77 results on '"Matthew C. Lorincz"'
Search Results
2. Evolution of imprinting via lineage-specific insertion of retroviral promoters
- Author
-
Aaron B. Bogutz, Julie Brind’Amour, Hisato Kobayashi, Kristoffer N. Jensen, Kazuhiko Nakabayashi, Hiroo Imai, Matthew C. Lorincz, and Louis Lefebvre
- Subjects
Science - Abstract
Although many species-specific imprinted genes have been identified, how the evolutionary switch from biallelic to imprinted expression occurs is still unknown. Here authors find that lineage-specific ERVs active as oocyte promoters can induce de novo DNA methylation at gDMRs and imprinting.
- Published
- 2019
- Full Text
- View/download PDF
3. ZFP57 regulation of transposable elements and gene expression within and beyond imprinted domains
- Author
-
Hui Shi, Ruslan Strogantsev, Nozomi Takahashi, Anastasiya Kazachenka, Matthew C. Lorincz, Myriam Hemberger, and Anne C. Ferguson-Smith
- Subjects
DNA methylation ,KZFPs ,ZFP57 ,Transposable elements ,Embryonic stem cells ,Genetics ,QH426-470 - Abstract
Abstract Background KRAB zinc finger proteins (KZFPs) represent one of the largest families of DNA-binding proteins in vertebrate genomes and appear to have evolved to silence transposable elements (TEs) including endogenous retroviruses through sequence-specific targeting of repressive chromatin states. ZFP57 is required to maintain the post-fertilization DNA methylation memory of parental origin at genomic imprints. Here we conduct RNA-seq and ChIP-seq analyses in normal and ZFP57 mutant mouse ES cells to understand the relative importance of ZFP57 at imprints, unique and repetitive regions of the genome. Results Over 80% of ZFP57 targets are TEs, however, ZFP57 is not essential for their repression. The remaining targets lie within unique imprinted and non-imprinted sequences. Though the loss of ZFP57 influences imprinted genes as expected, the majority of unique gene targets lose H3K9me3 with little effect on DNA methylation and very few exhibit alterations in expression. Comparison of ZFP57 mutants with DNA methyltransferase-deleted ES cells (TKO) identifies a remarkably similar pattern of H3K9me3 loss across the genome. These data define regions where H3K9me3 is secondary to DNA methylation and we propose that ZFP57 is the principal if not sole methylation-sensitive KZFP in mouse ES cells. Finally, we examine dynamics of DNA and H3K9 methylation during pre-implantation development and show that sites bound by ZFP57 in ES cells maintain DNA methylation and H3K9me3 at imprints and at non-imprinted regions on the maternally inherited chromosome throughout preimplantation development. Conclusion Our analyses suggest the evolution of a rare DNA methylation-sensitive KZFP that is not essential for repeat silencing, but whose primary function is to maintain DNA methylation and repressive histone marks at germline-derived imprinting control regions.
- Published
- 2019
- Full Text
- View/download PDF
4. LTR retrotransposons transcribed in oocytes drive species-specific and heritable changes in DNA methylation
- Author
-
Julie Brind’Amour, Hisato Kobayashi, Julien Richard Albert, Kenjiro Shirane, Akihiko Sakashita, Asuka Kamio, Aaron Bogutz, Tasuku Koike, Mohammad M. Karimi, Louis Lefebvre, Tomohiro Kono, and Matthew C. Lorincz
- Subjects
Science - Abstract
De novo DNA methylation during mouse oogenesis occurs within transcribed regions. Here the authors investigate the role of species-specific long terminal repeats (LTRs)-initiated transcription units in regulating the oocyte methylome, identifying syntenic regions in mouse, rat and human with divergent DNA methylation associated with private LITs.
- Published
- 2018
- Full Text
- View/download PDF
5. Development and application of an integrated allele-specific pipeline for methylomic and epigenomic analysis (MEA)
- Author
-
Julien Richard Albert, Tasuku Koike, Hamid Younesy, Richard Thompson, Aaron B. Bogutz, Mohammad M. Karimi, and Matthew C. Lorincz
- Subjects
Epigenomics ,Allele-specific ,Allelic ,RNA-seq ,Chromatin immunoprecipitation ,ChIP ,Biotechnology ,TP248.13-248.65 ,Genetics ,QH426-470 - Abstract
Abstract Background Allele-specific transcriptional regulation, including of imprinted genes, is essential for normal mammalian development. While the regulatory regions controlling imprinted genes are associated with DNA methylation (DNAme) and specific histone modifications, the interplay between transcription and these epigenetic marks at allelic resolution is typically not investigated genome-wide due to a lack of bioinformatic packages that can process and integrate multiple epigenomic datasets with allelic resolution. In addition, existing ad-hoc software only consider SNVs for allele-specific read discovery. This limitation omits potentially informative INDELs, which constitute about one fifth of the number of SNVs in mice, and introduces a systematic reference bias in allele-specific analyses. Results Here, we describe MEA, an INDEL-aware Methylomic and Epigenomic Allele-specific analysis pipeline which enables user-friendly data exploration, visualization and interpretation of allelic imbalance. Applying MEA to mouse embryonic datasets yields robust allele-specific DNAme maps and low reference bias. We validate allele-specific DNAme at known differentially methylated regions and show that automated integration of such methylation data with RNA- and ChIP-seq datasets yields an intuitive, multidimensional view of allelic gene regulation. MEA uncovers numerous novel dynamically methylated loci, highlighting the sensitivity of our pipeline. Furthermore, processing and visualization of epigenomic datasets from human brain reveals the expected allele-specific enrichment of H3K27ac and DNAme at imprinted as well as novel monoallelically expressed genes, highlighting MEA’s utility for integrating human datasets of distinct provenance for genome-wide analysis of allelic phenomena. Conclusions Our novel pipeline for standardized allele-specific processing and visualization of disparate epigenomic and methylomic datasets enables rapid analysis and navigation with allelic resolution. MEA is freely available as a Docker container at https://github.com/julienrichardalbert/MEA.
- Published
- 2018
- Full Text
- View/download PDF
6. Histone H3K9 Methyltransferase G9a in Oocytes Is Essential for Preimplantation Development but Dispensable for CG Methylation Protection
- Author
-
Wan Kin Au Yeung, Julie Brind’Amour, Yu Hatano, Kazuo Yamagata, Robert Feil, Matthew C. Lorincz, Makoto Tachibana, Yoichi Shinkai, and Hiroyuki Sasaki
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Mammalian histone methyltransferase G9a (also called EHMT2) deposits H3K9me2 on chromatin and is essential for postimplantation development. However, its role in oogenesis and preimplantation development remains poorly understood. We show that H3K9me2-enriched chromatin domains in mouse oocytes are generally depleted of CG methylation, contrasting with their association in embryonic stem and somatic cells. Oocyte-specific disruption of G9a results in reduced H3K9me2 enrichment and impaired reorganization of heterochromatin in oocytes, but only a modest reduction in CG methylation is detected. Furthermore, in both oocytes and 2-cell embryos, G9a depletion has limited impact on the expression of genes and retrotransposons. Although their CG methylation is minimally affected, preimplantation embryos derived from such oocytes show abnormal chromosome segregation and frequent developmental arrest. Our findings illuminate the functional importance of G9a independent of CG methylation in preimplantation development and call into question the proposed role for H3K9me2 in CG methylation protection in zygotes. : Au Yeung et al. report that H3K9 methyltransferase G9a in mouse oocytes is essential for preimplantation development. Contrary to the previous model, however, maternal G9a is dispensable for CG methylation protection in zygotes and is instead important for chromatin reorganization in oocytes and proper chromosome segregation in preimplantation embryos. Keywords: oocyte, preimplantation embryo, histone modification, DNA methylation, G9a, H3K9me2, chromatin organization, chromosome segregation
- Published
- 2019
- Full Text
- View/download PDF
7. Inter-Strain Epigenomic Profiling Reveals a Candidate IAP Master Copy in C3H Mice
- Author
-
Rita Rebollo, Mariana Galvão-Ferrarini, Liane Gagnier, Ying Zhang, Ardian Ferraj, Christine R. Beck, Matthew C. Lorincz, and Dixie L. Mager
- Subjects
transposable elements ,endogenous retrovirus ,mouse ,C3H ,IAP ,epigenetics ,Microbiology ,QR1-502 - Abstract
Insertions of endogenous retroviruses cause a significant fraction of mutations in inbred mice but not all strains are equally susceptible. Notably, most new Intracisternal A particle (IAP) ERV mutagenic insertions have occurred in C3H mice. We show here that strain-specific insertional polymorphic IAPs accumulate faster in C3H/HeJ mice, relative to other sequenced strains, and that IAP transcript levels are higher in C3H/HeJ embryonic stem (ES) cells compared to other ES cells. To investigate the mechanism for high IAP activity in C3H mice, we identified 61 IAP copies in C3H/HeJ ES cells enriched with H3K4me3 (a mark of active promoters) and, among those tested, all are unmethylated in C3H/HeJ ES cells. Notably, 13 of the 61 are specific to C3H/HeJ and are members of the non-autonomous 1Δ1 IAP subfamily that is responsible for nearly all new insertions in C3H. One copy is full length with intact open reading frames and hence potentially capable of providing proteins in trans to other 1Δ1 elements. This potential “master copy” is present in other strains, including 129, but its 5’ long terminal repeat (LTR) is methylated in 129 ES cells. Thus, the unusual IAP activity in C3H may be due to reduced epigenetic repression coupled with the presence of a master copy.
- Published
- 2020
- Full Text
- View/download PDF
8. Repression of germline genes by PRC1.6 and SETDB1 in the early embryo precedes DNA methylation-mediated silencing
- Author
-
Akihiko Okuda, Ayumu Suzuki, Kentaro Mochizuki, Kenjiro Shirane, Haruhiko Koseki, Kousuke Uranishi, Aaron B. Bogutz, Jafar Sharif, Matthew C. Lorincz, and Sanne M. Janssen
- Subjects
Epigenomics ,Male ,Methyltransferase ,General Physics and Astronomy ,E2F6 Transcription Factor ,Polycomb-Group Proteins ,Germline ,Epigenesis, Genetic ,Histones ,Mice ,0302 clinical medicine ,Genes, Reporter ,Basic Helix-Loop-Helix Transcription Factors ,Polycomb Repressive Complex 1 ,0303 health sciences ,Multidisciplinary ,biology ,Basic Helix-Loop-Helix Leucine Zipper Transcription Factors ,Gene Expression Regulation, Developmental ,Mouse Embryonic Stem Cells ,Cell biology ,Histone ,DNA methylation ,Female ,Signal Transduction ,Ubiquitin-Protein Ligases ,Science ,Green Fluorescent Proteins ,Mice, Transgenic ,macromolecular substances ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Gene silencing ,Animals ,Embryo Implantation ,Gene Silencing ,Psychological repression ,Gene ,030304 developmental biology ,Cell lineage ,Promoter ,General Chemistry ,Histone-Lysine N-Methyltransferase ,DNA Methylation ,Embryo, Mammalian ,Mice, Inbred C57BL ,Protein Subunits ,biology.protein ,030217 neurology & neurosurgery - Abstract
Silencing of a subset of germline genes is dependent upon DNA methylation (DNAme) post-implantation. However, these genes are generally hypomethylated in the blastocyst, implicating alternative repressive pathways before implantation. Indeed, in embryonic stem cells (ESCs), an overlapping set of genes, including germline “genome-defence” (GGD) genes, are upregulated following deletion of the H3K9 methyltransferase SETDB1 or subunits of the non-canonical PRC1 complex PRC1.6. Here, we show that in pre-implantation embryos and naïve ESCs (nESCs), hypomethylated promoters of germline genes bound by the PRC1.6 DNA-binding subunits MGA/MAX/E2F6 are enriched for RING1B-dependent H2AK119ub1 and H3K9me3. Accordingly, repression of these genes in nESCs shows a greater dependence on PRC1.6 than DNAme. In contrast, GGD genes are hypermethylated in epiblast-like cells (EpiLCs) and their silencing is dependent upon SETDB1, PRC1.6/RING1B and DNAme, with H3K9me3 and DNAme establishment dependent upon MGA binding. Thus, GGD genes are initially repressed by PRC1.6, with DNAme subsequently engaged in post-implantation embryos., Germline genes are repressed by DNA methylation and histone marks. Here, the authors show that specific germline genes hypomethylated pre-implantation are enriched for PRC1.6, H2AK119ub1 and H3K9me3, which coordinately repress their expression.
