Min/+ polyp-associated hypermethylation affects enhancers regulating intestinal differentiation, while hypomethylation targets AP-1 binding sites. This Infinium Mouse Methylation BeadChip (version MM285) is widely accessible to the research community and will accelerate high-sample-throughput studies in this important model organism.

6.
Nat Cell Biol ; 24(7): 1141-1153, 2022 07.
Article in English | MEDLINE | ID: mdl-35787683

ABSTRACT

Mutagenic purine-pyrimidine repeats can adopt the left-handed Z-DNA conformation. DNA breaks at potential Z-DNA sites can lead to somatic mutations in cancer or to germline mutations that are transmitted to the next generation. It is not known whether any mechanism exists in the germ line to control Z-DNA structure and DNA breaks at purine-pyrimidine repeats. Here we provide genetic, epigenomic and biochemical evidence for the existence of a biological process that erases Z-DNA specifically in germ cells of the mouse male foetus. We show that a previously uncharacterized zinc finger protein, ZBTB43, binds to and removes Z-DNA, preventing the formation of DNA double-strand breaks. By removing Z-DNA, ZBTB43 also promotes de novo DNA methylation at CG-containing purine-pyrimidine repeats in prospermatogonia. Therefore, the genomic and epigenomic integrity of the species is safeguarded by remodelling DNA structure in the mammalian germ line during a critical window of germline epigenome reprogramming.


Subject(s)
DNA, Z-Form , Animals , DNA/metabolism , DNA Methylation , DNA, Z-Form/metabolism , Epigenome , Germ Cells/metabolism , Male , Mammals/metabolism , Mice , Nucleic Acid Conformation , Purines/metabolism , Pyrimidines
7.
Sci Adv ; 8(22): eabn3815, 2022 06 03.
Article in English | MEDLINE | ID: mdl-35658030

ABSTRACT

Oxidative DNA damage has been linked to inflammation, cancer, and aging. Here, we have mapped two types of oxidative DNA damage, oxidized guanines produced by hydrogen peroxide and oxidized thymines created by potassium permanganate, at a single-base resolution. 8-Oxo-guanine occurs strictly dependent on the G/C sequence context and shows a pronounced peak at transcription start sites (TSSs). We determined the trinucleotide sequence pattern of guanine oxidation. This pattern shows high similarity to the cancer-associated single-base substitution signatures SBS18 and SBS36. SBS36 is found in colorectal cancers that carry mutations in MUTYH, encoding a repair enzyme that operates on 8-oxo-guanine mispairs. SBS18 is common in inflammation-associated upper gastrointestinal tract tumors including esophageal and gastric adenocarcinomas. Oxidized thymines induced by permanganate occur with a distinct dinucleotide specificity, 5'T-A/C, and are depleted at the TSS. Our data suggest that two cancer mutational signatures, SBS18 and SBS36, are caused by reactive oxygen species.


Subject(s)
Neoplasms , Upper Gastrointestinal Tract , DNA Damage , Guanine , Humans , Hydrogen Peroxide/pharmacology , Inflammation , Mutation , Oxidation-Reduction
8.
PLoS Genet ; 17(11): e1009908, 2021 11.
Article in English | MEDLINE | ID: mdl-34793451

ABSTRACT

EHMT2 is the main euchromatic H3K9 methyltransferase. Embryos with zygotic, or maternal mutation in the Ehmt2 gene exhibit variable developmental delay. To understand how EHMT2 prevents variable developmental delay we performed RNA sequencing of mutant and somite stage-matched normal embryos at 8.5-9.5 days of gestation. Using four-way comparisons between delayed and normal embryos we clarified what it takes to be normal and what it takes to develop. We identified differentially expressed genes, for example Hox genes that simply reflected the difference in developmental progression of wild type and the delayed mutant uterus-mate embryos. By comparing wild type and zygotic mutant embryos along the same developmental window we detected a role of EHMT2 in suppressing variation in the transcriptional switches. We identified transcription changes where precise switching during development occurred only in the normal but not in the mutant embryo. At the 6-somite stage, gastrulation-specific genes were not precisely switched off in the Ehmt2-/- zygotic mutant embryos, while genes involved in organ growth, connective tissue development, striated muscle development, muscle differentiation, and cartilage development were not precisely switched on. The Ehmt2mat-/+ maternal mutant embryos displayed high transcriptional variation consistent with their variable survival. Variable derepression of transcripts occurred dominantly in the maternally inherited allele. Transcription was normal in the parental haploinsufficient wild type embryos despite their delay, consistent with their good prospects. Global profiling of transposable elements revealed EHMT2 targeted DNA methylation and suppression at LTR repeats, mostly ERVKs. In Ehmt2-/- embryos, transcription over very long distances initiated from such misregulated 'driver' ERVK repeats, encompassing a multitude of misexpressed 'passenger' repeats. In summary, EHMT2 reduced transcriptional variation of developmental switch genes and developmentally switching repeat elements at the six-somite stage embryos. These findings establish EHMT2 as a suppressor of transcriptional and developmental variation at the transition between gastrulation and organ specification.


