Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.198
Filter
Add more filters

Publication year range
1.
Cell ; 187(1): 110-129.e31, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38181737

ABSTRACT

X chromosome inactivation (XCI) serves as a paradigm for RNA-mediated regulation of gene expression, wherein the long non-coding RNA XIST spreads across the X chromosome in cis to mediate gene silencing chromosome-wide. In female naive human pluripotent stem cells (hPSCs), XIST is in a dispersed configuration, and XCI does not occur, raising questions about XIST's function. We found that XIST spreads across the X chromosome and induces dampening of X-linked gene expression in naive hPSCs. Surprisingly, XIST also targets specific autosomal regions, where it induces repressive chromatin changes and gene expression dampening. Thereby, XIST equalizes X-linked gene dosage between male and female cells while inducing differences in autosomes. The dispersed Xist configuration and autosomal localization also occur transiently during XCI initiation in mouse PSCs. Together, our study identifies XIST as the regulator of X chromosome dampening, uncovers an evolutionarily conserved trans-acting role of XIST/Xist, and reveals a correlation between XIST/Xist dispersal and autosomal targeting.


Subject(s)
Genes, X-Linked , RNA, Long Noncoding , X Chromosome , Animals , Female , Humans , Male , Mice , Gene Silencing , RNA, Long Noncoding/genetics , X Chromosome/genetics , Pluripotent Stem Cells/metabolism
2.
Cell ; 185(12): 2164-2183.e25, 2022 06 09.
Article in English | MEDLINE | ID: mdl-35597241

ABSTRACT

X inactivation (XCI) is triggered by upregulation of XIST, which coats the chromosome in cis, promoting formation of a heterochromatic domain (Xi). XIST role beyond initiation of XCI is only beginning to be elucidated. Here, we demonstrate that XIST loss impairs differentiation of human mammary stem cells (MaSCs) and promotes emergence of highly tumorigenic and metastatic carcinomas. On the Xi, XIST deficiency triggers epigenetic changes and reactivation of genes overlapping Polycomb domains, including Mediator subunit MED14. MED14 overdosage results in increased Mediator levels and hyperactivation of the MaSC enhancer landscape and transcriptional program, making differentiation less favorable. We further demonstrate that loss of XIST and Xi transcriptional instability is common among human breast tumors of poor prognosis. We conclude that XIST is a gatekeeper of human mammary epithelium homeostasis, thus unveiling a paradigm in the control of somatic cell identity with potential consequences for our understanding of gender-specific malignancies.


Subject(s)
Mediator Complex/metabolism , Neoplastic Stem Cells/metabolism , RNA, Long Noncoding/metabolism , Breast Neoplasms/metabolism , Cell Differentiation , Epigenesis, Genetic , Humans , RNA, Long Noncoding/genetics , X Chromosome Inactivation
3.
Cell ; 184(7): 1790-1803.e17, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33735607

ABSTRACT

The long non-coding RNA (lncRNA) XIST establishes X chromosome inactivation (XCI) in female cells in early development and thereafter is thought to be largely dispensable. Here, we show XIST is continually required in adult human B cells to silence a subset of X-linked immune genes such as TLR7. XIST-dependent genes lack promoter DNA methylation and require continual XIST-dependent histone deacetylation. XIST RNA-directed proteomics and CRISPRi screen reveal distinctive somatic cell-type-specific XIST complexes and identify TRIM28 that mediates Pol II pausing at promoters of X-linked genes in B cells. Single-cell transcriptome data of female patients with either systemic lupus erythematosus or COVID-19 infection revealed XIST dysregulation, reflected by escape of XIST-dependent genes, in CD11c+ atypical memory B cells (ABCs). XIST inactivation with TLR7 agonism suffices to promote isotype-switched ABCs. These results indicate cell-type-specific diversification and function for lncRNA-protein complexes and suggest expanded roles for XIST in sex-differences in biology and medicine.


