Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Neurobiol Dis ; 198: 106538, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38789057

ABSTRACT

Aging is the main risk factor of cognitive neurodegenerative diseases such as Alzheimer's disease, with epigenome alterations as a contributing factor. Here, we compared transcriptomic/epigenomic changes in the hippocampus, modified by aging and by tauopathy, an AD-related feature. We show that the cholesterol biosynthesis pathway is severely impaired in hippocampal neurons of tauopathic but not of aged mice pointing to vulnerability of these neurons in the disease. At the epigenomic level, histone hyperacetylation was observed at neuronal enhancers associated with glutamatergic regulations only in the tauopathy. Lastly, a treatment of tau mice with the CSP-TTK21 epi-drug that restored expression of key cholesterol biosynthesis genes counteracted hyperacetylation at neuronal enhancers and restored object memory. As acetyl-CoA is the primary substrate of both pathways, these data suggest that the rate of the cholesterol biosynthesis in hippocampal neurons may trigger epigenetic-driven changes, that may compromise the functions of hippocampal neurons in pathological conditions.

2.
J Exp Clin Cancer Res ; 42(1): 301, 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-37957685

ABSTRACT

BACKGROUND: Inherited defects in the base-excision repair gene MBD4 predispose individuals to adenomatous polyposis and colorectal cancer, which is characterized by an accumulation of C > T transitions resulting from spontaneous deamination of 5'-methylcytosine. METHODS: Here, we have investigated the potential role of MBD4 in regulating DNA methylation levels using genome-wide transcriptome and methylome analyses. Additionally, we have elucidated its function through a series of in vitro experiments. RESULTS: Here we show that the protein MBD4 is required for DNA methylation maintenance and G/T mismatch repair. Transcriptome and methylome analyses reveal a genome-wide hypomethylation of promoters, gene bodies and repetitive elements in the absence of MBD4 in vivo. Methylation mark loss is accompanied by a broad transcriptional derepression phenotype affecting promoters and retroelements with low methylated CpG density. MBD4 in vivo forms a complex with the mismatch repair proteins (MMR), which exhibits high bi-functional glycosylase/AP-lyase endonuclease specific activity towards methylated DNA substrates containing a G/T mismatch. Experiments using recombinant proteins reveal that the association of MBD4 with the MMR protein MLH1 is required for this activity. CONCLUSIONS: Our data identify MBD4 as an enzyme specifically designed to repair deaminated 5-methylcytosines and underscores its critical role in safeguarding against methylation damage. Furthermore, it illustrates how MBD4 functions in normal and pathological conditions.


Subject(s)
DNA Repair , Retroelements , Humans , DNA Mismatch Repair , Recombinant Proteins/genetics , DNA Methylation , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism
3.
Cell Rep ; 42(11): 113363, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37924516

ABSTRACT

Super-enhancers (SEs) are stretches of enhancers ensuring a high level of expression of key genes associated with cell function. The identification of cancer-specific SE-driven genes is a powerful means for the development of innovative therapeutic strategies. Here, we identify a MITF/SOX10/TFIIH-dependent SE promoting the expression of BAHCC1 in a broad panel of melanoma cells. BAHCC1 is highly expressed in metastatic melanoma and is required for tumor engraftment, growth, and dissemination. Integrative genomics analyses reveal that BAHCC1 is a transcriptional regulator controlling expression of E2F/KLF-dependent cell-cycle and DNA-repair genes. BAHCC1 associates with BRG1-containing remodeling complexes at the promoters of these genes. BAHCC1 silencing leads to decreased cell proliferation and delayed DNA repair. Consequently, BAHCC1 deficiency cooperates with PARP inhibition to induce melanoma cell death. Our study identifies BAHCC1 as an SE-driven gene expressed in melanoma and demonstrates how its inhibition can be exploited as a therapeutic target.