- Published
- 2021
9. Profiling Histone Methylation in Low Numbers of Cells
- Author
-
Julie, Brind'Amour and Matthew C, Lorincz
- Subjects
Histones ,Chromatin Immunoprecipitation ,High-Throughput Nucleotide Sequencing ,Sequence Analysis, DNA ,Methylation ,Protein Processing, Post-Translational - Abstract
Chromatin immunoprecipitation (ChIP) enables the study of DNA-protein interactions. When coupled with high-throughput sequencing (ChIP-seq), this method allows the generation of genome-wide profiles of the distribution of specific proteins in a given cellular context. Typical ChIP-seq experiments require millions of cells as input material and thus are not ideal to study many in vivo cell populations. Here, we describe an ultra-low-input native ChIP-seq method, ULI-NChIP-seq, to profile histone modification patterns in as low as 150 cells.
- Published
- 2022
10. Transcription shapes genome-wide histone acetylation patterns
- Author
-
Zhen Cheng Liu, Matthew C. Lorincz, LeAnn J. Howe, Julie Brind’Amour, Anastasia Kuzmin, Kristoffer N. Jensen, and Benjamin J. E. Martin
- Subjects
0301 basic medicine ,Science ,General Physics and Astronomy ,RNA polymerase II ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Transcription (biology) ,parasitic diseases ,Histone post-translational modifications ,Nucleosome ,Gene ,Histone Acetyltransferases ,Multidisciplinary ,biology ,Chemistry ,Acetylation ,General Chemistry ,DNA ,Cell biology ,Chromatin ,030104 developmental biology ,Histone ,biology.protein ,Transcription ,030217 neurology & neurosurgery ,Histone analysis - Abstract
Histone acetylation is a ubiquitous hallmark of transcription, but whether the link between histone acetylation and transcription is causal or consequential has not been addressed. Using immunoblot and chromatin immunoprecipitation-sequencing in S. cerevisiae, here we show that the majority of histone acetylation is dependent on transcription. This dependency is partially explained by the requirement of RNA polymerase II (RNAPII) for the interaction of H4 histone acetyltransferases (HATs) with gene bodies. Our data also confirms the targeting of HATs by transcription activators, but interestingly, promoter-bound HATs are unable to acetylate histones in the absence of transcription. Indeed, HAT occupancy alone poorly predicts histone acetylation genome-wide, suggesting that HAT activity is regulated post-recruitment. Consistent with this, we show that histone acetylation increases at nucleosomes predicted to stall RNAPII, supporting the hypothesis that this modification is dependent on nucleosome disruption during transcription. Collectively, these data show that histone acetylation is a consequence of RNAPII promoting both the recruitment and activity of histone acetyltransferases., Histone acetylation is a ubiquitous hallmark of transcription. Here the authors provide evidence that the majority of histone acetylation is dependent on transcription, specifically due to the requirement of RNAPII for the recruitment and activity of histone acetyltransferases.
- Published
- 2021
11. Maternal DNMT3A-dependent de novo methylation of the paternal genome inhibits gene expression in the early embryo
- Author
-
Julien Richard Albert, Wan Kin Au Yeung, Hiroyuki Sasaki, Aaron B. Bogutz, Keisuke Toriyama, Matthew C. Lorincz, Hisato Kobayashi, Ryutaro Hirasawa, and Julie Brind’Amour
- Subjects
0301 basic medicine ,Epigenomics ,Male ,Science ,Bisulfite sequencing ,General Physics and Astronomy ,Biology ,Genome ,General Biochemistry, Genetics and Molecular Biology ,Article ,DNA Methyltransferase 3A ,03 medical and health sciences ,0302 clinical medicine ,Animals ,DNA (Cytosine-5-)-Methyltransferases ,Paternal Inheritance ,lcsh:Science ,Alleles ,Genetics ,Multidisciplinary ,Zygote ,DNA methylation ,Gene Expression Regulation, Developmental ,Imprinting ,General Chemistry ,Spermatozoa ,Gene regulation ,030104 developmental biology ,Blastocyst ,CpG site ,Mice, Inbred DBA ,Embryogenesis ,embryonic structures ,Oocytes ,CpG Islands ,Female ,lcsh:Q ,Maternal Inheritance ,Reprogramming ,030217 neurology & neurosurgery - Abstract
De novo DNA methylation (DNAme) during mammalian spermatogenesis yields a densely methylated genome, with the exception of CpG islands (CGIs), which are hypomethylated in sperm. While the paternal genome undergoes widespread DNAme loss before the first S-phase following fertilization, recent mass spectrometry analysis revealed that the zygotic paternal genome is paradoxically also subject to a low level of de novo DNAme. However, the loci involved, and impact on transcription were not addressed. Here, we employ allele-specific analysis of whole-genome bisulphite sequencing data and show that a number of genomic regions, including several dozen CGI promoters, are de novo methylated on the paternal genome by the 2-cell stage. A subset of these promoters maintains DNAme through development to the blastocyst stage. Consistent with paternal DNAme acquisition, many of these loci are hypermethylated in androgenetic blastocysts but hypomethylated in parthenogenetic blastocysts. Paternal DNAme acquisition is lost following maternal deletion of Dnmt3a, with a subset of promoters, which are normally transcribed from the paternal allele in blastocysts, being prematurely transcribed at the 4-cell stage in maternal Dnmt3a knockout embryos. These observations uncover a role for maternal DNMT3A activity in post-fertilization epigenetic reprogramming and transcriptional silencing of the paternal genome., The paternal genome in mice undergoes widespread DNA methylation loss post-fertilization. Here, the authors apply allele-specific analysis of WGBS data to show that a number of genomic regions are simultaneously de novo methylated on the paternal genome dependent on maternal DNMT3A activity, which induces transcriptional silencing of this allele in the early embryo.
- Published
- 2020
12. NSD1-deposited H3K36me2 directs de novo methylation in the mouse male germline and counteracts Polycomb-associated silencing
- Author
-
Takashi Ito, Matthew C. Lorincz, Kenjiro Shirane, and Fumihito Miura
- Subjects
0303 health sciences ,Methyltransferase ,Euchromatin ,Methylation ,Biology ,Germline ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,SETD2 ,Genetics ,Gene silencing ,Genomic imprinting ,Gene ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
De novo DNA methylation (DNAme) in mammalian germ cells is dependent on DNMT3A and DNMT3L. However, oocytes and spermatozoa show distinct patterns of DNAme. In mouse oocytes, de novo DNAme requires the lysine methyltransferase (KMTase) SETD2, which deposits H3K36me3. We show here that SETD2 is dispensable for de novo DNAme in the male germline. Instead, the lysine methyltransferase NSD1, which broadly deposits H3K36me2 in euchromatic regions, plays a critical role in de novo DNAme in prospermatogonia, including at imprinted genes. However, males deficient in germline NSD1 show a more severe defect in spermatogenesis than Dnmt3l-/- males. Notably, unlike DNMT3L, NSD1 safeguards a subset of genes against H3K27me3-associated transcriptional silencing. In contrast, H3K36me2 in oocytes is predominantly dependent on SETD2 and coincides with H3K36me3. Furthermore, females with NSD1-deficient oocytes are fertile. Thus, the sexually dimorphic pattern of DNAme in mature mouse gametes is orchestrated by distinct profiles of H3K36 methylation.
- Published
- 2020
13. Profiling Histone Methylation in Low Numbers of Cells
- Author
-
Julie Brind’Amour and Matthew C. Lorincz
- Published
- 2022
14. Interplay between chromatin marks in development and disease
- Author
-
Sanne M, Janssen and Matthew C, Lorincz
- Subjects
Histones ,Mice ,Animals ,Humans ,Disease ,Growth and Development ,DNA Methylation ,Chromatin Assembly and Disassembly ,Protein Processing, Post-Translational ,Chromatin - Abstract
DNA methylation (DNAme) and histone post-translational modifications (PTMs) have important roles in transcriptional regulation. Although many reports have characterized the functions of such chromatin marks in isolation, recent genome-wide studies reveal surprisingly complex interactions between them. Here, we focus on the interplay between DNAme and methylation of specific lysine residues on the histone H3 tail. We describe the impact of genetic perturbation of the relevant methyltransferases in the mouse on the landscape of chromatin marks as well as the transcriptome. In addition, we discuss the specific neurodevelopmental growth syndromes and cancers resulting from pathogenic mutations in the human orthologues of these genes. Integrating these observations underscores the fundamental importance of crosstalk between DNA and histone H3 methylation in development and disease.
- Published
- 2021
15. Paternal MTHFR deficiency leads to hypomethylation of young retrotransposons and reproductive decline across two successive generations
- Author
-
Gurbet Karahan, Kenjiro Shirane, Jay M. Baltz, Matthew C. Lorincz, Taylor McClatchie, Sanne M. Janssen, Jacquetta M. Trasler, and Donovan Chan
- Subjects
Epigenomics ,Male ,Embryonic Germ Cells ,Mouse ,Retroelements ,Methylenetetrahydrofolate reductase deficiency ,Intergenerational epigenetic inheritance ,Retrotransposon ,Chromatin and Epigenetics ,Biology ,Andrology ,03 medical and health sciences ,Fathers ,Mice ,0302 clinical medicine ,Folic Acid ,medicine ,Animals ,Young retrotransposons ,Molecular Biology ,Methylenetetrahydrofolate Reductase (NADPH2) ,030304 developmental biology ,0303 health sciences ,Reproductive Biology ,DNA methylation ,Reproduction ,Male germ cell development ,Methylation ,medicine.disease ,Sperm ,Spermatozoa ,digestive system diseases ,Mice, Inbred C57BL ,Germ Cells ,Psychotic Disorders ,Muscle Spasticity ,Methylenetetrahydrofolate reductase ,MTHFR ,biology.protein ,Female ,Homocystinuria ,Reprogramming ,030217 neurology & neurosurgery ,Developmental Biology ,Research Article - Abstract
5,10-Methylenetetrahydrofolate reductase (MTHFR) is a crucial enzyme in the folate metabolic pathway with a key role in generating methyl groups. As MTHFR deficiency impacts male fertility and sperm DNA methylation, there is the potential for epimutations to be passed to the next generation. Here, we assessed whether the impact of MTHFR deficiency on testis morphology and sperm DNA methylation is exacerbated across generations in mouse. Although MTHFR deficiency in F1 fathers has only minor effects on sperm counts and testis weights and histology, F2 generation sons show further deterioration in reproductive parameters. Extensive loss of DNA methylation is observed in both F1 and F2 sperm, with >80% of sites shared between generations, suggestive of regions consistently susceptible to MTHFR deficiency. These regions are generally methylated during late embryonic germ cell development and are enriched in young retrotransposons. As retrotransposons are resistant to reprogramming of DNA methylation in embryonic germ cells, their hypomethylated state in the sperm of F1 males could contribute to the worsening reproductive phenotype observed in F2 MTHFR-deficient males, compatible with the intergenerational passage of epimutations., Summary: MTHFR deficiency results in sperm DNA hypomethylation of regions enriched in potentially active young retrotransposons that are targets of a piRNA- and DNMT3C-dependent de novo DNA methylation pathway, leading to reproductive defects that are more severe in subsequent generations.