Subject(s)
B-Lymphocytes/immunology , COVID-19 , Lupus Erythematosus, Systemic , RNA, Long Noncoding/physiology , Toll-Like Receptor 7/immunology , X Chromosome Inactivation , COVID-19/genetics , COVID-19/immunology , Cell Line , DNA Methylation , Female , Gene Silencing , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology
4.
Cell ; 184(25): 6174-6192.e32, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34813726

ABSTRACT

The lncRNA Xist forms ∼50 diffraction-limited foci to transcriptionally silence one X chromosome. How this small number of RNA foci and interacting proteins regulate a much larger number of X-linked genes is unknown. We show that Xist foci are locally confined, contain ∼2 RNA molecules, and nucleate supramolecular complexes (SMACs) that include many copies of the critical silencing protein SPEN. Aggregation and exchange of SMAC proteins generate local protein gradients that regulate broad, proximal chromatin regions. Partitioning of numerous SPEN molecules into SMACs is mediated by their intrinsically disordered regions and essential for transcriptional repression. Polycomb deposition via SMACs induces chromatin compaction and the increase in SMACs density around genes, which propagates silencing across the X chromosome. Our findings introduce a mechanism for functional nuclear compartmentalization whereby crowding of transcriptional and architectural regulators enables the silencing of many target genes by few RNA molecules.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Mitochondrial Proteins/metabolism , RNA, Long Noncoding/metabolism , X Chromosome/metabolism , Animals , Cell Line , Embryonic Stem Cells , Fibroblasts , Gene Silencing , Humans , Mice , Protein Binding , X Chromosome Inactivation
5.
Annu Rev Biochem ; 89: 255-282, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32259458

ABSTRACT

Facultative heterochromatin (fHC) concerns the developmentally regulated heterochromatinization of different regions of the genome and, in the case of the mammalian X chromosome and imprinted loci, of only one allele of a homologous pair. The formation of fHC participates in the timely repression of genes, by resisting strong trans activators. In this review, we discuss the molecular mechanisms underlying the establishment and maintenance of fHC in mammals using a mouse model. We focus on X-chromosome inactivation (XCI) as a paradigm for fHC but also relate it to genomic imprinting and homeobox (Hox) gene cluster repression. A vital role for noncoding transcription and/or transcripts emerges as the general principle of triggering XCI and canonical imprinting. However, other types of fHC are established through an unknown mechanism, independent of noncoding transcription (Hox clusters and noncanonical imprinting). We also extensively discuss polycomb-group repressive complexes (PRCs), which frequently play a vital role in fHC maintenance.


Subject(s)
Gene Expression Regulation, Developmental , Genomic Imprinting , Heterochromatin/metabolism , Polycomb-Group Proteins/genetics , X Chromosome Inactivation , X Chromosome/metabolism , Animals , Chromatin Assembly and Disassembly , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Female , Gene Silencing , Heterochromatin/chemistry , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histones/genetics , Histones/metabolism , Humans , Male , Oocytes/cytology , Oocytes/growth & development , Oocytes/metabolism , Polycomb-Group Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Spermatozoa/cytology , Spermatozoa/growth & development , Spermatozoa/metabolism , X Chromosome/chemistry
6.
Cell ; 182(1): 127-144.e23, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32502394

ABSTRACT

Before zygotic genome activation (ZGA), the quiescent genome undergoes reprogramming to transition into the transcriptionally active state. However, the mechanisms underlying euchromatin establishment during early embryogenesis remain poorly understood. Here, we show that histone H4 lysine 16 acetylation (H4K16ac) is maintained from oocytes to fertilized embryos in Drosophila and mammals. H4K16ac forms large domains that control nucleosome accessibility of promoters prior to ZGA in flies. Maternal depletion of MOF acetyltransferase leading to H4K16ac loss causes aberrant RNA Pol II recruitment, compromises the 3D organization of the active genomic compartments during ZGA, and causes downregulation of post-zygotically expressed genes. Germline depletion of histone deacetylases revealed that other acetyl marks cannot compensate for H4K16ac loss in the oocyte. Moreover, zygotic re-expression of MOF was neither able to restore embryonic viability nor onset of X chromosome dosage compensation. Thus, maternal H4K16ac provides an instructive function to the offspring, priming future gene activation.