Subject(s)
Melanoma , Humans , Cell Line, Tumor , Melanoma/pathology , Regulatory Sequences, Nucleic Acid , Genomic Instability , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Enhancer Elements, Genetic , Proteins/metabolism
4.
Nutrients ; 15(21)2023 Nov 05.
Article in English | MEDLINE | ID: mdl-37960344

ABSTRACT

Early-life exposure to high-fat diets (HF) can program metabolic and cognitive alterations in adult offspring. Although the hippocampus plays a crucial role in memory and metabolic homeostasis, few studies have reported the impact of maternal HF on this structure. We assessed the effects of maternal HF during lactation on physiological, metabolic, and cognitive parameters in young adult offspring mice. To identify early-programming mechanisms in the hippocampus, we developed a multi-omics strategy in male and female offspring. Maternal HF induced a transient increased body weight at weaning, and a mild glucose intolerance only in 3-month-old male mice with no change in plasma metabolic parameters in adult male and female offspring. Behavioral alterations revealed by a Barnes maze test were observed both in 6-month-old male and female mice. The multi-omics strategy unveiled sex-specific transcriptomic and proteomic modifications in the hippocampus of adult offspring. These studies that were confirmed by regulon analysis show that, although genes whose expression was modified by maternal HF were different between sexes, the main pathways affected were similar with mitochondria and synapses as main hippocampal targets of maternal HF. The effects of maternal HF reported here may help to better characterize sex-dependent molecular pathways involved in cognitive disorders and neurodegenerative diseases.


Subject(s)
Diet, High-Fat , Prenatal Exposure Delayed Effects , Animals , Mice , Female , Male , Humans , Diet, High-Fat/adverse effects , Obesity/etiology , Obesity/metabolism , Multiomics , Proteomics , Lactation , Hippocampus/metabolism , Maternal Nutritional Physiological Phenomena/physiology , Prenatal Exposure Delayed Effects/metabolism
5.
Prog Neurobiol ; 227: 102483, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37327984

ABSTRACT

Cytoplasmic mislocalization of the nuclear Fused in Sarcoma (FUS) protein is associated to amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Cytoplasmic FUS accumulation is recapitulated in the frontal cortex and spinal cord of heterozygous Fus∆NLS/+ mice. Yet, the mechanisms linking FUS mislocalization to hippocampal function and memory formation are still not characterized. Herein, we show that in these mice, the hippocampus paradoxically displays nuclear FUS accumulation. Multi-omic analyses showed that FUS binds to a set of genes characterized by the presence of an ETS/ELK-binding motifs, and involved in RNA metabolism, transcription, ribosome/mitochondria and chromatin organization. Importantly, hippocampal nuclei showed a decompaction of the neuronal chromatin at highly expressed genes and an inappropriate transcriptomic response was observed after spatial training of Fus∆NLS/+ mice. Furthermore, these mice lacked precision in a hippocampal-dependent spatial memory task and displayed decreased dendritic spine density. These studies shows that mutated FUS affects epigenetic regulation of the chromatin landscape in hippocampal neurons, which could participate in FTD/ALS pathogenic events. These data call for further investigation in the neurological phenotype of FUS-related diseases and open therapeutic strategies towards epigenetic drugs.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Animals , Mice , Amyotrophic Lateral Sclerosis/genetics , Chromatin/metabolism , Epigenesis, Genetic , Frontotemporal Dementia/genetics , Hippocampus/metabolism , Mutation , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism
6.
Cell Genom ; 3(4): 100295, 2023 Apr 12.
Article in English | MEDLINE | ID: mdl-37082140

ABSTRACT

Sea urchins are emblematic models in developmental biology and display several characteristics that set them apart from other deuterostomes. To uncover the genomic cues that may underlie these specificities, we generated a chromosome-scale genome assembly for the sea urchin Paracentrotus lividus and an extensive gene expression and epigenetic profiles of its embryonic development. We found that, unlike vertebrates, sea urchins retained ancestral chromosomal linkages but underwent very fast intrachromosomal gene order mixing. We identified a burst of gene duplication in the echinoid lineage and showed that some of these expanded genes have been recruited in novel structures (water vascular system, Aristotle's lantern, and skeletogenic micromere lineage). Finally, we identified gene-regulatory modules conserved between sea urchins and chordates. Our results suggest that gene-regulatory networks controlling development can be conserved despite extensive gene order rearrangement.