- Published
- 2021
16. Setting the chromatin stage in oocytes
- Author
-
Matthew C. Lorincz and Julie Brind’Amour
- Subjects
0303 health sciences ,Zygote ,biology ,Lysine ,Cell Biology ,Nuclear reprogramming ,Cell biology ,Chromatin ,03 medical and health sciences ,0302 clinical medicine ,Histone ,030220 oncology & carcinogenesis ,biology.protein ,Maternal to zygotic transition ,Demethylase ,030304 developmental biology ,Demethylation - Abstract
Post-translational histone modifications are important regulators of nuclear reprogramming. A study now reveals that histone lysine demethylase KDM4A-mediated H3K9me3 demethylation in mammalian oocytes is essential for zygotic genome activation and preimplantation development.
- Published
- 2020
17. NSD1-deposited H3K36me2 directs de novo methylation in the mouse male germline and counteracts Polycomb-associated silencing
- Author
-
Kenjiro, Shirane, Fumihito, Miura, Takashi, Ito, and Matthew C, Lorincz
- Subjects
Male ,Mice, Knockout ,Nuclear Proteins ,Polycomb-Group Proteins ,Histone-Lysine N-Methyltransferase ,DNA Methylation ,Spermatozoa ,DNA Methyltransferase 3A ,Histones ,Mice ,Oocytes ,Animals ,Humans ,Female ,DNA (Cytosine-5-)-Methyltransferases ,Spermatogenesis ,Transcription Factors - Abstract
De novo DNA methylation (DNAme) in mammalian germ cells is dependent on DNMT3A and DNMT3L. However, oocytes and spermatozoa show distinct patterns of DNAme. In mouse oocytes, de novo DNAme requires the lysine methyltransferase (KMTase) SETD2, which deposits H3K36me3. We show here that SETD2 is dispensable for de novo DNAme in the male germline. Instead, the lysine methyltransferase NSD1, which broadly deposits H3K36me2 in euchromatic regions, plays a critical role in de novo DNAme in prospermatogonia, including at imprinted genes. However, males deficient in germline NSD1 show a more severe defect in spermatogenesis than Dnmt3l
- Published
- 2020
18. Setting the chromatin stage in oocytes
- Author
-
Julie, Brind'Amour and Matthew C, Lorincz
- Subjects
Histones ,Zygote ,Oocytes ,Chromatin ,Article - Abstract
The importance of germline-inherited posttranslational histone modifications on priming early mammalian development is just emerging1–4. Histone H3 lysine 9 (H3K9) trimethylation is associated with heterochromatin and gene repression during cell-fate change5, while histone H3 lysine 4 (H3K4) trimethylation marks active gene promoters6. Mature oocytes are transcriptionally quiescent and possess remarkably broad domains of H3K4me3 (bdH3K4me3)1,2. It remains unknown as to which factors contribute to the maintenance of the bdH3K4me3 landscape. Lysine-specific demethylase 4A (KDM4A) demethylates H3K9me3 at promoters marked by H3K4me3 in actively transcribing somatic cells7. Here, we report that KDM4A-mediated H3K9me3 demethylation at bdH3K4me3 in oocytes is crucial for normal preimplantation development and zygotic genome activation (ZGA) after fertilization. Loss of KDM4A in oocytes causes aberrant H3K9me3 spreading over bdH3K4me3, resulting in insufficient transcriptional activation of genes, endogenous retroviral elements and long terminal repeat initiated chimeric transcripts during ZGA. The catalytic activity of KDM4A is essential for normal epigenetic reprogramming and preimplantation development. Hence, KDM4A plays a crucial role in preserving maternal epigenome integrity required for proper ZGA and transfer of developmental control to the embryo.
- Published
- 2020
19. Maternal DNMT3A-dependent de novo methylation of the zygotic paternal genome inhibits gene expression in the early embryo
- Author
-
Julien Richard Albert, Hisato Kobayashi, Keisuke Toriyama, Wan Kin Au Yeung, Julie Brind’Amour, Matthew C. Lorincz, Hiroyuki Sasaki, Aaron B. Bogutz, and Ryutaro Hirasawa
- Subjects
Genetics ,0303 health sciences ,Zygote ,Bisulfite sequencing ,Promoter ,Methylation ,Biology ,Genome ,03 medical and health sciences ,0302 clinical medicine ,CpG site ,embryonic structures ,Allele ,Reprogramming ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
De novo DNA methylation (DNAme) during mammalian spermatogenesis yields a densely methylated genome, with the exception of CpG islands (CGIs), which are hypomethylated in sperm. Following fertilization, the paternal genome undergoes widespread DNAme loss before the first S-phase. Paradoxically, recent mass spectrometry analysis revealed that a low level of de novo DNAme occurs exclusively on the zygotic paternal genome. However, the loci involved and impact on genic transcription was not addressed. Here, we employ allele-specific analysis of wholegenome bisulphite sequencing (WGBS) data and show that a number of genomic regions, including several dozen CGI promoters, are de novo methylated on the paternal genome in 2-cell embryos. A subset of these promoters maintains DNAme through development to the blastocyst stage. Consistent with zygotic paternal DNAme acquisition (PDA), many of these loci are hypermethylated in androgenetic blastocysts but hypomethylated in parthenogenetic blastocysts. Strikingly, PDA is lost following maternal deletion ofDnmt3a. Furthermore, a subset of promoters showing PDA which are normally transcribed from the paternal allele in blastocysts show premature transcription at the 4-cell stage in maternalDnmt3aknockout embryos. These observations uncover an unexpected role for maternal DNMT3A activity in postfertilization epigenetic reprogramming and transcriptional silencing of the paternal genome.
- Published
- 2020
20. Evolution of imprinting via lineage-specific insertion of retroviral promoters
- Author
-
Matthew C. Lorincz, Hiroo Imai, Hisato Kobayashi, Louis Lefebvre, Kazuhiko Nakabayashi, Julie Brind’Amour, Kristoffer N. Jensen, and Aaron B. Bogutz
- Subjects
Epigenomics ,Male ,Primates ,0301 basic medicine ,Pan troglodytes ,Science ,General Physics and Astronomy ,Endogenous retrovirus ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Evolution, Molecular ,Genomic Imprinting ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Species Specificity ,Transcription (biology) ,Animals ,Humans ,Imprinting (psychology) ,Promoter Regions, Genetic ,lcsh:Science ,Gene ,030304 developmental biology ,Genetics ,0303 health sciences ,Multidisciplinary ,Terminal Repeat Sequences ,Imprinting ,Promoter ,Haplorhini ,General Chemistry ,DNA Methylation ,Germ Cells ,Retroviridae ,030104 developmental biology ,Differentially methylated regions ,DNA methylation ,Oocytes ,Macaca ,Epigenetics ,Female ,lcsh:Q ,Genomic imprinting ,030217 neurology & neurosurgery - Abstract
Imprinted genes are expressed from a single parental allele, with the other allele often silenced by DNA methylation (DNAme) established in the germline. While species-specific imprinted orthologues have been documented, the molecular mechanisms underlying the evolutionary switch from biallelic to imprinted expression are unknown. During mouse oogenesis, gametic differentially methylated regions (gDMRs) acquire DNAme in a transcription-guided manner. Here we show that oocyte transcription initiating in lineage-specific endogenous retroviruses (ERVs) is likely responsible for DNAme establishment at 4/6 mouse-specific and 17/110 human-specific imprinted gDMRs. The latter are divided into Catarrhini- or Hominoidea-specific gDMRs embedded within transcripts initiating in ERVs specific to these primate lineages. Strikingly, imprinting of the maternally methylated genes Impact and Slc38a4 was lost in the offspring of female mice harboring deletions of the relevant murine-specific ERVs upstream of these genes. Our work reveals an evolutionary mechanism whereby maternally silenced genes arise from biallelically expressed progenitors., Although many species-specific imprinted genes have been identified, how the evolutionary switch from biallelic to imprinted expression occurs is still unknown. Here authors find that lineage-specific ERVs active as oocyte promoters can induce de novo DNA methylation at gDMRs and imprinting.
- Published
- 2019
21. The majority of histone acetylation is a consequence of transcription
- Author
-
Matthew C. Lorincz, LeAnn J. Howe, Julie Brind’Amour, Zhen Cheng Liu, Anastasia Kuzmin, Kristoffer N. Jensen, and Benjamin J. E. Martin
- Subjects
Histone Acetyltransferases ,0303 health sciences ,Transcriptional activity ,030302 biochemistry & molecular biology ,Biology ,Cell biology ,Chromatin ,Histone H4 ,03 medical and health sciences ,Histone ,Acetylation ,Transcription (biology) ,parasitic diseases ,biology.protein ,Gene ,030304 developmental biology - Abstract
Histone acetylation is a ubiquitous hallmark of transcriptional activity, but whether the link is of a causal or consequential nature is still a matter of debate. In this study we resolve this question. Using both immunoblot analysis and chromatin immunoprecipitation-sequencing (ChIP-seq) in S. cerevisiae, we show that the majority of histone acetylation is dependent on transcription. Loss of histone H4 acetylation upon transcription inhibition is partially explained by depletion of histone acetyltransferases (HATs) from gene bodies, implicating transcription in HAT targeting. Despite this, HAT occupancy alone poorly predicts histone acetylation, suggesting that HAT activity is regulated at a step post-recruitment. Collectively, these data show that the majority of histone acetylation is a consequence of RNAPII promoting both the recruitment and activity of histone acetyltransferases.