Subject(s)
Histones/metabolism , Lysine/metabolism , Transcriptional Activation/genetics , Acetylation , Animals , Base Sequence , Chromosome Segregation/genetics , Conserved Sequence , Dosage Compensation, Genetic , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryo, Nonmammalian/metabolism , Evolution, Molecular , Female , Genome , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Male , Mammals/genetics , Mice , Mutation/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleosomes/metabolism , Oocytes/metabolism , Promoter Regions, Genetic , RNA Polymerase II/metabolism , X Chromosome/metabolism , Zygote/metabolism
7.
Cell ; 176(1-2): 182-197.e23, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30595450

ABSTRACT

During development, the precise relationships between transcription and chromatin modifications often remain unclear. We use the X chromosome inactivation (XCI) paradigm to explore the implication of chromatin changes in gene silencing. Using female mouse embryonic stem cells, we initiate XCI by inducing Xist and then monitor the temporal changes in transcription and chromatin by allele-specific profiling. This reveals histone deacetylation and H2AK119 ubiquitination as the earliest chromatin alterations during XCI. We show that HDAC3 is pre-bound on the X chromosome and that, upon Xist coating, its activity is required for efficient gene silencing. We also reveal that first PRC1-associated H2AK119Ub and then PRC2-associated H3K27me3 accumulate initially at large intergenic domains that can then spread into genes only in the context of histone deacetylation and gene silencing. Our results reveal the hierarchy of chromatin events during the initiation of XCI and identify key roles for chromatin in the early steps of transcriptional silencing.


Subject(s)
Chromatin/metabolism , X Chromosome Inactivation/genetics , X Chromosome Inactivation/physiology , Acetylation , Animals , Chromatin/genetics , Embryonic Stem Cells , Epigenomics/methods , Female , Gene Silencing , Histone Deacetylases/metabolism , Histones/metabolism , Mice , Polycomb-Group Proteins/metabolism , Protein Processing, Post-Translational , RNA, Long Noncoding/metabolism , Transcription, Genetic , Ubiquitination , X Chromosome/metabolism
8.
Annu Rev Biochem ; 87: 323-350, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29668306

ABSTRACT

X chromosome regulation represents a prime example of an epigenetic phenomenon where coordinated regulation of a whole chromosome is required. In flies, this is achieved by transcriptional upregulation of X chromosomal genes in males to equalize the gene dosage differences in females. Chromatin-bound proteins and long noncoding RNAs (lncRNAs) constituting a ribonucleoprotein complex known as the male-specific lethal (MSL) complex or the dosage compensation complex mediate this process. MSL complex members decorate the male X chromosome, and their absence leads to male lethality. The male X chromosome is also enriched with histone H4 lysine 16 acetylation (H4K16ac), indicating that the chromatin compaction status of the X chromosome also plays an important role in transcriptional activation. How the X chromosome is specifically targeted and how dosage compensation is mechanistically achieved are central questions for the field. Here, we review recent advances, which reveal a complex interplay among lncRNAs, the chromatin landscape, transcription, and chromosome conformation that fine-tune X chromosome gene expression.


Subject(s)
Dosage Compensation, Genetic , X Chromosome/genetics , Animals , Chromatin/genetics , Chromatin/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Epigenesis, Genetic , Female , Genes, X-Linked , Histone Code/genetics , Humans , Male , Models, Genetic , Models, Molecular , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , X Chromosome/metabolism
9.
Cell ; 174(2): 406-421.e25, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29887375