7.
Proc Natl Acad Sci U S A ; 120(4): e2210593120, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36656860

ABSTRACT

Mitotic entry correlates with the condensation of the chromosomes, changes in histone modifications, exclusion of transcription factors from DNA, and the broad downregulation of transcription. However, whether mitotic condensation influences transcription in the subsequent interphase is unknown. Here, we show that preventing one chromosome to condense during mitosis causes it to fail resetting of transcription. Rather, in the following interphase, the affected chromosome contains unusually high levels of the transcription machinery, resulting in abnormally high expression levels of genes in cis, including various transcription factors. This subsequently causes the activation of inducible transcriptional programs in trans, such as the GAL genes, even in the absence of the relevant stimuli. Thus, mitotic chromosome condensation exerts stringent control on interphase gene expression to ensure the maintenance of basic cellular functions and cell identity across cell divisions. Together, our study identifies the maintenance of transcriptional homeostasis during interphase as an unexpected function of mitosis and mitotic chromosome condensation.


Subject(s)
Chromatin , Chromosomes , Chromatin/genetics , Chromosomes/genetics , Chromosomes/metabolism , Interphase/genetics , Mitosis/genetics , Transcription Factors/metabolism
8.
J Biomed Sci ; 29(1): 107, 2022 Dec 20.
Article in English | MEDLINE | ID: mdl-36539812

ABSTRACT

BACKGROUND: Spinocerebellar ataxia type 7 (SCA7) is a neurodegenerative disorder that primarily affects the cerebellum and retina. SCA7 is caused by a polyglutamine expansion in the ATXN7 protein, a subunit of the transcriptional coactivator SAGA that acetylates histone H3 to deposit narrow H3K9ac mark at DNA regulatory elements of active genes. Defective histone acetylation has been presented as a possible cause for gene deregulation in SCA7 mouse models. However, the topography of acetylation defects at the whole genome level and its relationship to changes in gene expression remain to be determined. METHODS: We performed deep RNA-sequencing and chromatin immunoprecipitation coupled to high-throughput sequencing to examine the genome-wide correlation between gene deregulation and alteration of the active transcription marks, e.g. SAGA-related H3K9ac, CBP-related H3K27ac and RNA polymerase II (RNAPII), in a SCA7 mouse retinopathy model. RESULTS: Our analyses revealed that active transcription marks are reduced at most gene promoters in SCA7 retina, while a limited number of genes show changes in expression. We found that SCA7 retinopathy is caused by preferential downregulation of hundreds of highly expressed genes that define morphological and physiological identities of mature photoreceptors. We further uncovered that these photoreceptor genes harbor unusually broad H3K9ac profiles spanning the entire gene bodies and have a low RNAPII pausing. This broad H3K9ac signature co-occurs with other features that delineate superenhancers, including broad H3K27ac, binding sites for photoreceptor specific transcription factors and expression of enhancer-related non-coding RNAs (eRNAs). In SCA7 retina, downregulated photoreceptor genes show decreased H3K9 and H3K27 acetylation and eRNA expression as well as increased RNAPII pausing, suggesting that superenhancer-related features are altered. CONCLUSIONS: Our study thus provides evidence that distinctive epigenetic configurations underlying high expression of cell-type specific genes are preferentially impaired in SCA7, resulting in a defect in the maintenance of identity features of mature photoreceptors. Our results also suggest that continuous SAGA-driven acetylation plays a role in preserving post-mitotic neuronal identity.


Subject(s)
Retinal Diseases , Spinocerebellar Ataxias , Mice , Animals , Spinocerebellar Ataxias/genetics , Transcription Factors/genetics , Disease Models, Animal , Retinal Diseases/genetics , Gene Expression , Epigenesis, Genetic
9.
J Clin Invest ; 132(22)2022 11 15.
Article in English | MEDLINE | ID: mdl-36377661

ABSTRACT

Cisplatin is a potent chemotherapeutic drug that is widely used in the treatment of various solid cancers. However, its clinical effectiveness is strongly limited by frequent severe adverse effects, in particular nephrotoxicity and chemotherapy-induced peripheral neuropathy. Thus, there is an urgent medical need to identify novel strategies that limit cisplatin-induced toxicity. In the present study, we show that the FDA-approved adenosine A2A receptor antagonist istradefylline (KW6002) protected from cisplatin-induced nephrotoxicity and neuropathic pain in mice with or without tumors. Moreover, we also demonstrate that the antitumoral properties of cisplatin were not altered by istradefylline in tumor-bearing mice and could even be potentiated. Altogether, our results support the use of istradefylline as a valuable preventive approach for the clinical management of patients undergoing cisplatin treatment.