- Published
- 2019
- Full Text
- View/download PDF
22. ZFP57 regulation of transposable elements and gene expression within and beyond imprinted domains
- Author
-
Anastasiya Kazachenka, Ruslan Strogantsev, Matthew C. Lorincz, Myriam Hemberger, Nozomi Takahashi, Hui Shi, Anne C. Ferguson-Smith, Apollo - University of Cambridge Repository, Takahashi, Nozomi [0000-0002-4267-1094], and Ferguson-Smith, Anne [0000-0003-4996-9990]
- Subjects
Transposable element ,Embryonic stem cells ,lcsh:QH426-470 ,Endogenous retrovirus ,KZFPs ,Biology ,Tripartite Motif-Containing Protein 28 ,Genome ,Cell Line ,Histones ,03 medical and health sciences ,chemistry.chemical_compound ,Genomic Imprinting ,Mice ,0302 clinical medicine ,Genetics ,Animals ,Molecular Biology ,ZFP57 ,030304 developmental biology ,0303 health sciences ,DNA methylation ,Binding Sites ,Research ,Chromatin ,Mice, Inbred C57BL ,Repressor Proteins ,lcsh:Genetics ,Histone ,chemistry ,biology.protein ,DNA Transposable Elements ,Genomic imprinting ,Transposable elements ,030217 neurology & neurosurgery ,DNA ,Protein Binding - Abstract
Background KRAB zinc finger proteins (KZFPs) represent one of the largest families of DNA-binding proteins in vertebrate genomes and appear to have evolved to silence transposable elements (TEs) including endogenous retroviruses through sequence-specific targeting of repressive chromatin states. ZFP57 is required to maintain the post-fertilization DNA methylation memory of parental origin at genomic imprints. Here we conduct RNA-seq and ChIP-seq analyses in normal and ZFP57 mutant mouse ES cells to understand the relative importance of ZFP57 at imprints, unique and repetitive regions of the genome. Results Over 80% of ZFP57 targets are TEs, however, ZFP57 is not essential for their repression. The remaining targets lie within unique imprinted and non-imprinted sequences. Though the loss of ZFP57 influences imprinted genes as expected, the majority of unique gene targets lose H3K9me3 with little effect on DNA methylation and very few exhibit alterations in expression. Comparison of ZFP57 mutants with DNA methyltransferase-deleted ES cells (TKO) identifies a remarkably similar pattern of H3K9me3 loss across the genome. These data define regions where H3K9me3 is secondary to DNA methylation and we propose that ZFP57 is the principal if not sole methylation-sensitive KZFP in mouse ES cells. Finally, we examine dynamics of DNA and H3K9 methylation during pre-implantation development and show that sites bound by ZFP57 in ES cells maintain DNA methylation and H3K9me3 at imprints and at non-imprinted regions on the maternally inherited chromosome throughout preimplantation development. Conclusion Our analyses suggest the evolution of a rare DNA methylation-sensitive KZFP that is not essential for repeat silencing, but whose primary function is to maintain DNA methylation and repressive histone marks at germline-derived imprinting control regions. Electronic supplementary material The online version of this article (10.1186/s13072-019-0295-4) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
23. Epigenetic regulation of unique genes and repetitive elements by the KRAB zinc finger protein ZFP57
- Author
-
Anne C. Ferguson-Smith, Matthew C. Lorincz, Anastasiya Kazachenka, Ruslan Strogantsev, Myriam Hemberger, HaoTian H. Shi, and Nozomi Takahashi
- Subjects
Genetics ,0303 health sciences ,Methylation ,Biology ,Chromatin ,03 medical and health sciences ,0302 clinical medicine ,Histone ,DNA methylation ,biology.protein ,Epigenetics ,Genomic imprinting ,Gene ,Reprogramming ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
BackgroundKRAB-zinc finger proteins (KZFPs) represent one of the largest families of DNA binding proteins in vertebrate genomes and appear to have evolved to silence transposable elements (TEs) including endogenous retroviruses through sequence-specific targeting of repressive chromatin states. ZFP57 is required to maintain the post-fertilization DNA methylation memory of parental-origin at genomic imprints along with ZFP445 which is specific for imprints. However, ZFP57 has multiple methylated genomic targets. Here we conduct RNA-seq and ChIP-seq analyses in normal and ZFP57 mutant mouse ES cells to understand the relative importance of ZFP57 at unique and repetitive regions of the genome.ResultsOver 80% of ZFP57 targets are TEs, however, ZFP57 is not essential for their repression. The remaining targets lie within unique imprinted and non-imprinted sequences. Though loss of ZFP57 influences imprinted genes as expected, the majority of unique gene targets lose H3K9me3 with little effect on DNA methylation and very few exhibiting alterations in expression. Comparison with DNA methyltransferase-deleted ES cells (TKO) identifies remarkably similar losses of H3K9me3 and changes in expression, defining regions where H3K9me3 is secondary to DNA methylation. We show that ZFP57 is the principal methylation-sensitive KZFP recruiting KAP1 and H3K9me3 in ES cells. Finally, like imprints, other unique targets of ZFP57 are enriched for germline-derived DNA methylation including oocyte-specific methylation that is resistant to post-fertilisation epigenetic reprogramming.ConclusionOur analyses suggest the evolution of a rare DNA methylation-sensitive KZFP that is not essential for repeat silencing, but whose primary function is to maintain DNA methylation and repressive histone marks at germline derived imprinting control regions.
- Published
- 2019
24. Vertebrate diapause preserves organisms long term through Polycomb complex members
- Author
-
Matthew C. Lorincz, Alejandro Sánchez Alvarado, Julie Brind’Amour, Chi-Kuo Hu, Wei Wang, Param Priya Singh, G. Adam Reeves, and Anne Brunet
- Subjects
Polycomb-Group Proteins ,Diapause ,Biology ,Article ,Nothobranchius furzeri ,Transcriptome ,Histones ,03 medical and health sciences ,0302 clinical medicine ,biology.animal ,Animals ,Killifish ,Muscle, Skeletal ,Gene ,Organism ,030304 developmental biology ,Polycomb Repressive Complex 1 ,0303 health sciences ,Multidisciplinary ,Killifishes ,Vertebrate ,Gene Expression Regulation, Developmental ,biology.organism_classification ,Cell biology ,Chromatin ,Vertebrates ,Mutation ,030217 neurology & neurosurgery - Abstract
Putting vertebrate development on hold Suspended animation is an often-used device in science fiction, but it also exists in several forms in nature: hibernation, torpor, and diapause. Hu et al. studied diapause in the African turquoise killifish, a vertebrate model system (see the Perspective by Van Gilst). They found that diapause protects a complex living organism without trade-offs for future growth, fertility, and even life span. Diapause is actively regulated, with a dynamic switch to specific Polycomb complex members. One Polycomb member, CBX7, is critical for the regulation of organ genes and is involved in muscle preservation and diapause maintenance. This work illuminates the mechanisms that underlie suspended life. Science , this issue p. 870 ; see also p. 851
- Published
- 2018
25. Histone H3 lysine 4 trimethylation in sperm is transmitted to the embryo and associated with diet-induced phenotypes in the offspring
- Author
-
Julie Brind’Amour, Ariane Lismer, Sarah Kimmins, Christine Lafleur, Vanessa Dumeaux, Romain Lambrot, and Matthew C. Lorincz
- Subjects
Male ,Histone H3 Lysine 4 ,Offspring ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Congenital Abnormalities ,Epigenesis, Genetic ,Histones ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,Animals ,Epigenetics ,Enhancer ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Gene Expression Regulation, Developmental ,Embryo ,Cell Biology ,DNA Methylation ,Embryo, Mammalian ,Spermatozoa ,Sperm ,Chromatin ,Diet ,Cell biology ,Mice, Inbred C57BL ,Phenotype ,Animals, Newborn ,H3K4me3 ,Female ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Summary A father's lifestyle impacts offspring health; yet, the underlying molecular mechanisms remain elusive. We hypothesized that a diet that changes methyl donor availability will alter the sperm and embryo epigenomes to impact embryonic gene expression and development. Here, we demonstrate that a folate-deficient (FD) diet alters histone H3 lysine 4 trimethylation (H3K4me3) in sperm at developmental genes and putative enhancers. A subset of H3K4me3 alterations in sperm are retained in the pre-implantation embryo and associated with deregulated embryonic gene expression. Using a genetic mouse model in which sires have pre-existing altered H3K4me2/3 in sperm, we show that a FD diet exacerbates alterations in sperm H3K4me3 and embryonic gene expression, leading to an increase in developmental defect severity. These findings imply that paternal H3K4me3 is transmitted to the embryo and influences gene expression and development. It further suggests that epigenetic errors can accumulate in sperm to worsen offspring developmental outcomes.
- Published
- 2021
26. Activation of Endogenous Retroviruses in Dnmt1 −/− ESCs Involves Disruption of SETDB1-Mediated Repression by NP95 Binding to Hemimethylated DNA
- Author
-
Osamu Ohara, Yoichi Shinkai, Jafar Sharif, Makoto Nakanishi, Haruhiko Koseki, Kayoko Katsuyama, Yoko Mizutani-Koseki, Midori Shimada, Julie Brind’Amour, Tomoyuki Ishikura, Hehuang Xie, Preeti Goyal, Takaho A. Endo, Mohammad M. Karimi, Manabu Nakayama, Matthew C. Lorincz, Zhixiong Sun, and Ming-an Sun
- Subjects
DNA (Cytosine-5-)-Methyltransferase 1 ,0301 basic medicine ,Placenta ,Ubiquitin-Protein Ligases ,Cellular differentiation ,Gene Dosage ,Endogenous retrovirus ,Biology ,Models, Biological ,Histones ,Mice ,03 medical and health sciences ,Protein Domains ,Pregnancy ,Genetics ,Animals ,DNA (Cytosine-5-)-Methyltransferases ,Gene Silencing ,Epigenetics ,Mice, Knockout ,Regulation of gene expression ,urogenital system ,Lysine ,Endogenous Retroviruses ,Gene Expression Regulation, Developmental ,Nuclear Proteins ,Cell Differentiation ,Mouse Embryonic Stem Cells ,DNA ,Histone-Lysine N-Methyltransferase ,Cell Biology ,DNA Methylation ,Trophoblasts ,Genes, Intracisternal A-Particle ,030104 developmental biology ,DNA demethylation ,Histone ,Genetic Loci ,Mutation ,embryonic structures ,DNA methylation ,CCAAT-Enhancer-Binding Proteins ,DNMT1 ,biology.protein ,Molecular Medicine ,Female ,Virus Activation ,Protein Binding - Abstract
Repression of endogenous retroviruses (ERVs) in mammals involves several epigenetic mechanisms. Acute loss of the maintenance methyltransferase Dnmt1 induces widespread DNA demethylation and transcriptional activation of ERVs, including CpG-rich IAP (intracisternal A particle) proviruses. Here, we show that this effect is not due simply to a loss of DNA methylation. Conditional deletions reveal that both Dnmt1 and Np95 are essential for maintenance DNA methylation. However, while IAPs are derepressed in Dnmt1-ablated embryos and embryonic stem cells (ESCs), these ERVs remain silenced when Np95 is deleted alone or in combination with Dnmt1. This paradoxical phenotype results from an ectopic interaction between NP95 and the H3K9 methyltransferase SETDB1. Normally, SETDB1 maintains silencing of IAPs, but in the absence of DNMT1, prolonged binding of NP95 to hemimethylated DNA transiently disrupts SETDB1-dependent H3K9me3 deposition. Thus, our observations reveal an unexpected antagonistic interplay between two repressive pathways involved in retroviral silencing in mammalian cells.
- Published
- 2016
27. LTR retrotransposons transcribed in oocytes drive species-specific and heritable changes in DNA methylation
- Author
-
Asuka Kamio, Mohammad M. Karimi, Julien Richard Albert, Kenjiro Shirane, Matthew C. Lorincz, Louis Lefebvre, Tomohiro Kono, Akihiko Sakashita, Hisato Kobayashi, Tasuku Koike, Aaron B. Bogutz, and Julie Brind’Amour
- Subjects
0301 basic medicine ,Retroelements ,Transcription, Genetic ,Science ,Inheritance Patterns ,General Physics and Astronomy ,Retrotransposon ,Biology ,Synteny ,Genome ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Species Specificity ,Animals ,Humans ,RNA, Messenger ,Epigenetics ,lcsh:Science ,Mammals ,Regulation of gene expression ,Genetics ,Polymorphism, Genetic ,Multidisciplinary ,Terminal Repeat Sequences ,Promoter ,General Chemistry ,DNA Methylation ,Long terminal repeat ,Rats ,Mice, Inbred C57BL ,030104 developmental biology ,Gene Expression Regulation ,CpG site ,Fertilization ,DNA methylation ,Oocytes ,CpG Islands ,DNA, Intergenic ,lcsh:Q - Abstract
De novo DNA methylation (DNAme) during mouse oogenesis occurs within transcribed regions enriched for H3K36me3. As many oocyte transcripts originate in long terminal repeats (LTRs), which are heterogeneous even between closely related mammals, we examined whether species-specific LTR-initiated transcription units (LITs) shape the oocyte methylome. Here we identify thousands of syntenic regions in mouse, rat, and human that show divergent DNAme associated with private LITs, many of which initiate in lineage-specific LTR retrotransposons. Furthermore, CpG island (CGI) promoters methylated in mouse and/or rat, but not human oocytes, are embedded within rodent-specific LITs and vice versa. Notably, at a subset of such CGI promoters, DNAme persists on the maternal genome in fertilized and parthenogenetic mouse blastocysts or in human placenta, indicative of species-specific epigenetic inheritance. Polymorphic LITs are also responsible for disparate DNAme at promoter CGIs in distantly related mouse strains, revealing that LITs also promote intra-species divergence in CGI DNAme., De novo DNA methylation during mouse oogenesis occurs within transcribed regions. Here the authors investigate the role of species-specific long terminal repeats (LTRs)-initiated transcription units in regulating the oocyte methylome, identifying syntenic regions in mouse, rat and human with divergent DNA methylation associated with private LITs.
