Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters










Publication year range
1.
Cell ; 187(3): 733-749.e16, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38306984

ABSTRACT

Autoimmune diseases disproportionately affect females more than males. The XX sex chromosome complement is strongly associated with susceptibility to autoimmunity. Xist long non-coding RNA (lncRNA) is expressed only in females to randomly inactivate one of the two X chromosomes to achieve gene dosage compensation. Here, we show that the Xist ribonucleoprotein (RNP) complex comprising numerous autoantigenic components is an important driver of sex-biased autoimmunity. Inducible transgenic expression of a non-silencing form of Xist in male mice introduced Xist RNP complexes and sufficed to produce autoantibodies. Male SJL/J mice expressing transgenic Xist developed more severe multi-organ pathology in a pristane-induced lupus model than wild-type males. Xist expression in males reprogrammed T and B cell populations and chromatin states to more resemble wild-type females. Human patients with autoimmune diseases displayed significant autoantibodies to multiple components of XIST RNP. Thus, a sex-specific lncRNA scaffolds ubiquitous RNP components to drive sex-biased immunity.


Subject(s)
Autoantibodies , Autoimmune Diseases , RNA, Long Noncoding , Animals , Female , Humans , Male , Mice , Autoantibodies/genetics , Autoimmune Diseases/genetics , Autoimmunity/genetics , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , X Chromosome/genetics , X Chromosome/metabolism , X Chromosome Inactivation , Sex Characteristics
2.
Epigenomes ; 7(4)2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37987303

ABSTRACT

For about 30 years, SPEN has been the subject of research in many different fields due to its variety of functions and its conservation throughout a wide spectrum of species, like worms, arthropods, and vertebrates. To date, 216 orthologues have been documented. SPEN had been studied for its role in gene regulation in the context of cell signaling, including the NOTCH or nuclear hormone receptor signaling pathways. More recently, SPEN has been identified as a major regulator of initiation of chromosome-wide gene silencing during X chromosome inactivation (XCI) in mammals, where its function remains to be fully understood. Dependent on the biological context, SPEN functions via mechanisms which include different domains. While some domains of SPEN are highly conserved in sequence and secondary structure, species-to-species differences exist that might lead to mechanistic differences. Initiation of XCI appears to be different between humans and mice, which raises additional questions about the extent of generalization of SPEN's function in XCI. In this review, we dissect the mechanism of SPEN in XCI. We discuss its subregions and domains, focusing on its role as a major regulator. We further highlight species-related research, specifically of mouse and human SPEN, with the aim to reveal and clarify potential species-to-species differences in SPEN's function.

3.
EMBO J ; 42(23): e113955, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37850882

ABSTRACT

Recent studies have reported the differentiation of pluripotent cells into oocytes in vitro. However, the developmental competence of in vitro-generated oocytes remains low. Here, we perform a comprehensive comparison of mouse germ cell development in vitro over all culture steps versus in vivo with the goal to understand mechanisms underlying poor oocyte quality. We show that the in vitro differentiation of primordial germ cells to growing oocytes and subsequent follicle growth is critical for competence for preimplantation development. Systematic transcriptome analysis of single oocytes that were subjected to different culture steps identifies genes that are normally upregulated during oocyte growth to be susceptible for misregulation during in vitro oogenesis. Many misregulated genes are Polycomb targets. Deregulation of Polycomb repression is therefore a key cause and the earliest defect known in in vitro oocyte differentiation. Conversely, structurally normal in vitro-derived oocytes fail at zygotic genome activation and show abnormal acquisition of 5-hydroxymethylcytosine on maternal chromosomes. Our data identify epigenetic regulation at an early stage of oogenesis limiting developmental competence and suggest opportunities for future improvements.


Subject(s)
Epigenesis, Genetic , Oocytes , Female , Animals , Mice , Ovarian Follicle , Oogenesis/genetics , Germ Cells
4.
PLoS Biol ; 20(3): e3001596, 2022 03.
Article in English | MEDLINE | ID: mdl-35353806

ABSTRACT

Hedgehog (HH) signaling is important for embryonic pattering and stem cell differentiation. The G protein-coupled receptor (GPCR) Smoothened (SMO) is the key HH signal transducer modulating both transcription-dependent and transcription-independent responses. We show that SMO protects naive mouse embryonic stem cells (ESCs) from dissociation-induced cell death. We exploited this SMO dependency to perform a genetic screen in haploid ESCs where we identify the Golgi proteins TMED2 and TMED10 as factors for SMO regulation. Super-resolution microscopy shows that SMO is normally retained in the endoplasmic reticulum (ER) and Golgi compartments, and we demonstrate that TMED2 binds to SMO, preventing localization to the plasma membrane. Mutation of TMED2 allows SMO accumulation at the plasma membrane, recapitulating early events after HH stimulation. We demonstrate the physiologic relevance of this interaction in neural differentiation, where TMED2 functions to repress HH signal strength. Identification of TMED2 as a binder and upstream regulator of SMO opens the way for unraveling the events in the ER-Golgi leading to HH signaling activation.


Subject(s)
Hedgehog Proteins , Receptors, G-Protein-Coupled , Animals , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Membrane Proteins , Mice , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/genetics , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Vesicular Transport Proteins
5.
Stem Cell Reports ; 17(1): 43-52, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34919812

ABSTRACT

Mammalian haploid cells have applications for genetic screening and substituting gametic genomes. Here, we characterize a culture system for obtaining haploid primordial germ cell-like cells (PGCLCs) from haploid mouse embryonic stem cells (ESCs). We find that haploid cells show predisposition for PGCLCs, whereas a large fraction of somatic cells becomes diploid. Characterization of the differentiating haploid ESCs (haESCs) reveals that Xist is activated from and colocalizes with the single X chromosome. This observation suggests that X chromosome inactivation (XCI) is initiated in haploid cells consistent with a model where autosomal blocking factors set a threshold for X-linked activators. We further find that Xist expression is lost at later timepoints in differentiation, which likely reflects the loss of X-linked activators. In vitro differentiation of haploid PGCLCs can be a useful approach for future studies of potential X-linked activators of Xist.