Subject(s)
Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/metabolism , Transcription, Genetic , Animals , CpG Islands , DNA Methylation , Female , Haploinsufficiency , Histone-Lysine N-Methyltransferase/genetics , Mice , Transcriptome
9.
Epigenomics ; 13(16): 1299-1314, 2021 08.
Article in English | MEDLINE | ID: mdl-34519223

ABSTRACT

Aim: Paternal allele-specific expression of noncanonical imprinted genes in the extraembryonic lineages depends on an H3K27me3-based imprint in the oocyte, which is not a lasting mark. We hypothesized that EHMT2, the main euchromatic H3K9 dimethyltransferase, also has a role in controlling noncanonical imprinting. Methods: We carried out allele-specific total RNA-seq analysis in the ectoplacental cone of somite-matched 8.5 days post coitum embryos using reciprocal mouse crosses. Results: We found that the maternal allele of noncanonical imprinted genes was derepressed from its ERVK promoter in the Ehmt2-/- ectoplacental cone. In Ehmt2-/- embryos, loss of DNA methylation accompanied biallelic derepression of the ERVK promoters. Canonical imprinting and imprinted X chromosome inactivation were generally undisturbed. Conclusion: EHMT2 is essential for repressing the maternal allele in noncanonical imprinting.


Subject(s)
Gene Expression Regulation , Genomic Imprinting , Histone-Lysine N-Methyltransferase/metabolism , Animals , Biomarkers , DNA Methylation , Female , Male , Mice , Mice, Knockout , Mice, Transgenic , Placenta/metabolism , Pregnancy , Promoter Regions, Genetic , Spermatozoa , Whole Genome Sequencing
10.
Cell Rep ; 34(6): 108729, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33567274

ABSTRACT

Beckwith-Wiedemann syndrome (BWS) and Silver-Russell syndrome (SRS) are imprinting disorders manifesting as aberrant fetal growth and severe postnatal-growth-related complications. Based on the insulator model, one-third of BWS cases and two-thirds of SRS cases are consistent with misexpression of insulin-like growth factor 2 (IGF2), an important facilitator of fetal growth. We propose that the IGF2-dependent BWS and SRS cases can be identified by prenatal diagnosis and can be prevented by prenatal intervention targeting IGF2. We test this hypothesis using our mouse models of IGF2-dependent BWS and SRS. We find that genetically normalizing IGF2 levels in a double rescue experiment corrects the fetal overgrowth phenotype in the BWS model and the growth retardation in the SRS model. In addition, we pharmacologically rescue the BWS growth phenotype by reducing IGF2 signaling during late gestation. This animal study encourages clinical investigations to target IGF2 for prenatal diagnosis and prenatal prevention in human BWS and SRS.


Subject(s)
Beckwith-Wiedemann Syndrome , Gene Targeting , Insulin-Like Growth Factor II , Prenatal Diagnosis , Silver-Russell Syndrome , Animals , Beckwith-Wiedemann Syndrome/diagnosis , Beckwith-Wiedemann Syndrome/embryology , Beckwith-Wiedemann Syndrome/genetics , Beckwith-Wiedemann Syndrome/therapy , Disease Models, Animal , Female , Humans , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Mice , Pregnancy , Silver-Russell Syndrome/diagnosis , Silver-Russell Syndrome/embryology , Silver-Russell Syndrome/genetics , Silver-Russell Syndrome/therapy
11.
Methods Mol Biol ; 2198: 147-157, 2021.
Article in English | MEDLINE | ID: mdl-32822029