Subject(s)
Antineoplastic Agents , Neuralgia , Animals , Mice , Cisplatin/adverse effects , Purines/pharmacology , Neuralgia/chemically induced , Receptor, Adenosine A2A , Antineoplastic Agents/adverse effects
10.
Nat Commun ; 13(1): 4412, 2022 07 29.
Article in English | MEDLINE | ID: mdl-35906245

ABSTRACT

Gonadal sexual fate in mammals is determined during embryonic development and must be actively maintained in adulthood. In the mouse ovary, oestrogen receptors and FOXL2 protect ovarian granulosa cells from transdifferentiation into Sertoli cells, their testicular counterpart. However, the mechanism underlying their protective effect is unknown. Here, we show that TRIM28 is required to prevent female-to-male sex reversal of the mouse ovary after birth. We found that upon loss of Trim28, ovarian granulosa cells transdifferentiate to Sertoli cells through an intermediate cell type, different from gonadal embryonic progenitors. TRIM28 is recruited on chromatin in the proximity of FOXL2 to maintain the ovarian pathway and to repress testicular-specific genes. The role of TRIM28 in ovarian maintenance depends on its E3-SUMO ligase activity that regulates the sex-specific SUMOylation profile of ovarian-specific genes. Our study identifies TRIM28 as a key factor in protecting the adult ovary from the testicular pathway.


Subject(s)
Ovary , Sumoylation , Animals , Female , Male , Mammals/metabolism , Mice , Ovary/metabolism , Sertoli Cells/metabolism , Testis/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tripartite Motif-Containing Protein 28/genetics , Tripartite Motif-Containing Protein 28/metabolism
11.
Nucleic Acids Res ; 50(13): 7350-7366, 2022 07 22.
Article in English | MEDLINE | ID: mdl-35766398

ABSTRACT

The histone variant H3.3 is encoded by two distinct genes, H3f3a and H3f3b, exhibiting identical amino-acid sequence. H3.3 is required for spermatogenesis, but the molecular mechanism of its spermatogenic function remains obscure. Here, we have studied the role of each one of H3.3A and H3.3B proteins in spermatogenesis. We have generated transgenic conditional knock-out/knock-in (cKO/KI) epitope-tagged FLAG-FLAG-HA-H3.3B (H3.3BHA) and FLAG-FLAG-HA-H3.3A (H3.3AHA) mouse lines. We show that H3.3B, but not H3.3A, is required for spermatogenesis and male fertility. Analysis of the molecular mechanism unveils that the absence of H3.3B led to alterations in the meiotic/post-meiotic transition. Genome-wide RNA-seq reveals that the depletion of H3.3B in meiotic cells is associated with increased expression of the whole sex X and Y chromosomes as well as of both RLTR10B and RLTR10B2 retrotransposons. In contrast, the absence of H3.3B resulted in down-regulation of the expression of piRNA clusters. ChIP-seq experiments uncover that RLTR10B and RLTR10B2 retrotransposons, the whole sex chromosomes and the piRNA clusters are markedly enriched of H3.3. Taken together, our data dissect the molecular mechanism of H3.3B functions during spermatogenesis and demonstrate that H3.3B, depending on its chromatin localization, is involved in either up-regulation or down-regulation of expression of defined large chromatin regions.