ABSTRACT

Mammalian chromosomes are partitioned into A/B compartments and topologically associated domains (TADs). The inactive X (Xi) chromosome, however, adopts a distinct conformation without evident compartments or TADs. Here, through exploration of an architectural protein, structural-maintenance-of-chromosomes hinge domain containing 1 (SMCHD1), we probe how the Xi is reconfigured during X chromosome inactivation. A/B compartments are first fused into "S1" and "S2" compartments, coinciding with Xist spreading into gene-rich domains. SMCHD1 then binds S1/S2 compartments and merges them to create a compartment-less architecture. Contrary to current views, TADs remain on the Xi but in an attenuated state. Ablating SMCHD1 results in a persistent S1/S2 organization and strengthening of TADs. Furthermore, loss of SMCHD1 causes regional defects in Xist spreading and erosion of heterochromatic silencing. We present a stepwise model for Xi folding, where SMCHD1 attenuates a hidden layer of Xi architecture to facilitate Xist spreading.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Chromosomes, Mammalian/chemistry , X Chromosome Inactivation , Alleles , Animals , Cell Line , Chromosomal Proteins, Non-Histone/genetics , Chromosomes, Mammalian/metabolism , DNA Methylation , Female , Heterochromatin/metabolism , Histones/genetics , Histones/metabolism , Male , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Principal Component Analysis , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
10.
Cell ; 171(1): 85-102.e23, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28867287

ABSTRACT

Chromatin modification and higher-order chromosome structure play key roles in gene regulation, but their functional interplay in controlling gene expression is elusive. We have discovered the machinery and mechanism underlying the dynamic enrichment of histone modification H4K20me1 on hermaphrodite X chromosomes during C. elegans dosage compensation and demonstrated H4K20me1's pivotal role in regulating higher-order chromosome structure and X-chromosome-wide gene expression. The structure and the activity of the dosage compensation complex (DCC) subunit DPY-21 define a Jumonji demethylase subfamily that converts H4K20me2 to H4K20me1 in worms and mammals. Selective inactivation of demethylase activity eliminates H4K20me1 enrichment in somatic cells, elevates X-linked gene expression, reduces X chromosome compaction, and disrupts X chromosome conformation by diminishing the formation of topologically associating domains (TADs). Unexpectedly, DPY-21 also associates with autosomes of germ cells in a DCC-independent manner to enrich H4K20me1 and trigger chromosome compaction. Our findings demonstrate the direct link between chromatin modification and higher-order chromosome structure in long-range regulation of gene expression.


Subject(s)
Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Gene Expression Regulation , X Chromosome/chemistry , Amino Acid Sequence , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Carrier Proteins/genetics , Dosage Compensation, Genetic , Embryo, Nonmammalian/metabolism , Jumonji Domain-Containing Histone Demethylases/chemistry , Jumonji Domain-Containing Histone Demethylases/metabolism , Models, Molecular , Mutation , Piperidines/metabolism , Sequence Alignment , Thiophenes/metabolism
11.
Mol Cell ; 82(1): 190-208.e17, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34932975

ABSTRACT

Developmental genes such as Xist, which initiates X chromosome inactivation, are controlled by complex cis-regulatory landscapes, which decode multiple signals to establish specific spatiotemporal expression patterns. Xist integrates information on X chromosome dosage and developmental stage to trigger X inactivation in the epiblast specifically in female embryos. Through a pooled CRISPR screen in differentiating mouse embryonic stem cells, we identify functional enhancer elements of Xist at the onset of random X inactivation. Chromatin profiling reveals that X-dosage controls the promoter-proximal region, while differentiation cues activate several distal enhancers. The strongest distal element lies in an enhancer cluster associated with a previously unannotated Xist-enhancing regulatory transcript, which we named Xert. Developmental cues and X-dosage are thus decoded by distinct regulatory regions, which cooperate to ensure female-specific Xist upregulation at the correct developmental time. With this study, we start to disentangle how multiple, functionally distinct regulatory elements interact to generate complex expression patterns in mammals.


Subject(s)
Enhancer Elements, Genetic , Genetic Loci , Mouse Embryonic Stem Cells/metabolism , Promoter Regions, Genetic , RNA, Long Noncoding/genetics , X Chromosome Inactivation , X Chromosome , Animals , Cell Differentiation , Cell Line , Female , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Mice, Transgenic , Up-Regulation
12.
Genes Dev ; 36(7-8): 483-494, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35483741