- Published
- 2018
28. ChAsE: chromatin analysis and exploration tool
- Author
-
Mohammad M. Karimi, Cydney B. Nielsen, Matthew C. Lorincz, Torsten Möller, Hamid Younesy, and Steven J.M. Jones
- Subjects
0301 basic medicine ,Statistics and Probability ,Source code ,Computer science ,media_common.quotation_subject ,Interface (computing) ,Biochemistry ,Plot (graphics) ,03 medical and health sciences ,Software ,Animals ,Cluster Analysis ,Humans ,Cluster analysis ,Molecular Biology ,Interactive visualization ,media_common ,Information retrieval ,business.industry ,Usability ,Applications Notes ,Chromatin ,Computer Science Applications ,Computational Mathematics ,030104 developmental biology ,Computational Theory and Mathematics ,Programming Languages ,business - Abstract
Summary : We present ChAsE, a cross-platform desktop application developed for interactive visualization, exploration and clustering of epigenomic data such as ChIP-seq experiments. ChAsE is designed and developed in close collaboration with several groups of biologists and bioinformaticians with a focus on usability and interactivity. Data can be analyzed through k-means clustering, specifying presence or absence of signal in epigenetic data and performing set operations between clusters. Results can be explored in an interactive heat map and profile plot interface and exported for downstream analysis or as high quality figures suitable for publications. Availability and Implementation: Software, source code (MIT License), data and video tutorials available at http://chase.cs.univie.ac.at . Contact : mkarimi@brc.ubc.ca or torsten.moeller@univie.ac.at Supplementary information: Supplementary data are available at Bioinformatics online.
- Published
- 2016
29. Evidence for Converging DNA Methylation Pathways in Placenta and Cancer
- Author
-
Matthew C. Lorincz and Dirk Schübeler
- Subjects
0301 basic medicine ,Somatic cell ,Carcinogenesis ,Placenta ,Biology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Pregnancy ,Neoplasms ,medicine ,Animals ,Humans ,Promoter Regions, Genetic ,Molecular Biology ,Genetics ,Cancer ,Promoter ,Cell Biology ,DNA Methylation ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,CpG site ,DNA methylation ,CpG Islands ,Female ,Developmental Biology - Abstract
In mammals, the canonical somatic DNA methylation landscape is established upon specification of the embryo proper and subsequently disrupted within many cancer types1-4. However, the underlying mechanisms that direct this genome-scale transformation remain elusive, with no clear model for its systematic acquisition or potential developmental utility5,6. Here we analyzed global remethylation from the mouse preimplantation embryo into the early epiblast and extraembryonic ectoderm. We show that these two states acquire highly divergent genomic distributions with substantial disruption of bimodal, CpG density-dependent methylation in the placental progenitor7,8. The extraembryonic epigenome includes specific de novo methylation at hundreds of embryonically-protected CpG island promoters particularly those that are associated with key developmental regulators and orthologously methylated across most human cancer types9. Our data suggest that the evolutionary innovation of extraembryonic tissues may have required cooption of DNA methylation-based suppression as an alternative to the embryonically utilized Polycomb group proteins, which coordinate germlayer formation in response to extraembryonic cues10. Moreover, we establish that this decision is made deterministically downstream of promiscuously utilized, and frequently oncogenic, signaling pathways via a novel combination of epigenetic cofactors. Methylation of developmental gene promoters during tumorigenesis may therefore reflect the misappropriation of an innate trajectory and the spontaneous reacquisition of a latent, developmentally-encoded epigenetic landscape.
- Published
- 2017
30. Epigenetic modifier drugs trigger widespread transcription of endogenous retroviruses
- Author
-
Dixie L. Mager and Matthew C. Lorincz
- Subjects
0301 basic medicine ,Regulation of gene expression ,Genetics ,Epigenomics ,Epigenetic modifier ,Direct effects ,Endogenous Retroviruses ,Endogenous retrovirus ,Biology ,03 medical and health sciences ,030104 developmental biology ,Transcription (biology) ,Humans ,Epigenetics ,Gene - Abstract
A study in this issue demonstrates that epigenome-modifying drugs used in cancer chemotherapy induce transcription from thousands of previously unannotated transcription start sites, most of which are derived from ancient endogenous retroviruses (ERVs). This work, coupled with previous related findings, suggests that induction of ERVs, rather than direct effects on specific genes, may have a central role in the cellular responses to such agents and, in turn, their therapeutic efficacy.
- Published
- 2017
31. Regulation of DNA methylation turnover at LTR retrotransposons and imprinted loci by the histone methyltransferase Setdb1
- Author
-
Preeti Goyal, Matthew C. Lorincz, Danny Leung, Wei Xie, Tingting Du, Keith E. Szulwach, Yujing Li, Peng Jin, Bing Ren, Ulrich Wagner, and Ah Young Lee
- Subjects
Male ,Retroelements ,Biology ,DNA Methyltransferase 3A ,Epigenesis, Genetic ,Histones ,Genomic Imprinting ,Mice ,Epigenetics of physical exercise ,Histone methylation ,Animals ,DNA (Cytosine-5-)-Methyltransferases ,Epigenetics ,RNA-Directed DNA Methylation ,Cells, Cultured ,Embryonic Stem Cells ,Epigenomics ,Mice, Knockout ,Genetics ,Multidisciplinary ,Endogenous Retroviruses ,Histone-Lysine N-Methyltransferase ,DNA Methylation ,Biological Sciences ,Histone methyltransferase ,DNA methylation ,Female ,Reprogramming - Abstract
During mammalian development, DNA methylation patterns need to be reset in primordial germ cells (PGCs) and preimplantation embryos. However, many LTR retrotransposons and imprinted genes are impervious to such global epigenetic reprogramming via hitherto undefined mechanisms. Here, we report that a subset of such genomic regions are resistant to widespread erasure of DNA methylation in mouse embryonic stem cells (mESCs) lacking the de novo DNA methyltransferases (Dnmts) Dnmt3a and Dnmt3b. Intriguingly, these loci are enriched for H3K9me3 in mESCs, implicating this mark in DNA methylation homeostasis. Indeed, deletion of the H3K9 methyltransferase SET domain bifurcated 1 (Setdb1) results in reduced H3K9me3 and DNA methylation levels at specific loci, concomitant with increased 5-hydroxymethylation (5hmC) and ten-eleven translocation 1 binding. Taken together, these data reveal that Setdb1 promotes the persistence of DNA methylation in mESCs, likely reflecting one mechanism by which DNA methylation is maintained at LTR retrotransposons and imprinted genes during developmental stages when DNA methylation is reprogrammed.
- Published
- 2014
32. HP1 proteins safeguard embryonic stem cells
- Author
-
Kristoffer N. Jensen and Matthew C. Lorincz
- Subjects
0301 basic medicine ,Cell specific ,Multidisciplinary ,Heterochromatin ,Biology ,Embryonic stem cell ,DNA sequencing ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,Heterochromatin protein 1 ,Stem cell ,Peptide sequence ,Gene - Abstract
A complex that includes the protein HP1 binds to specific regulatory DNA sequences to promote local compaction of genomic regions and inhibit associated genes that drive differentiation of specific cell lineages. A complex that includes the protein HP1 binds to specific regulatory DNA sequences to promote local compaction of genomic regions and inhibit associated genes that drive differentiation of specific cell lineages.
- Published
- 2018
33. An Interactive Analysis and Exploration Tool for Epigenomic Data
- Author
-
Mohammad M. Karimi, Torsten Möller, Hamid Younesy, Steven J.M. Jones, Olivia Alder, Cydney B. Nielsen, Rebecca L. Cullum, and Matthew C. Lorincz
- Subjects
Computer science ,Interface (computing) ,SIGNAL (programming language) ,Data mining ,computer.software_genre ,Cluster analysis ,Computer Graphics and Computer-Aided Design ,computer ,Abstraction (linguistics) ,Visualization ,Domain (software engineering) ,Epigenomics - Abstract
In this design study, we present an analysis and abstraction of the data and tasks related to the domain of epigenomics, and the design and implementation of an interactive tool to facilitate data analysis and visualization in this domain. Epigenomic data can be grouped into subsets either by k-means clustering or by querying for combinations of presence or absence of signal (on/off) in different epigenomic experiments. These steps can easily be interleaved and the comparison of different workflows is explicitly supported. We took special care to contain the exponential expansion of possible on/off combinations by creating a novel querying interface. An interactive heat map facilitates the exploration and comparison of different clusters. We validated our iterative design by working closely with two groups of biologists on different biological problems. Both groups quickly found new insight into their data as well as claimed that our tool would save them several hours or days of work over using existing tools.
- Published
- 2013
34. Correction: hnRNP K Coordinates Transcriptional Silencing by SETDB1 in Embryonic Stem Cells
- Author
-
Mohammad M. Karimi, Leonard J. Foster, Matthew C. Lorincz, Vered Dulberg, Peter Thompson, Kyung-Mee Moon, and Carol Chen
- Subjects
0301 basic medicine ,Cancer Research ,lcsh:QH426-470 ,Statement (logic) ,Geo database ,Computational biology ,Accession number (bioinformatics) ,Biology ,Bioinformatics ,Embryonic stem cell ,Data availability ,03 medical and health sciences ,lcsh:Genetics ,030104 developmental biology ,0302 clinical medicine ,Genetics ,Gene silencing ,Molecular Biology ,030217 neurology & neurosurgery ,Genetics (clinical) ,Ecology, Evolution, Behavior and Systematics - Abstract
The Data Availability statement for this paper does not include a GEO accession number for the RNA-seq data presented. These data are available from the GEO database (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc={"type":"entrez-geo","attrs":{"text":"GSE84386","term_id":"84386"}}GSE84386). The complete statement is
- Published
- 2016
35. Long terminal repeats: from parasitic elements to building blocks of the transcriptional regulatory repertoire
- Author
-
Todd S. Macfarlan, Matthew C. Lorincz, and Peter Thompson
- Subjects
0301 basic medicine ,DNA Replication ,Gene Expression Regulation, Viral ,Transcription, Genetic ,viruses ,Endogenous retrovirus ,RNA polymerase II ,Retrotransposon ,Virus Replication ,Article ,03 medical and health sciences ,Animals ,Humans ,Promoter Regions, Genetic ,Molecular Biology ,Gene ,Genetics ,Regulation of gene expression ,biology ,Endogenous Retroviruses ,Terminal Repeat Sequences ,RNA-Directed DNA Polymerase ,Cell Biology ,Exaptation ,Chromatin Assembly and Disassembly ,Long terminal repeat ,030104 developmental biology ,Regulatory sequence ,DNA, Viral ,Host-Pathogen Interactions ,biology.protein ,RNA, Long Noncoding ,RNA Polymerase II - Abstract
The life cycle of endogenous retroviruses (ERVs), also called long terminal repeat (LTR) retrotransposons, begins with transcription by RNA polymerase II followed by reverse transcription and re-integration into the host genome. While most ERVs are relics of ancient integration events, "young" proviruses competent for retrotransposition-found in many mammals, but not humans-represent an ongoing threat to host fitness. As a consequence, several restriction pathways have evolved to suppress their activity at both transcriptional and post-transcriptional stages of the viral life cycle. Nevertheless, accumulating evidence has revealed that LTR sequences derived from distantly related ERVs have been exapted as regulatory sequences for many host genes in a wide range of cell types throughout mammalian evolution. Here, we focus on emerging themes from recent studies cataloging the diversity of ERV LTRs acting as important transcriptional regulatory elements in mammals and explore the molecular features that likely account for LTR exaptation in developmental and tissue-specific gene regulation.