Subject(s)
Histones , RNA, Small Interfering/metabolism , Retroelements , Spermatogenesis , Animals , Chromatin/genetics , Histones/genetics , Histones/metabolism , Male , Mice , Sex Chromosomes/metabolism
12.
J Clin Invest ; 132(12)2022 06 15.
Article in English | MEDLINE | ID: mdl-35536645

ABSTRACT

Caffeine is the most widely consumed psychoactive substance in the world. Strikingly, the molecular pathways engaged by its regular consumption remain unclear. We herein addressed the mechanisms associated with habitual (chronic) caffeine consumption in the mouse hippocampus using untargeted orthogonal omics techniques. Our results revealed that chronic caffeine exerts concerted pleiotropic effects in the hippocampus at the epigenomic, proteomic, and metabolomic levels. Caffeine lowered metabolism-related processes (e.g., at the level of metabolomics and gene expression) in bulk tissue, while it induced neuron-specific epigenetic changes at synaptic transmission/plasticity-related genes and increased experience-driven transcriptional activity. Altogether, these findings suggest that regular caffeine intake improves the signal-to-noise ratio during information encoding, in part through fine-tuning of metabolic genes, while boosting the salience of information processing during learning in neuronal circuits.


Subject(s)
Caffeine , Proteomics , Animals , Caffeine/metabolism , Caffeine/pharmacology , Hippocampus/metabolism , Learning , Mice , Neuronal Plasticity/physiology
13.
Nucleic Acids Res ; 50(3): 1351-1369, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35100417

ABSTRACT

Tight control of gene expression networks required for adipose tissue formation and plasticity is essential for adaptation to energy needs and environmental cues. However, the mechanisms that orchestrate the global and dramatic transcriptional changes leading to adipocyte differentiation remain to be fully unraveled. We investigated the regulation of nascent transcription by the sumoylation pathway during adipocyte differentiation using SLAMseq and ChIPseq. We discovered that the sumoylation pathway has a dual function in differentiation; it supports the initial downregulation of pre-adipocyte-specific genes, while it promotes the establishment of the mature adipocyte transcriptional program. By characterizing endogenous sumoylome dynamics in differentiating adipocytes by mass spectrometry, we found that sumoylation of specific transcription factors like PPARγ/RXR and their co-factors are associated with the transcription of adipogenic genes. Finally, using RXR as a model, we found that sumoylation may regulate adipogenic transcription by supporting the chromatin occurrence of transcription factors. Our data demonstrate that the sumoylation pathway supports the rewiring of transcriptional networks required for formation of functional adipocytes. This study also provides the scientists in the field of cellular differentiation and development with an in-depth resource of the dynamics of the SUMO-chromatin landscape, SUMO-regulated transcription and endogenous sumoylation sites during adipocyte differentiation.


Subject(s)
Adipogenesis , Sumoylation , Adipocytes/metabolism , Adipogenesis/genetics , Cell Differentiation/genetics , Chromatin/genetics , Chromatin/metabolism , Transcription Factors/metabolism
14.
Nat Commun ; 12(1): 5446, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34521844

ABSTRACT

EOMES and T-BET are related T-box transcription factors that control natural killer (NK) cell development. Here we demonstrate that EOMES and T-BET regulate largely distinct gene sets during this process. EOMES is dominantly expressed in immature NK cells and drives early lineage specification by inducing hallmark receptors and functions. By contrast, T-BET is dominant in mature NK cells, where it induces responsiveness to IL-12 and represses the cell cycle, likely through transcriptional repressors. Regardless, many genes with distinct functions are co-regulated by the two transcription factors. By generating two gene-modified mice facilitating chromatin immunoprecipitation of endogenous EOMES and T-BET, we show a strong overlap in their DNA binding targets, as well as extensive epigenetic changes during NK cell differentiation. Our data thus suggest that EOMES and T-BET may distinctly govern, via differential expression and co-factors recruitment, NK cell maturation by inserting partially overlapping epigenetic regulations.