ABSTRACT

Genomic imprinting regulates parental origin-dependent monoallelic gene expression. It is mediated by either germline differential methylation of DNA (canonical imprinting) or oocyte-derived H3K27me3 (noncanonical imprinting) in mice. Depletion of Eed, an essential component of Polycomb repressive complex 2, results in genome-wide loss of H3K27me3 in oocytes, which causes loss of noncanonical imprinting (LOI) in embryos. Although Eed maternal KO (matKO) embryos show partial lethality after implantation, it is unknown whether LOI itself contributes to the developmental phenotypes of these embryos, which makes it unclear whether noncanonical imprinting is developmentally relevant. Here, by combinatorial matKO of Xist, a noncanonical imprinted gene whose LOI causes aberrant transient maternal X-chromosome inactivation (XCI) at preimplantation, we show that prevention of the transient maternal XCI greatly restores the development of Eed matKO embryos. Moreover, we found that the placentae of Eed matKO embryos are remarkably enlarged in a manner independent of Xist LOI. Heterozygous deletion screening of individual autosomal noncanonical imprinted genes suggests that LOI of the Sfmbt2 miRNA cluster chromosome 2 miRNA cluster (C2MC), solute carrier family 38 member 4 (Slc38a4), and Gm32885 contributes to the placental enlargement. Taken together, our study provides evidence that Xist imprinting sustains embryonic development and that autosomal noncanonical imprinting restrains placental overgrowth.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Animals , Embryonic Development/genetics , Female , Histones/metabolism , Mice , Placenta , Pregnancy , RNA, Long Noncoding/genetics , Repressor Proteins/genetics , X Chromosome Inactivation
13.
Mol Cell ; 81(9): 1970-1987.e9, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33725485

ABSTRACT

Depletion of architectural factors globally alters chromatin structure but only modestly affects gene expression. We revisit the structure-function relationship using the inactive X chromosome (Xi) as a model. We investigate cohesin imbalances by forcing its depletion or retention using degron-tagged RAD21 (cohesin subunit) or WAPL (cohesin release factor). Cohesin loss disrupts the Xi superstructure, unveiling superloops between escapee genes with minimal effect on gene repression. By contrast, forced cohesin retention markedly affects Xi superstructure, compromises spreading of Xist RNA-Polycomb complexes, and attenuates Xi silencing. Effects are greatest at distal chromosomal ends, where looping contacts with the Xist locus are weakened. Surprisingly, cohesin loss creates an Xi superloop, and cohesin retention creates Xi megadomains on the active X chromosome. Across the genome, a proper cohesin balance protects against aberrant inter-chromosomal interactions and tempers Polycomb-mediated repression. We conclude that a balance of cohesin eviction and retention regulates X inactivation and inter-chromosomal interactions across the genome.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Embryonic Stem Cells/metabolism , Gene Silencing , Polycomb-Group Proteins/metabolism , RNA, Long Noncoding/metabolism , X Chromosome Inactivation , X Chromosome , Animals , Cell Cycle Proteins/genetics , Cell Line , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Mice , Nucleic Acid Conformation , Polycomb-Group Proteins/genetics , Protein Conformation , Proteins/genetics , Proteins/metabolism , RNA, Long Noncoding/genetics , Structure-Activity Relationship , Cohesins
14.
Genes Dev ; 35(13-14): 1035-1054, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34168040

ABSTRACT

G9a is a histone methyltransferase responsible for the dimethylation of histone H3 at lysine 9 (H3K9me2). G9a plays key roles in transcriptional silencing of developmentally regulated genes, but its role in X-chromosome inactivation (XCI) has been under debate. Here, we uncover a female-specific function of G9a and demonstrate that deleting G9a has a disproportionate impact on the X chromosome relative to the rest of the genome. G9a deficiency causes a failure of XCI and female-specific hypersensitivity to drug inhibition of H3K9me2. We show that G9a interacts with Tsix and Xist RNAs, and that competitive inhibition of the G9a-RNA interaction recapitulates the XCI defect. During XCI, Xist recruits G9a to silence X-linked genes on the future inactive X. In parallel on the future Xa, Tsix recruits G9a to silence Xist in cis Thus, RNA tethers G9a for allele-specific targeting of the H3K9me2 modification and the G9a-RNA interaction is essential for XCI.