ABSTRACT

DNA methylation undergoes dynamic changes at the genome-wide scale during the early steps of mammalian embryo development. Immunochemical detection of 5-methylcytosine (5mC) in the zygote has led to the discovery that a global loss of DNA methylation takes place soon after fertilization, occurring rapidly in the paternal pronucleus. Using the same method employed above, which detects modified bases in the denatured single stranded DNA, we showed that this active DNA "demethylation" in the paternal pronucleus involves oxidation of 5mC to 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and 5-carboxycytosine (5caC) by the TET3 enzyme. By immunostaining of genetically altered zygotes we revealed that the maternal pronucleus is protected from TET3-mediated oxidation by histone H3K9 methyltransferase enzymes, EHMT2 and SETDB1. The same assays are also applicable for visualizing the temporal and spatial distribution of the modified cytosine residues in preimplantation embryos. Here, we provide a detailed protocol for detecting 5mC, 5hmC, 5fC, and 5caC in mouse zygotes and preimplantation-stage embryos using antibodies raised against modified cytosine species.


Subject(s)
Blastocyst/metabolism , Cytosine/metabolism , DNA Methylation , Embryo, Mammalian/metabolism , Embryonic Development/genetics , 5-Methylcytosine/metabolism , Animals , Cytosine/analogs & derivatives , Embryo, Mammalian/embryology , Humans , Immunochemistry/methods , Mammals , Microscopy, Confocal , Zygote/metabolism
12.
Methods Mol Biol ; 2198: 321-331, 2021.
Article in English | MEDLINE | ID: mdl-32822042

ABSTRACT

DNA cytosine modification is an important epigenetic mechanism that serves critical functions in a variety of biological processes in development and disease. 5-Methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) are the two most common epigenetic marks found in the mammalian genome. 5hmC is generated from 5mC by the ten-eleven translocation (TET) family of dioxygenase enzymes. This modification can reach substantial levels in certain cell types such as embryonic stem cells and neurons. Standard bisulfite sequencing techniques cannot distinguish between 5mC and 5hmC. Therefore, the method of TET-assisted bisulfite sequencing has been developed for detecting 5hmC specifically. The method is based on protection of 5hmC by glycosylation followed by complete oxidation of both 5mC and 5fC to 5caC, which converts to uracil after bisulfite treatment leaving only 5hmC remaining as a cytosine signal after PCR and sequencing. The method requires a highly active TET protein for the conversion steps. Here, we present an efficient TET protein purification method and a streamlined TAB-sequencing protocol for 5hmC analysis at single base resolution.


Subject(s)
5-Methylcytosine/analogs & derivatives , Epigenomics/methods , Sequence Analysis, DNA/methods , 5-Methylcytosine/analysis , 5-Methylcytosine/chemistry , Animals , Cytosine/analysis , Cytosine/metabolism , DNA/genetics , DNA Methylation/genetics , Dioxygenases/genetics , Dioxygenases/metabolism , Epigenesis, Genetic/genetics , Genome , Humans , Oxidation-Reduction , Sulfites/chemistry
13.
Sci Rep ; 10(1): 20636, 2020 11 26.
Article in English | MEDLINE | ID: mdl-33244015

ABSTRACT

A battery of chromatin modifying enzymes play essential roles in remodeling the epigenome in the zygote and cleavage stage embryos, when the maternal genome is the sole contributor. Here we identify an exemption. DOT1L methylates lysine 79 in the globular domain of histone H3 (H3K79). Dot1l is an essential gene, as homozygous null mutant mouse embryos exhibit multiple developmental abnormalities and die before 11.5 days of gestation. To test if maternally deposited DOT1L is required for embryo development, we carried out a conditional Dot1l knockout in growing oocytes using the Zona pellucida 3-Cre (Zp3-Cre) transgenic mice. We found that the resulting maternal mutant Dot1lmat-/+ offspring displayed normal development and fertility, suggesting that the expression of the paternally inherited copy of Dot1l in the embryo is sufficient to support development. In addition, Dot1l maternal deletion did not affect the parental allele-specific expression of imprinted genes, indicating that DOT1L is not needed for imprint establishment in the oocyte or imprint protection in the zygote. In summary, uniquely and as opposed to other histone methyltransferases and histone marks, maternal DOT1L deposition and H3K79 methylation in the zygote and in the preimplantation stage embryo is dispensable for mouse development.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Animals , Blastocyst/metabolism , Cell Proliferation/physiology , Embryo, Mammalian/metabolism , Embryonic Development/physiology , Histone Methyltransferases/metabolism , Histones/metabolism , Lysine/metabolism , Methylation , Mice , Mice, Knockout , Mice, Transgenic , Models, Animal , Oocytes/metabolism , Zygote/metabolism
14.
J Mol Biol ; 432(6): 1718-1730, 2020 Mar 13.
Article in English | MEDLINE | ID: mdl-31401118