Subject(s)
Cell Cycle/genetics , Cell Lineage/genetics , Killer Cells, Natural/immunology , T-Box Domain Proteins/genetics , Animals , Base Sequence , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , CD11b Antigen/genetics , CD11b Antigen/immunology , Cell Cycle/drug effects , Cell Cycle/immunology , Cell Differentiation , Cell Lineage/drug effects , Cell Lineage/immunology , Epigenesis, Genetic/immunology , Interleukin-12/pharmacology , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Protein Binding , Spleen/cytology , Spleen/immunology , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/immunology , Transcription, Genetic , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
15.
J Exp Med ; 218(10)2021 10 04.
Article in English | MEDLINE | ID: mdl-34459852

ABSTRACT

Our understanding of cell fate decisions in hematopoietic stem cells is incomplete. Here, we show that the transcription factor Helios is highly expressed in murine hematopoietic stem and progenitor cells (HSPCs), where it is required to suppress the separation of the platelet/megakaryocyte lineage from the HSPC pool. Helios acts mainly in quiescent cells, where it directly represses the megakaryocyte gene expression program in cells as early as the stem cell stage. Helios binding promotes chromatin compaction, notably at the regulatory regions of platelet-specific genes recognized by the Gata2 and Runx1 transcriptional activators, implicated in megakaryocyte priming. Helios null HSPCs are biased toward the megakaryocyte lineage at the expense of the lymphoid and partially resemble cells of aging animals. We propose that Helios acts as a guardian of HSPC pluripotency by continuously repressing the megakaryocyte fate, which in turn allows downstream lymphoid priming to take place. These results highlight the importance of negative and positive priming events in lineage commitment.


Subject(s)
DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/physiology , Megakaryocytes/physiology , Transcription Factors/metabolism , Animals , Cell Differentiation , DNA-Binding Proteins/genetics , Female , Gene Expression Profiling , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Lymphocytes/cytology , Lymphocytes/physiology , Male , Megakaryocytes/cytology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocytes/cytology , T-Lymphocytes/physiology , Transcription Factors/genetics
16.
Science ; 372(6549)2021 Jun 25.
Article in English | MEDLINE | ID: mdl-34324427

ABSTRACT

The Rett syndrome protein MeCP2 was described as a methyl-CpG-binding protein, but its exact function remains unknown. Here we show that mouse MeCP2 is a microsatellite binding protein that specifically recognizes hydroxymethylated CA repeats. Depletion of MeCP2 alters chromatin organization of CA repeats and lamina-associated domains and results in nucleosome accumulation on CA repeats and genome-wide transcriptional dysregulation. The structure of MeCP2 in complex with a hydroxymethylated CA repeat reveals a characteristic DNA shape, with considerably modified geometry at the 5-hydroxymethylcytosine, which is recognized specifically by Arg133, a key residue whose mutation causes Rett syndrome. Our work identifies MeCP2 as a microsatellite DNA binding protein that targets the 5hmC-modified CA-rich strand and maintains genome regions nucleosome-free, suggesting a role for MeCP2 dysfunction in Rett syndrome.


Subject(s)
Dinucleotide Repeats , Methyl-CpG-Binding Protein 2/metabolism , Microsatellite Repeats , Nucleosomes/metabolism , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/chemistry , 5-Methylcytosine/metabolism , Animals , Cells, Cultured , Chromatin/chemistry , Chromatin/metabolism , Chromatin/ultrastructure , Cytosine/chemistry , Cytosine/metabolism , DNA Methylation , Embryonic Stem Cells/metabolism , Fibroblasts , Frontal Lobe/metabolism , Methyl-CpG-Binding Protein 2/chemistry , Methyl-CpG-Binding Protein 2/genetics , Mice , Neurons/metabolism , Nucleic Acid Conformation , Oxidation-Reduction , Protein Binding , Protein Domains , Rett Syndrome/genetics , Rett Syndrome/metabolism , Transcription, Genetic
17.
Cell Metab ; 33(3): 513-530.e8, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33539777

ABSTRACT

Polycystic ovary syndrome (PCOS) is the most common reproductive and metabolic disorder affecting women of reproductive age. PCOS has a strong heritable component, but its pathogenesis has been unclear. Here, we performed RNA sequencing and genome-wide DNA methylation profiling of ovarian tissue from control and third-generation PCOS-like mice. We found that DNA hypomethylation regulates key genes associated with PCOS and that several of the differentially methylated genes are also altered in blood samples from women with PCOS compared with healthy controls. Based on this insight, we treated the PCOS mouse model with the methyl group donor S-adenosylmethionine and found that it corrected their transcriptomic, neuroendocrine, and metabolic defects. These findings show that the transmission of PCOS traits to future generations occurs via an altered landscape of DNA methylation and propose methylome markers as a possible diagnostic landmark for the condition, while also identifying potential candidates for epigenetic-based therapy.