- Published
- 2016
36. Dynamic and flexible H3K9me3 bridging via HP1β dimerization establishes a plastic state of condensed chromatin
- Author
-
Tuncay Baubec, Sarah Kreuz, Henning Urlaub, Carol Chen, Matthew C. Lorincz, Dirk Schwarzer, Wiebke H. Pohl, Szabolcs Soeroes, Bastian Zimmermann, Thomas Jenuwein, Miroslav Nikolov, Inti A. De La Rosa-Velázquez, Wolfgang Fischle, Aleksandar Chernev, Peter Walla, Bryan J. Wilkins, Heinz Neumann, Nils Kost, Hans Michael Zenn, Kyoko Hiragami-Hamada, University of Zurich, and Fischle, Wolfgang
- Subjects
Models, Molecular ,0301 basic medicine ,Chromosomal Proteins, Non-Histone ,General Physics and Astronomy ,Crystallography, X-Ray ,Chromodomain ,Histones ,Non-histone protein ,Heterochromatin ,Histone code ,Multidisciplinary ,Chromatin binding ,10226 Department of Molecular Mechanisms of Disease ,3100 General Physics and Astronomy ,Chromatin ,Nucleosomes ,embryonic structures ,Protein Binding ,endocrine system ,animal structures ,Science ,Blotting, Western ,Molecular Sequence Data ,Static Electricity ,1600 General Chemistry ,Biology ,Methylation ,Article ,General Biochemistry, Genetics and Molecular Biology ,Chromatin remodeling ,03 medical and health sciences ,Histone H1 ,1300 General Biochemistry, Genetics and Molecular Biology ,Cell Line, Tumor ,Humans ,Amino Acid Sequence ,ChIA-PET ,Sequence Homology, Amino Acid ,Lysine ,General Chemistry ,Molecular biology ,Kinetics ,030104 developmental biology ,Microscopy, Fluorescence ,Chromobox Protein Homolog 5 ,Biophysics ,570 Life sciences ,biology ,Protein Multimerization ,chromatin ,H3K9me3 - Abstract
Histone H3 trimethylation of lysine 9 (H3K9me3) and proteins of the heterochromatin protein 1 (HP1) family are hallmarks of heterochromatin, a state of compacted DNA essential for genome stability and long-term transcriptional silencing. The mechanisms by which H3K9me3 and HP1 contribute to chromatin condensation have been speculative and controversial. Here we demonstrate that human HP1β is a prototypic HP1 protein exemplifying most basal chromatin binding and effects. These are caused by dimeric and dynamic interaction with highly enriched H3K9me3 and are modulated by various electrostatic interfaces. HP1β bridges condensed chromatin, which we postulate stabilizes the compacted state. In agreement, HP1β genome-wide localization follows H3K9me3-enrichment and artificial bridging of chromatin fibres is sufficient for maintaining cellular heterochromatic conformation. Overall, our findings define a fundamental mechanism for chromatin higher order structural changes caused by HP1 proteins, which might contribute to the plastic nature of condensed chromatin., Heterochromatin protein 1 (HP1), including HP1 α, β and γ, is a family of non-histone chromatin factors thought to be involved in chromatin organization. Here, the authors show that dimeric HP1β interacts dynamically with H3K9me3, a hallmark of heterochromatin, and bridges condensed chromatin.
- Published
- 2016
37. DNA Methylation and SETDB1/H3K9me3 Regulate Predominantly Distinct Sets of Genes, Retroelements, and Chimeric Transcripts in mESCs
- Author
-
Martin Hirst, Yoichi Shinkai, Irina A. Maksakova, Misha Bilenky, Matthew C. Lorincz, Danny Leung, Steven J.M. Jones, Jie Xin Tang, Mohammad M. Karimi, Preeti Goyal, and Dixie L. Mager
- Subjects
DNA (Cytosine-5-)-Methyltransferase 1 ,Methyltransferase ,Retroelements ,Transcription, Genetic ,Endogenous retrovirus ,Article ,DNA Methyltransferase 3A ,Histones ,03 medical and health sciences ,Histone H3 ,Mice ,0302 clinical medicine ,Genetics ,Animals ,DNA (Cytosine-5-)-Methyltransferases ,Protein Methyltransferases ,Gene ,Cells, Cultured ,Embryonic Stem Cells ,030304 developmental biology ,Regulation of gene expression ,Mice, Knockout ,0303 health sciences ,biology ,Lysine ,Endogenous Retroviruses ,Histone-Lysine N-Methyltransferase ,Cell Biology ,DNA Methylation ,Histone ,Gene Expression Regulation ,DNA methylation ,biology.protein ,DNMT1 ,Molecular Medicine ,030217 neurology & neurosurgery - Abstract
DNA methylation and histone H3 lysine 9 trimethylation (H3K9me3) play important roles in silencing of genes and retroelements. However, a comprehensive comparison of genes and repetitive elements repressed by these pathways has not been reported. Here we show that in mouse embryonic stem cells (mESCs), the genes up-regulated following deletion of the H3K9 methyltransferase Setdb1 are distinct from those de-repressed in mESC deficient in the DNA methyltransferases Dnmt1, Dnmt3a and Dnmt3b, with the exception of a small number of primarily germline-specific genes. Numerous endogenous retroviruses (ERVs) lose H3K9me3 and are concomitantly de-repressed exclusively in SETDB1 knockout mESCs. Strikingly, ~15% of up-regulated genes are induced in association with de-repression of promoter proximal ERVs, half in the context of “chimaeric” transcripts that initiate within these retroelements and splice to genic exons. Thus, SETDB1 plays a previously unappreciated yet critical role in inhibiting aberrant gene transcription by suppressing the expression of proximal ERVs.
- Published
- 2011
- Full Text
- View/download PDF
38. Lysine methyltransferase G9a is required for de novo DNA methylation and the establishment, but not the maintenance, of proviral silencing
- Author
-
Dixie L. Mager, Kevin Dong, Danny Leung, Makoto Tachibana, Matthew C. Lorincz, Ruth Appanah, Yoichi Shinkai, Fabio M.V. Rossi, Preeti Goyal, Bernhard Lehnertz, Irina A. Maksakova, and Sandra Lee
- Subjects
Chromatin Immunoprecipitation ,Methyltransferase ,Blotting, Western ,Genetic Vectors ,Endogenous retrovirus ,Mice ,Retrovirus ,Proviruses ,Murine leukemia virus ,Animals ,Gene silencing ,DNA (Cytosine-5-)-Methyltransferases ,Gene Silencing ,Embryonic Stem Cells ,Mice, Knockout ,Multidisciplinary ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Endogenous Retroviruses ,Histone-Lysine N-Methyltransferase ,Methylation ,DNA Methylation ,Biological Sciences ,Flow Cytometry ,biology.organism_classification ,Molecular biology ,DNA methylation ,Moloney murine leukemia virus ,Chromatin immunoprecipitation - Abstract
Methylation on lysine 9 of histone H3 (H3K9me) and DNA methylation play important roles in the transcriptional silencing of specific genes and repetitive elements. Both marks are detected on class I and II endogenous retroviruses (ERVs) in murine embryonic stem cells (mESCs). Recently, we reported that the H3K9-specific lysine methyltransferase (KMTase) Eset/Setdb1/KMT1E is required for H3K9me3 and the maintenance of silencing of ERVs in mESCs. In contrast, G9a/Ehmt2/KMT1C is dispensable, despite the fact that this KMTase is required for H3K9 dimethylation (H3K9me2) and efficient DNA methylation of these retroelements. Transcription of the exogenous retrovirus (XRV) Moloney murine leukemia virus is rapidly extinguished after integration in mESCs, concomitant with de novo DNA methylation. However, the role that H3K9 KMTases play in this process has not been addressed. Here, we demonstrate that G9a, but not Suv39h1 or Suv39h2, is required for silencing of newly integrated Moloney murine leukemia virus–based vectors in mESCs. The silencing defect in G9a −/− cells is accompanied by a reduction of H3K9me2 at the proviral LTR, indicating that XRVs are direct targets of G9a. Furthermore, de novo DNA methylation of newly integrated proviruses is impaired in the G9a −/− line, phenocopying proviral DNA methylation and silencing defects observed in Dnmt3a-deficient mESCs. Once established, however, maintenance of silencing of XRVs, like ERVs, is dependent exclusively on the KMTase Eset. Taken together, these observations reveal that in mESCs, the H3K9 KMTase G9a is required for the establishment, but not for the maintenance, of silencing of newly integrated proviruses.
- Published
- 2011
39. DNA methylation in ES cells requires the lysine methyltransferase G9a but not its catalytic activity
- Author
-
Makoto Tachibana, Yoichi Shinkai, Dixie L. Mager, Hao W Yang, Irina A. Maksakova, Lucia L. Lam, Dirk Schübeler, Ruth Appanah, Fabio Mohn, Kevin Dong, Danny Leung, Matthew C. Lorincz, and Sandra Lee
- Subjects
Chromosomal Proteins, Non-Histone ,Mice, Transgenic ,Biology ,Article ,Catalysis ,Gene Expression Regulation, Enzymologic ,General Biochemistry, Genetics and Molecular Biology ,Histones ,Mice ,Epigenetics of physical exercise ,Histone methylation ,Animals ,Epigenetics ,Molecular Biology ,RNA-Directed DNA Methylation ,Embryonic Stem Cells ,Epigenomics ,Models, Genetic ,General Immunology and Microbiology ,General Neuroscience ,EZH2 ,Histone-Lysine N-Methyltransferase ,Methyltransferases ,DNA Methylation ,Molecular biology ,Mice, Inbred C57BL ,Chromobox Protein Homolog 5 ,Histone methyltransferase ,DNA methylation ,Mice, Inbred CBA ,CpG Islands - Abstract
Histone H3K9 methylation is required for DNA methylation and silencing of repetitive elements in plants and filamentous fungi. In mammalian cells however, deletion of the H3K9 histone methyltransferases (HMTases) Suv39h1 and Suv39h2 does not affect DNA methylation of the endogenous retrovirus murine leukaemia virus, indicating that H3K9 methylation is dispensable for DNA methylation of retrotransposons, or that a different HMTase is involved. We demonstrate that embryonic stem (ES) cells lacking the H3K9 HMTase G9a show a significant reduction in DNA methylation of retrotransposons, major satellite repeats and densely methylated CpG-rich promoters. Surprisingly, demethylated retrotransposons remain transcriptionally silent in G9a(-/-) cells, and show only a modest decrease in H3K9me2 and no decrease in H3K9me3 or HP1alpha binding, indicating that H3K9 methylation per se is not the relevant trigger for DNA methylation. Indeed, introduction of catalytically inactive G9a transgenes partially 'rescues' the DNA methylation defect observed in G9a(-/-) cells. Taken together, these observations reveal that H3K9me3 and HP1alpha recruitment to retrotransposons occurs independent of DNA methylation in ES cells and that G9a promotes DNA methylation independent of its HMTase activity.
- Published
- 2008
40. Systematic Identification of Factors for Provirus Silencing in Embryonic Stem Cells
- Author
-
Chadi A. El Farran, Hao Fei Wang, Yuin-Han Loh, Jayantha Gunaratne, James J. Collins, Matthew C. Lorincz, Yat Li, Vinay Tergaonkar, Lingyi Chen, Sharon Schlesinger, Tit Meng Lim, Stephen P. Goff, Frederic Bard, Hongchao Guo, Tao Yu, Hu Li, Germaine Yen Lin Goh, Yu Fen Samantha Seah, Hai Tong Fang, George Q. Daley, Suat Peng Neo, Bin Xia Yang, Massachusetts Institute of Technology. Department of Biological Engineering, and Collins, James J.