Subject(s)
Chromosomes, Human, X , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Methyltransferases , RNA, Long Noncoding , Female , Histones/metabolism , Humans , Methyltransferases/genetics , RNA, Long Noncoding/genetics , X Chromosome Inactivation/genetics
15.
Mol Cell ; 79(5): 836-845.e7, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32649884

ABSTRACT

The inactive X chromosome (Xi) is inherently susceptible to genomic aberrations. Replication stress (RS) has been proposed as an underlying cause, but the mechanisms that protect from Xi instability remain unknown. Here, we show that macroH2A1.2, an RS-protective histone variant enriched on the Xi, is required for Xi integrity and female survival. Mechanistically, macroH2A1.2 counteracts its structurally distinct and equally Xi-enriched alternative splice variant, macroH2A1.1. Comparative proteomics identified a role for macroH2A1.1 in alternative end joining (alt-EJ), which accounts for Xi anaphase defects in the absence of macroH2A1.2. Genomic instability was rescued by simultaneous depletion of macroH2A1.1 or alt-EJ factors, and mice deficient for both macroH2A1 variants harbor no overt female defects. Notably, macroH2A1 splice variant imbalance affected alt-EJ capacity also in tumor cells. Together, these findings identify macroH2A1 splicing as a modulator of genome maintenance that ensures Xi integrity and may, more broadly, predict DNA repair outcome in malignant cells.


Subject(s)
Alternative Splicing , DNA Repair , Epigenesis, Genetic , Genomic Instability , Histones/physiology , Anaphase , Animals , Cell Line , Chromosomal Instability , Chromosomes, Human, X , Female , Histones/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
Genes Dev ; 34(11-12): 733-744, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32482714

ABSTRACT

The X inactive-specific transcript (Xist) gene is the master regulator of X chromosome inactivation in mammals. Xist produces a long noncoding (lnc)RNA that accumulates over the entire length of the chromosome from which it is transcribed, recruiting factors to modify underlying chromatin and silence X-linked genes in cis Recent years have seen significant progress in identifying important functional elements in Xist RNA, their associated RNA-binding proteins (RBPs), and the downstream pathways for chromatin modification and gene silencing. In this review, we summarize progress in understanding both how these pathways function in Xist-mediated silencing and the complex interplay between them.


Subject(s)
Proteins/metabolism , RNA, Long Noncoding/metabolism , X Chromosome Inactivation/genetics , DNA-Binding Proteins/metabolism , Gene Silencing/physiology , Methyltransferases/metabolism , RNA, Long Noncoding/genetics , RNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Lamin B Receptor
17.
EMBO J ; 42(9): e112962, 2023 05 02.
Article in English | MEDLINE | ID: mdl-36929479

ABSTRACT

Human in vitro oogenesis provides a framework for clarifying the mechanism of human oogenesis. To create its benchmark, it is vital to promote in vitro oogenesis using a model physiologically close to humans. Here, we establish a foundation for in vitro oogenesis in cynomolgus (cy) monkeys (Macaca fascicularis): cy female embryonic stem cells harboring one active and one inactive X chromosome (Xa and Xi, respectively) differentiate robustly into primordial germ cell-like cells, which in xenogeneic reconstituted ovaries develop efficiently into oogonia and, remarkably, further into meiotic oocytes at the zygotene stage. This differentiation entails comprehensive epigenetic reprogramming, including Xi reprogramming, yet Xa and Xi remain epigenetically asymmetric with, as partly observed in vivo, incomplete Xi reactivation. In humans and monkeys, the Xi epigenome in pluripotent stem cells functions as an Xi-reprogramming determinant. We further show that developmental pathway over-activations with suboptimal up-regulation of relevant meiotic genes impede in vitro meiotic progression. Cy in vitro oogenesis exhibits critical homology with the human system, including with respect to bottlenecks, providing a salient model for advancing human in vitro oogenesis.