Subject(s)
Epigenesis, Genetic , Polycystic Ovary Syndrome/genetics , Animals , Anti-Mullerian Hormone/pharmacology , Anti-Mullerian Hormone/therapeutic use , Case-Control Studies , DNA Methylation/drug effects , Disease Models, Animal , Female , Genetic Predisposition to Disease , Humans , Luteinizing Hormone/blood , Male , Mice , Mice, Inbred C57BL , Mixed Function Oxygenases/genetics , Ovary/metabolism , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/pathology , Prenatal Care , Proto-Oncogene Proteins/genetics , S-Adenosylmethionine/pharmacology , S-Adenosylmethionine/therapeutic use , Transcriptome/drug effects
18.
EMBO Rep ; 22(3): e51009, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33512761

ABSTRACT

Histone post-translational modifications (PTMs) are key players in chromatin regulation. The identification of novel histone acylations raises important questions regarding their role in transcription. In this study, we characterize the role of an acylation on the lateral surface of the histone octamer, H3K122 succinylation (H3K122succ), in chromatin function and transcription. Using chromatin succinylated at H3K122 in in vitro transcription assays, we show that the presence of H3K122succ is sufficient to stimulate transcription. In line with this, we found in our ChIP assays H3K122succ enriched on promoters of active genes and H3K122succ enrichment scaling with gene expression levels. Furthermore, we show that the co-activators p300/CBP can succinylate H3K122 and identify sirtuin 5 (SIRT5) as a new desuccinylase. By applying single molecule FRET assays, we demonstrate a direct effect of H3K122succ on nucleosome stability, indicating an important role for histone succinylation in modulating chromatin dynamics. Together, these data provide the first insights into the mechanisms underlying transcriptional regulation by H3K122succ.


Subject(s)
Histones , Nucleosomes , Chromatin/genetics , Gene Expression Regulation , Histones/genetics , Histones/metabolism , Nucleosomes/genetics , Protein Processing, Post-Translational
19.
PLoS One ; 16(1): e0246570, 2021.
Article in English | MEDLINE | ID: mdl-33513189

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0242211.].

20.
J Mol Biol ; 433(6): 166659, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33010306

ABSTRACT

Epigenetic modifications and nucleosome positioning play an important role in modulating gene expression. However, how the patterns of epigenetic modifications and nucleosome positioning are established around promoters is not well understood. Here, we have addressed these questions in a series of genome-wide experiments coupled to a novel bioinformatic analysis approach. Our data reveal a clear correlation between CpG density, promoter activity and accumulation of active or repressive histone marks. CGI boundaries define the chromatin promoter regions that will be epigenetically modified. CpG-rich promoters are targeted by histone modifications and histone variants, while CpG-poor promoters are regulated by DNA methylation. CGIs boundaries, but not transcriptional activity, are essential determinants of H2A.Z positioning in vicinity of the promoters, suggesting that the presence of H2A.Z is not related to transcriptional control. Accordingly, H2A.Z depletion has no impact on gene expression of arrested mouse embryonic fibroblasts. Therefore, the underlying DNA sequence, the promoter CpG density and, to a lesser extent, transcriptional activity, are key factors implicated in promoter chromatin architecture.


Subject(s)
CpG Islands , Epigenesis, Genetic , Epigenome , Histones/genetics , Promoter Regions, Genetic , Protein Processing, Post-Translational , Animals , Chromatin/metabolism , Chromatin/ultrastructure , Chromatin Assembly and Disassembly , Computational Biology/methods , DNA Methylation , Embryo, Mammalian , Fibroblasts/cytology , Fibroblasts/metabolism , Histones/chemistry , Histones/deficiency , Histones/metabolism , Mice , Mice, Knockout , Primary Cell Culture , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...