ABSTRACT

5-Methylcytosine (5mC), the major modified DNA base in mammalian cells, can be oxidized enzymatically to 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and 5-carboxylcytosine (5caC) by the Ten-Eleven-Translocation (TET) family of proteins. Whereas 5fC and 5caC are recognized and removed by base excision repair proteins, the 5hmC base accumulates to substantial levels in certain cell types such as brain-derived neurons and is viewed as a relatively stable DNA base. As such, the existence of "reader" proteins that recognize 5hmC would be a logical assumption, and various searches have been undertaken to identify proteins that specifically bind to 5hmC and the other oxidized 5mC bases. However, the existence of definitive 5hmC "readers" has remained unclear and proteins interacting specifically with 5fC or 5caC are also very few. On the other hand, 5hmC is incapable of interacting with a number of proteins that recognize 5mC at CpG sequences, suggesting that 5hmC is an anti-reader modification that may serve to displace 5mC readers from DNA. In this review article, we discuss candidate proteins that may interact with oxidized 5mC bases.

15.
Proc Natl Acad Sci U S A ; 116(22): 10834-10841, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31088968

ABSTRACT

Genome-wide DNA "demethylation" in the zygote involves global TET3-mediated oxidation of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and 5-carboxylcytosine (5caC) in the paternal pronucleus. Asymmetrically enriched histone H3K9 methylation in the maternal pronucleus was suggested to protect the underlying DNA from 5mC conversion. We hypothesized that an H3K9 methyltransferase enzyme, either EHMT2 or SETDB1, must be expressed in the oocyte to specify the asymmetry of 5mC oxidation. To test these possibilities, we genetically deleted the catalytic domain of either EHMT2 or SETDB1 in growing oocytes and achieved significant reduction of global H3K9me2 or H3K9me3 levels, respectively, in the maternal pronucleus. We found that the asymmetry of global 5mC oxidation was significantly reduced in the zygotes that carried maternal mutation of either the Ehmt2 or Setdb1 genes. Whereas the levels of 5hmC, 5fC, and 5caC increased, 5mC levels decreased in the mutant maternal pronuclei. H3K9me3-rich rings around the nucleolar-like bodies retained 5mC in the maternal mutant zygotes, suggesting that the pericentromeric heterochromatin regions are protected from DNA demethylation independently of EHMT2 and SETDB1. We observed that the maternal pronuclei expanded in size in the mutant zygotes and contained a significantly increased number of nucleolar-like bodies compared with normal zygotes. These findings suggest that oocyte-derived EHMT2 and SETDB1 enzymes have roles in regulating 5mC oxidation and in the structural aspects of zygote development.


Subject(s)
5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Oocytes/metabolism , Animals , Female , Histone-Lysine N-Methyltransferase/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Oxidation-Reduction , Zygote/metabolism
16.
Nat Commun ; 10(1): 2046, 2019 05 03.
Article in English | MEDLINE | ID: mdl-31053723

ABSTRACT

Impaired neuronal processes, including dopamine imbalance, are central to the pathogenesis of major psychosis, but the molecular origins are unclear. Here we perform a multi-omics study of neurons isolated from the prefrontal cortex in schizophrenia and bipolar disorder (n = 55 cases and 27 controls). DNA methylation, transcriptomic, and genetic-epigenetic interactions in major psychosis converged on pathways of neurodevelopment, synaptic activity, and immune functions. We observe prominent hypomethylation of an enhancer within the insulin-like growth factor 2 (IGF2) gene in major psychosis neurons. Chromatin conformation analysis revealed that this enhancer targets the nearby tyrosine hydroxylase (TH) gene responsible for dopamine synthesis. In patients, we find hypomethylation of the IGF2 enhancer is associated with increased TH protein levels. In mice, Igf2 enhancer deletion disrupts the levels of TH protein and striatal dopamine, and induces transcriptional and proteomic abnormalities affecting neuronal structure and signaling. Our data suggests that epigenetic activation of the enhancer at IGF2 may enhance dopamine synthesis associated with major psychosis.