- Subjects
Genetics ,0303 health sciences ,TRIM28 ,Biochemistry, Genetics and Molecular Biology(all) ,Endogenous Retroviruses ,SUMO protein ,Endogenous retrovirus ,Biology ,Provirus ,Mi-2/NuRD complex ,General Biochemistry, Genetics and Molecular Biology ,Article ,Chromatin ,03 medical and health sciences ,0302 clinical medicine ,Proviruses ,Animals ,Epigenetics ,Psychological repression ,030217 neurology & neurosurgery ,Embryonic Stem Cells ,030304 developmental biology - Abstract
SummaryEmbryonic stem cells (ESCs) repress the expression of exogenous proviruses and endogenous retroviruses (ERVs). Here, we systematically dissected the cellular factors involved in provirus repression in embryonic carcinomas (ECs) and ESCs by a genome-wide siRNA screen. Histone chaperones (Chaf1a/b), sumoylation factors (Sumo2/Ube2i/Sae1/Uba2/Senp6), and chromatin modifiers (Trim28/Eset/Atf7ip) are key determinants that establish provirus silencing. RNA-seq analysis uncovered the roles of Chaf1a/b and sumoylation modifiers in the repression of ERVs. ChIP-seq analysis demonstrates direct recruitment of Chaf1a and Sumo2 to ERVs. Chaf1a reinforces transcriptional repression via its interaction with members of the NuRD complex (Kdm1a, Hdac1/2) and Eset, while Sumo2 orchestrates the provirus repressive function of the canonical Zfp809/Trim28/Eset machinery by sumoylation of Trim28. Our study reports a genome-wide atlas of functional nodes that mediate proviral silencing in ESCs and illuminates the comprehensive, interconnected, and multi-layered genetic and epigenetic mechanisms by which ESCs repress retroviruses within the genome.
- Published
- 2015
41. On the role of H3.3 in retroviral silencing
- Author
-
Matthew C. Lorincz, Geoffrey J. Faulkner, Warren Wu, Keiko Ozato, Brenda Wu, Mohammad M. Karimi, Rita Rebollo, Dixie L. Mager, Todd S. Macfarlan, Adam D. Ewing, Mahesh Bachu, Rui Kamada, Gernot Wolf, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Medical Genetics, University Hospital of North-Norway, Terry Fox Laboratory, BC Cancer Agency (BCCRC)-British Columbia Cancer Agency Research Centre, Mater Research Institute, and University of Queensland [Brisbane]
- Subjects
0301 basic medicine ,Multidisciplinary ,comparative analysis ,business.industry ,[SDV]Life Sciences [q-bio] ,MEDLINE ,Computational biology ,histone ,Biology ,analyse comparative ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Text mining ,pcr ,rétrovirus endogène ,Gene silencing ,business ,030217 neurology & neurosurgery - Abstract
On the role of H3.3 in retroviral silencing
- Published
- 2015
42. Ultra-low-input native ChIP-seq for rare cell populations
- Author
-
Julie Brind'Amour, Sheng Liu, Matthew Hudson, Carol Chen, Mohammad M Karimi, and Matthew C Lorincz
- Subjects
Cell culture ,biology.protein ,General Earth and Planetary Sciences ,Embryo ,Epigenetics ,Biology ,Chip ,Chromatin immunoprecipitation ,DNA sequencing ,General Environmental Science ,Cell biology ,Micrococcal nuclease ,Chromatin - Abstract
Combined chromatin immunoprecipitation and next generation sequencing (ChIP-seq) has become an extremely popular method to generate genome-wide epigenetic profiles from numerous cell lines and tissue types. Typical ChIP-seq experiments require large number of cells, making them illadapted to the study of rare cell populations. This procedure describes an ultra-low-input (ULI) micrococcal nuclease-based native ChIP (NChIP) and sequencing library construction method to generate genome-wide chromatin profiles from as few as 10 cells (Brind’Amour et al., 10.1038/ncomms7033). In addition, ULI-NChIP-seq has been validated in vivo, by generation of H3K9me3 and H3K27me3 profiles from E13.5 primordial germ cells isolated from single embryos (Liu, Brind’Amour et al., 10.1101/gad.244848.114).
- Published
- 2015
43. Setdb1 is required for germline development and silencing of H3K9me3-marked endogenous retroviruses in primordial germ cells
- Author
-
Hiroyuki Sasaki, Louis Lefebvre, Yoichi Shinkai, Mohammad M. Karimi, Aaron B. Bogutz, Sheng Liu, Julie Brind’Amour, Kenjiro Shirane, and Matthew C. Lorincz
- Subjects
Male ,Chromatin Immunoprecipitation ,Transcription, Genetic ,Endogenous retrovirus ,Retrotransposon ,Germline ,Gametogenesis ,Histones ,Gene Knockout Techniques ,Mice ,Genetics ,Animals ,Gene Silencing ,biology ,Endogenous Retroviruses ,Methylation ,Histone-Lysine N-Methyltransferase ,DNA Methylation ,Molecular biology ,Long terminal repeat ,Histone ,Germ Cells ,DNA methylation ,embryonic structures ,biology.protein ,Female ,Virus Activation ,Erratum ,Chromatin immunoprecipitation ,Gene Deletion ,Developmental Biology ,Research Paper - Abstract
Transcription of endogenous retroviruses (ERVs) is inhibited by de novo DNA methylation during gametogenesis, a process initiated after birth in oocytes and at approximately embryonic day 15.5 (E15.5) in prospermatogonia. Earlier in germline development, the genome, including most retrotransposons, is progressively demethylated. Young ERVK and ERV1 elements, however, retain intermediate methylation levels. As DNA methylation reaches a low point in E13.5 primordial germ cells (PGCs) of both sexes, we determined whether retrotransposons are marked by H3K9me3 and H3K27me3 using a recently developed low-input ChIP-seq (chromatin immunoprecipitation [ChIP] combined with deep sequencing) method. Although these repressive histone modifications are found predominantly on distinct genomic regions in E13.5 PGCs, they concurrently mark partially methylated long terminal repeats (LTRs) and LINE1 elements. Germline-specific conditional knockout of the H3K9 methyltransferase SETDB1 yields a decrease of both marks and DNA methylation at H3K9me3-enriched retrotransposon families. Strikingly, Setdb1 knockout E13.5 PGCs show concomitant derepression of many marked ERVs, including intracisternal A particle (IAP), ETn, and ERVK10C elements, and ERV-proximal genes, a subset in a sex-dependent manner. Furthermore, Setdb1 deficiency is associated with a reduced number of male E13.5 PGCs and postnatal hypogonadism in both sexes. Taken together, these observations reveal that SETDB1 is an essential guardian against proviral expression prior to the onset of de novo DNA methylation in the germline.
- Published
- 2015
44. hnRNP K Coordinates Transcriptional Silencing by SETDB1 in Embryonic Stem Cells
- Author
-
Carol Chen, Kyung Mee Moon, Matthew C. Lorincz, Mohammad M. Karimi, Vered Dulberg, Peter Thompson, and Leonard J. Foster
- Subjects
Cancer Research ,Small interfering RNA ,lcsh:QH426-470 ,Retroelements ,Transcription, Genetic ,SUMO protein ,Endogenous retrovirus ,Biology ,environment and public health ,Heterogeneous-Nuclear Ribonucleoprotein K ,Mice ,Transcription (biology) ,Genetics ,Silencer Elements, Transcriptional ,Gene silencing ,Animals ,Gene Silencing ,RNA, Small Interfering ,Molecular Biology ,Genetics (clinical) ,Ecology, Evolution, Behavior and Systematics ,Embryonic Stem Cells ,Mice, Knockout ,Endogenous Retroviruses ,Sumoylation ,Correction ,Histone-Lysine N-Methyltransferase ,DNA Methylation ,Molecular biology ,Embryonic stem cell ,Chromatin ,lcsh:Genetics ,Germ Cells ,DNA methylation ,Research Article - Abstract
Retrotransposition of endogenous retroviruses (ERVs) poses a substantial threat to genome stability. Transcriptional silencing of a subset of these parasitic elements in early mouse embryonic and germ cell development is dependent upon the lysine methyltransferase SETDB1, which deposits H3K9 trimethylation (H3K9me3) and the co-repressor KAP1, which binds SETDB1 when SUMOylated. Here we identified the transcription co-factor hnRNP K as a novel binding partner of the SETDB1/KAP1 complex in mouse embryonic stem cells (mESCs) and show that hnRNP K is required for ERV silencing. RNAi-mediated knockdown of hnRNP K led to depletion of H3K9me3 at ERVs, concomitant with de-repression of proviral reporter constructs and specific ERV subfamilies, as well as a cohort of germline-specific genes directly targeted by SETDB1. While hnRNP K recruitment to ERVs is dependent upon KAP1, SETDB1 binding at these elements requires hnRNP K. Furthermore, an intact SUMO conjugation pathway is necessary for SETDB1 recruitment to proviral chromatin and depletion of hnRNP K resulted in reduced SUMOylation at ERVs. Taken together, these findings reveal a novel regulatory hierarchy governing SETDB1 recruitment and in turn, transcriptional silencing in mESCs., Author Summary Retroelements, including endogenous retroviruses (ERVs), pose a significant threat to genome stability. In mouse embryonic stem (ES) cells, the enzyme SETDB1 safeguards the genome against transcription of specific ERVs by depositing a repressive mark H3K9 trimethylation (H3K9me3). Although SETDB1 is recruited to ERVs by its binding partner KAP1, the molecular basis of this silencing pathway is not clear. Using biochemical and genetic approaches, we identified hnRNP K as a novel component of this silencing pathway that facilitates the recruitment of SETDB1 to ERVs to promote their repression. HnRNP K binds to ERV sequences via KAP1 and subsequently promotes SETDB1 binding. Together, our results reveal a novel function for hnRNP K in transcriptional silencing of ERVs and demonstrate a new regulatory mechanism governing the deposition of H3K9me3 by SETDB1 in ES cells.
- Published
- 2015
45. Primate-specific endogenous retrovirus-driven transcription defines naive-like stem cells
- Author
-
Tamás Raskó, Jichang Wang, Avazeh T. Ghanbarian, Gerald G. Schumann, Huiqiang Cai, Gangcai Xie, Zoltán Ivics, Zsuzsanna Izsvák, Matthew C. Lorincz, Wei Chen, Laurence D. Hurst, Manvendra K. Singh, Nina V. Fuchs, Daniel Besser, Alessandro Prigione, and Attila Szvetnik
- Subjects
Genetic Markers ,Homeobox protein NANOG ,Rex1 ,Induced Pluripotent Stem Cells ,Endogenous retrovirus ,Biology ,Stem cell marker ,03 medical and health sciences ,0302 clinical medicine ,Humans ,Induced pluripotent stem cell ,Cells, Cultured ,Embryonic Stem Cells ,030304 developmental biology ,Genetics ,0303 health sciences ,Multidisciplinary ,Gene Expression Profiling ,Endogenous Retroviruses ,Embryonic stem cell ,Cell biology ,Endothelial stem cell ,DNA Transposable Elements ,RNA, Long Noncoding ,Stem cell ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
Naive embryonic stem cells hold great promise for research and therapeutics as they have broad and robust developmental potential. While such cells are readily derived from mouse blastocysts it has not been possible to isolate human equivalents easily1, 2, although human naive-like cells have been artificially generated (rather than extracted) by coercion of human primed embryonic stem cells by modifying culture conditions2, 3, 4 or through transgenic modification5. Here we show that a sub-population within cultures of human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) manifests key properties of naive state cells. These naive-like cells can be genetically tagged, and are associated with elevated transcription of HERVH, a primate-specific endogenous retrovirus. HERVH elements provide functional binding sites for a combination of naive pluripotency transcription factors, including LBP9, recently recognized as relevant to naivety in mice6. LBP9–HERVH drives hESC-specific alternative and chimaeric transcripts, including pluripotency-modulating long non-coding RNAs. Disruption of LBP9, HERVH and HERVH-derived transcripts compromises self-renewal. These observations define HERVH expression as a hallmark of naive-like hESCs, and establish novel primate-specific transcriptional circuitry regulating pluripotency.