Subject(s)
Oocytes , Oogenesis , Animals , Female , Humans , Macaca fascicularis , Oogenesis/physiology , Ovary , Embryonic Stem Cells
18.
Development ; 151(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38813842

ABSTRACT

This Review elucidates the regulatory principles of random monoallelic expression by focusing on two well-studied examples: the X-chromosome inactivation regulator Xist and the olfactory receptor gene family. Although the choice of a single X chromosome or olfactory receptor occurs in different developmental contexts, common gene regulatory principles guide monoallelic expression in both systems. In both cases, an event breaks the symmetry between genetically and epigenetically identical copies of the gene, leading to the expression of one single random allele, stabilized through negative feedback control. Although many regulatory steps that govern the establishment and maintenance of monoallelic expression have been identified, key pieces of the puzzle are still missing. We provide an overview of the current knowledge and models for the monoallelic expression of Xist and olfactory receptors. We discuss their similarities and differences, and highlight open questions and approaches that could guide the study of other monoallelically expressed genes.


Subject(s)
Alleles , RNA, Long Noncoding , Receptors, Odorant , X Chromosome Inactivation , Animals , Humans , X Chromosome Inactivation/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Receptors, Odorant/genetics , Receptors, Odorant/metabolism , Gene Expression Regulation, Developmental
19.
Mol Cell ; 75(3): 523-537.e10, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31256989

ABSTRACT

Long noncoding RNAs (lncRNAs) cause Polycomb repressive complexes (PRCs) to spread over broad regions of the mammalian genome. We report that in mouse trophoblast stem cells, the Airn and Kcnq1ot1 lncRNAs induce PRC-dependent chromatin modifications over multi-megabase domains. Throughout the Airn-targeted domain, the extent of PRC-dependent modification correlated with intra-nuclear distance to the Airn locus, preexisting genome architecture, and the abundance of Airn itself. Specific CpG islands (CGIs) displayed characteristics indicating that they nucleate the spread of PRCs upon exposure to Airn. Chromatin environments surrounding Xist, Airn, and Kcnq1ot1 suggest common mechanisms of PRC engagement and spreading. Our data indicate that lncRNA potency can be tightly linked to lncRNA abundance and that within lncRNA-targeted domains, PRCs are recruited to CGIs via lncRNA-independent mechanisms. We propose that CGIs that autonomously recruit PRCs interact with lncRNAs and their associated proteins through three-dimensional space to nucleate the spread of PRCs in lncRNA-targeted domains.


Subject(s)
RNA, Long Noncoding/genetics , Animals , Chromatin/genetics , Chromatin Assembly and Disassembly , CpG Islands/genetics , Genome/genetics , Genomic Imprinting/genetics , Humans , Mice , Polycomb Repressive Complex 1/genetics , Promoter Regions, Genetic , Stem Cells/metabolism , Trophoblasts/metabolism
20.
Genes Dev ; 33(21-22): 1591-1612, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31601616

ABSTRACT

Genome rearrangements that occur during evolution impose major challenges on regulatory mechanisms that rely on three-dimensional genome architecture. Here, we developed a scaffolding algorithm and generated chromosome-length assemblies from Hi-C data for studying genome topology in three distantly related Drosophila species. We observe extensive genome shuffling between these species with one synteny breakpoint after approximately every six genes. A/B compartments, a set of large gene-dense topologically associating domains (TADs), and spatial contacts between high-affinity sites (HAS) located on the X chromosome are maintained over 40 million years, indicating architectural conservation at various hierarchies. Evolutionary conserved genes cluster in the vicinity of HAS, while HAS locations appear evolutionarily flexible, thus uncoupling functional requirement of dosage compensation from individual positions on the linear X chromosome. Therefore, 3D architecture is preserved even in scenarios of thousands of rearrangements highlighting its relevance for essential processes such as dosage compensation of the X chromosome.


Subject(s)
Drosophila/genetics , Evolution, Molecular , Gene Expression Regulation/genetics , Genome, Insect/genetics , Algorithms , Animals , Chromosomes, Insect/genetics , Conserved Sequence , Dosage Compensation, Genetic , Drosophila/embryology , Embryo, Nonmammalian
SELECTION OF CITATIONS
SEARCH DETAIL