Subject(s)
Bipolar Disorder/genetics , Dopamine/biosynthesis , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Insulin-Like Growth Factor II/genetics , Schizophrenia/genetics , Tyrosine 3-Monooxygenase/genetics , Adult , Aged , Animals , Bipolar Disorder/pathology , DNA Methylation , Female , Gene Expression Profiling , Humans , Male , Mice , Mice, Knockout , Middle Aged , Neurons/pathology , Prefrontal Cortex/cytology , Prefrontal Cortex/pathology , Proteomics , Schizophrenia/pathology , Transcriptome/genetics , Tyrosine 3-Monooxygenase/metabolism , Young Adult
18.
Elife ; 52016 11 28.
Article in English | MEDLINE | ID: mdl-27892853

ABSTRACT

The germline genome of the binucleated ciliate Tetrahymena thermophila undergoes programmed chromosome breakage and massive DNA elimination to generate the somatic genome. Here, we present a complete sequence assembly of the germline genome and analyze multiple features of its structure and its relationship to the somatic genome, shedding light on the mechanisms of genome rearrangement as well as the evolutionary history of this remarkable germline/soma differentiation. Our results strengthen the notion that a complex, dynamic, and ongoing interplay between mobile DNA elements and the host genome have shaped Tetrahymena chromosome structure, locally and globally. Non-standard outcomes of rearrangement events, including the generation of short-lived somatic chromosomes and excision of DNA interrupting protein-coding regions, may represent novel forms of developmental gene regulation. We also compare Tetrahymena's germline/soma differentiation to that of other characterized ciliates, illustrating the wide diversity of adaptations that have occurred within this phylum.


Subject(s)
Gene Rearrangement , Genome, Protozoan , Tetrahymena thermophila/genetics , Sequence Analysis, DNA
19.
Genome Biol ; 17(1): 154, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27411809

ABSTRACT

In a recent paper, we described our efforts in search for evidence supporting epigenetic transgenerational inheritance caused by endocrine disrupter chemicals. One aspect of our study was to compare genome-wide DNA methylation changes in the vinclozolin-exposed fetal male germ cells (n = 3) to control samples (n = 3), their counterparts in the next, unexposed, generation (n = 3 + 3) and also in adult spermatozoa (n = 2 + 2) in both generations. We reported finding zero common hits in the intersection of these four comparisons. In our interpretation, this result did not support the notion that DNA methylation provides a mechanism for a vinclozolin-induced transgenerational male infertility phenotype. In response to criticism by Guerrero-Bosagna regarding our statistical power in the above study, here we provide power calculations to clarify the statistical power of our study and to show the validity of our conclusions. We also explain here how our data is misinterpreted in the commentary by Guerrero-Bosagna by leaving out important data points from consideration.Please see related Correspondence article: xxx (13059_2016_982) and related Research article: http://genomebiology.biomedcentral.com/articles/10.1186/s13059-015-0619-z.


Subject(s)
DNA Methylation/genetics , Endocrine Disruptors/toxicity , Epigenesis, Genetic , Oxazoles/toxicity , DNA Methylation/drug effects , Embryonic Germ Cells/drug effects , Embryonic Germ Cells/metabolism , Humans , Male , Spermatozoa/drug effects , Spermatozoa/pathology
20.
Genome Biol ; 17: 105, 2016 May 16.
Article in English | MEDLINE | ID: mdl-27184890

ABSTRACT

Abhay Sharma brings two arguments in favor of transgenerational epigenetic inheritance (TGEI) in mammals when criticizing our work. He uses probability calculations and finds that the probability of obtaining the number of common changes in the in utero-exposed prospermatogonia and the same cells in the next generation is significant in our study. He also compares our results to other published datasets and concludes that the probability for the observed overlap between independent studies is significant. We disagree with both arguments of Sharma and show here that his meta-analysis and statistical calculations are not correct.


Subject(s)
Cellular Reprogramming/genetics , DNA Methylation/genetics , Epigenesis, Genetic/drug effects , Genomic Imprinting/drug effects , Animals , Female , Male , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...