- Published
- 2014
- Full Text
- View/download PDF
46. An ultra-low-input native ChIP-seq protocol for genome-wide profiling of rare cell populations
- Author
-
Mohammad M. Karimi, Carol Chen, Julie Brind’Amour, Matthew C. Lorincz, Matthew Hudson, and Sheng Liu
- Subjects
Male ,Chromatin Immunoprecipitation ,Sequence analysis ,General Physics and Astronomy ,Computational biology ,Cell Separation ,Biology ,Bioinformatics ,Genome ,Polymerase Chain Reaction ,General Biochemistry, Genetics and Molecular Biology ,Histones ,Mice ,Sex Factors ,Animals ,Micrococcal Nuclease ,Epigenetics ,Embryonic Stem Cells ,Genetic Association Studies ,Oligonucleotide Array Sequence Analysis ,Multidisciplinary ,High-Throughput Nucleotide Sequencing ,General Chemistry ,Sequence Analysis, DNA ,Flow Cytometry ,Embryonic stem cell ,Histone ,Cell culture ,biology.protein ,Female ,Artifacts ,Chromatin immunoprecipitation ,Micrococcal nuclease - Abstract
Combined chromatin immunoprecipitation and next-generation sequencing (ChIP-seq) has enabled genome-wide epigenetic profiling of numerous cell lines and tissue types. A major limitation of ChIP-seq, however, is the large number of cells required to generate high-quality data sets, precluding the study of rare cell populations. Here, we present an ultra-low-input micrococcal nuclease-based native ChIP (ULI-NChIP) and sequencing method to generate genome-wide histone mark profiles with high resolution from as few as 10(3) cells. We demonstrate that ULI-NChIP-seq generates high-quality maps of covalent histone marks from 10(3) to 10(6) embryonic stem cells. Subsequently, we show that ULI-NChIP-seq H3K27me3 profiles generated from E13.5 primordial germ cells isolated from single male and female embryos show high similarity to recent data sets generated using 50-180 × more material. Finally, we identify sexually dimorphic H3K27me3 enrichment at specific genic promoters, thereby illustrating the utility of this method for generating high-quality and -complexity libraries from rare cell populations.
- Published
- 2014
47. Methylation-Mediated Proviral Silencing Is Associated with MeCP2 Recruitment and Localized Histone H3 Deacetylation
- Author
-
Matthew C. Lorincz, Dirk Schübeler, and Mark Groudine
- Subjects
Transcription, Genetic ,Chromosomal Proteins, Non-Histone ,Methyl-CpG-Binding Protein 2 ,Histone Deacetylases ,Cell Line ,Histones ,Histone H4 ,Mice ,Viral Proteins ,Histone H3 ,Proviruses ,Animals ,Gene Silencing ,Molecular Biology ,Transcriptional Regulation ,Binding Sites ,Integrases ,biology ,Endogenous Retroviruses ,Cell Biology ,DNA Methylation ,Provirus ,Histone H3 deacetylation ,Molecular biology ,Clone Cells ,DNA-Binding Proteins ,Repressor Proteins ,Histone ,DNA, Viral ,Gene Targeting ,DNA methylation ,biology.protein ,CpG Islands ,Histone deacetylase activity ,Moloney murine leukemia virus ,Chromatin immunoprecipitation - Abstract
The majority of 5-methylcytosine in mammalian DNA resides in endogenous transposable elements and is associated with the transcriptional silencing of these parasitic elements. Methylation also plays an important role in the silencing of exogenous retroviruses. One of the difficulties inherent in the study of proviral silencing is that the sites in which proviruses randomly integrate influence the probability of de novo methylation and expression. In order to compare methylated and unmethylated proviruses at the same genomic site, we used a recombinase-based targeting approach to introduce an in vitro methylated or unmethylated Moloney murine leukemia-based provirus in MEL cells. The methylated and unmethylated states are maintained in vivo, with the exception of the initially methylated proviral enhancer, which becomes demethylated in vivo. Although the enhancer is unmethylated and remodeled, the methylated provirus is transcriptionally silent. To further analyze the repressed state, histone acetylation status was determined by chromatin immunoprecipitation (ChIP) analyses, which revealed that localized histone H3 but not histone H4 hyperacetylation is inversely correlated with proviral methylation density. Since members of the methyl-CpG binding domain (MBD) family of proteins recruit histone deacetylase activity, these proteins may play a role in proviral repression. Interestingly, only MBD3 and MeCP2 are expressed in MEL cells. ChIPs with antibodies specific for these proteins revealed that only MeCP2 associates with the provirus in a methylation-dependent manner. Taken together, our results suggest that MeCP2 recruitment to a methylated provirus is sufficient for transcriptional silencing, despite the presence of a remodeled enhancer.
- Published
- 2001
48. Genomic Targeting of Methylated DNA: Influence of Methylation on Transcription, Replication, Chromatin Structure, and Histone Acetylation
- Author
-
Dirk Schübeler, Matthew C. Lorincz, Daniel M. Cimbora, Eric E. Bouhassira, Yong-Quing Feng, Agnes Telling, and Mark Groudine
- Subjects
DNA Replication ,Time Factors ,Transcription, Genetic ,Gene Expression ,Enhancer RNAs ,Biology ,Cell Line ,Histones ,Viral Proteins ,Epigenetics of physical exercise ,Genes, Reporter ,Histone methylation ,Animals ,Humans ,Histone code ,Transgenes ,Promoter Regions, Genetic ,Molecular Biology ,Epigenomics ,Cell Nucleus ,Integrases ,Pioneer factor ,Acetylation ,DNA ,DNA-Directed RNA Polymerases ,Cell Biology ,DNA Methylation ,Flow Cytometry ,Precipitin Tests ,Molecular biology ,Chromatin ,Artificial Gene Fusion ,Blotting, Southern ,Enhancer Elements, Genetic ,Gene Expression Regulation ,Histone methyltransferase ,Gene Targeting ,DNA methylation ,Plasmids - Abstract
We have developed a strategy to introduce in vitro-methylated DNA into defined chromosomal locations. Using this system, we examined the effects of methylation on transcription, chromatin structure, histone acetylation, and replication timing by targeting methylated and unmethylated constructs to marked genomic sites. At two sites, which support stable expression from an unmethylated enhancer-reporter construct, introduction of an in vitro-methylated but otherwise identical construct results in specific changes in transgene conformation and activity, including loss of the promoter DNase I-hypersensitive site, localized hypoacetyla- tion of histones H3 and H4 within the reporter gene, and a block to transcriptional initiation. Insertion of methylated constructs does not alter the early replication timing of the loci and does not result in de novo methylation of flanking genomic sequences. Methylation at the promoter and gene is stable over time, as is the repression of transcription. Surprisingly, sequences within the enhancer are demethylated, the hypersensitive site forms, and the enhancer is hyperacetylated. Nevertheless, the enhancer is unable to activate the methylated and hypoacetylated reporter. Our findings suggest that CpG methylation represses transcription by interfering with RNA polymerase initiation via a mechanism that involves localized histone deacetylation. This repression is dominant over a remodeled enhancer but neither results in nor requires region-wide changes in DNA replication or chromatin structure.
- Published
- 2000
49. Dynamic Analysis of Proviral Induction and De Novo Methylation: Implications for a Histone Deacetylase-Independent, Methylation Density-Dependent Mechanism of Transcriptional Repression
- Author
-
Matthew C. Lorincz, Mark Groudine, David I. K. Martin, Mark C. Walters, Dirk Schübeler, and Scott Goeke
- Subjects
Transcription, Genetic ,Green Fluorescent Proteins ,Biology ,Transfection ,Histone Deacetylases ,Mice ,Proviruses ,Histone methylation ,Histone H2A ,Tumor Cells, Cultured ,Animals ,Humans ,Sulfites ,Molecular Biology ,DNA Primers ,Epigenomics ,Transcriptional Regulation ,EZH2 ,Cell Biology ,DNA Methylation ,Molecular biology ,Recombinant Proteins ,Luminescent Proteins ,Gene Expression Regulation ,Histone methyltransferase ,DNA methylation ,Azacitidine ,Histone deacetylase activity ,Histone deacetylase ,Moloney murine leukemia virus ,Dinucleoside Phosphates - Abstract
Methylation of cytosines in the CpG dinucleotide is generally associated with transcriptional repression in mammalian cells, and recent findings implicate histone deacetylation in methylation-mediated repression. Analyses of histone acetylation in in vitro-methylated transfected plasmids support this model; however, little is known about the relationships among de novo DNA methylation, transcriptional repression, and histone acetylation state. To examine these relationships in vivo, we have developed a novel approach that permits the isolation and expansion of cells harboring expressing or silent retroviruses. MEL cells were infected with a Moloney murine leukemia virus encoding the green fluorescent protein (GFP), and single-copy, silent proviral clones were treated weekly with the histone deacetylase inhibitor trichostatin A or the DNA methylation inhibitor 5-azacytidine. Expression was monitored concurrently by flow cytometry, allowing for repeated phenotypic analysis over time, and proviral methylation was determined by Southern blotting and bisulfite methylation mapping. Shortly after infection, proviral expression was inducible and the reporter gene and proviral enhancer showed a low density of methylation. Over time, the efficacy of drug induction diminished, coincident with the accumulation of methyl-CpGs across the provirus. Bisulfite analysis of cells in which 5-azacytidine treatment induced GFP expression revealed measurable but incomplete demethylation of the provirus. Repression could be overcome in late-passage clones only by pretreatment with 5-azacytidine followed by trichostatin A, suggesting that partial demethylation reestablishes the trichostatin-inducible state. These experiments reveal the presence of a silencing mechanism which acts on densely methylated DNA and appears to function independently of histone deacetylase activity.
- Published
- 2000
50. Detection and Isolation of Gene-Corrected Cells in Gaucher Disease Via a Fluorescence-Activated Cell Sorter Assay for Lysosomal Glucocerebrosidase Activity
- Author
-
John A. Barranger, Zhenjun Diwu, Matthew C. Lorincz, William G. Kerr, and Leonard A. Herzenberg
- Subjects
Genetic enhancement ,Immunology ,CD34 ,Bone Marrow Stem Cell ,Cell Biology ,Hematology ,Glucocerebroside ,Biology ,Biochemistry ,Molecular biology ,Transplantation ,Haematopoiesis ,Stem cell ,Glucocerebrosidase - Abstract
Gaucher disease type 1 results from the accumulation of glucocerebroside in macrophages of the reticuloendothelial system, as a consequence of a deficiency in glucocerebrosidase (GC) activity. Recent improvements in the methodologies for introducing foreign genes into bone marrow stem cells have prompted several groups to test the efficacy of gene transfer therapy as a curative treatment for Gaucher disease. Limitations of this approach include the potential for insufficient engraftment of gene-corrected cells and incomplete transduction of hematopoietic stem cells using retroviral gene transfer. Overcoming these obstacles may be critical in the case of treatment for Gaucher disease type 1, because GC transduced cells have not been shown to have a growth advantage over noncorrected cells. Here, we describe the development and application of a novel, fluorescence-activated cell sorter based assay that directly quantitates GC activity at the single cell level. In a test of this application, fibroblasts from a Gaucher patient were transduced, and high expressing cells sorted based on GC activity. Reanalysis of cultured sorted fibroblasts reveals that these cells maintain high levels of enzymatic activity, compared with the heterogeneous population from which they were sorted. The assay is sufficiently sensitive to distinguish GC activity found in Gaucher patient monocytes from that in normal controls. Furthermore, preliminary results indicate that increased GC activity can be detected in transduced, CD34+ enriched peripheral blood mononuclear cells isolated from a Gaucher patient. This method should be a useful addition to current gene therapy protocols as a means to quantitatively assess gene correction of relevant cell populations and potentially purify transduced cells for transplantation.
- Published
- 1997
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.