Subject(s)
Embryonic Stem Cells/metabolism , Germ Cells/metabolism , Haploidy , RNA, Long Noncoding/genetics , X Chromosome , Animals , Biomarkers , Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Gene Dosage , Gene Expression Regulation, Developmental , Germ Cells/cytology , Mice , X Chromosome Inactivation
6.
EMBO Rep ; 22(9): e52190, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34309165

ABSTRACT

The reactivation of X-linked genes is observed in some primary breast tumors. Two active X chromosomes are also observed in female embryonic stem cells (ESCs), but whether double doses of X-linked genes affect DNA repair efficiency remains unclear. Here, we establish isogenic female/male ESCs and show that the female ESCs are more sensitive to camptothecin and have lower gene targeting efficiency than male ESCs, suggesting that homologous recombination (HR) efficiency is reduced in female ESCs. We also generate Xist-inducible female ESCs and show that the lower HR efficiency is restored when X chromosome inactivation is induced. Finally, we assess the X-linked genes with a role in DNA repair and find that Brcc3 is one of the genes involved in a network promoting proper HR. Our findings link the double doses of X-linked genes with lower DNA repair activity, and this may have relevance for common diseases in female patients, such as breast cancer.


Subject(s)
Embryonic Stem Cells , RNA, Long Noncoding , Female , Homologous Recombination , Humans , Male , X Chromosome , X Chromosome Inactivation/genetics
8.
Cell Rep ; 34(13): 108912, 2021 03 30.
Article in English | MEDLINE | ID: mdl-33789104

ABSTRACT

The fine-scale dynamics from euchromatin (EC) to facultative heterochromatin (fHC) has remained largely unclear. Here, we focus on Xist and its silencing initiator Tsix as a paradigm of transcription-mediated conversion from EC to fHC. In mouse epiblast stem cells, induction of Tsix recapitulates the conversion at the Xist promoter. Investigating the dynamics reveals that the conversion proceeds in a stepwise manner. Initially, a transient opened chromatin structure is observed. In the second step, gene silencing is initiated and dependent on Tsix, which is reversible and accompanied by simultaneous changes in multiple histone modifications. At the last step, maintenance of silencing becomes independent of Tsix and irreversible, which correlates with occupation of the -1 position of the transcription start site by a nucleosome and initiation of DNA methylation introduction. This study highlights the hierarchy of multiple chromatin events upon stepwise gene silencing establishment.


Subject(s)
Euchromatin/metabolism , Heterochromatin/metabolism , Promoter Regions, Genetic , RNA, Long Noncoding/genetics , Transcription, Genetic , Animals , CCCTC-Binding Factor/metabolism , DNA Methylation/genetics , Epigenesis, Genetic , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Silencing , Germ Layers/cytology , Histones/metabolism , Mice , Nucleosomes/metabolism , Protein Processing, Post-Translational , RNA, Long Noncoding/metabolism , Stem Cells/metabolism , YY1 Transcription Factor/metabolism
9.
Biology (Basel) ; 9(12)2020 Dec 16.
Article in English | MEDLINE | ID: mdl-33339109

ABSTRACT

The fibroblast growth factor (FGF) and the transforming growth factor-ß (TGF-ß) pathways are both involved in the maintenance of human embryonic stem cells (hESCs) and regulate the onset of their differentiation. Their converging functions have suggested that these pathways might share a wide range of overlapping targets. Published studies have focused on the long-term effects (24-48 h) of FGF and TGF-ß inhibition in hESCs, identifying direct and indirect target genes. In this study, we focused on the earliest transcriptome changes occurring between 3 and 9 h after FGF and TGF-ß inhibition to identify direct target genes only. Our analysis clearly shows that only a handful of target transcripts are common to both pathways. This is surprising in light of the previous literature, and has implications for models of cell signaling in human pluripotent cells. In addition, we identified STOX2 as a novel primary target of the TGF-ß signaling pathway. We show that STOX2 might act as a novel SMAD2/4 cofactor. Taken together, our results provide insights into the effect of cell signaling on the transcription profile of human pluripotent cells.

10.
J Vis Exp ; (165)2020 11 19.
Article in English | MEDLINE | ID: mdl-33283788

ABSTRACT

In organisms with sexual reproduction, germ cells are the source of totipotent cells that develop into new individuals. In mice, fertilization of an oocyte by a spermatozoon creates a totipotent zygote. Recently, several publications have reported that haploid embryonic stem cells (haESCs) can be a substitute for gametic genomes and contribute to embryos, which develop into mice. Here, we present a protocol to apply parthenogenetic haESCs as a substitute of sperm to construct embryos by intracytoplasmic injection into oocytes. This protocol consists of steps for preparing haESCs as sperm replacement, for injection of haESC chromosomes into oocytes, and for culture of semi-cloned embryos. The embryos can yield fertile semi-cloned mice after embryo transfer. Using haESCs as sperm replacement facilitates genome editing in the germline, studies of embryonic development, and investigation of genomic imprinting.


Subject(s)
Haploidy , Mouse Embryonic Stem Cells/cytology , Parthenogenesis , Spermatozoa/cytology , Animals , Cell Line , Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Female , Fibroblasts/cytology , Gene Editing/methods , Genome , Male , Mice , Microinjections , Mitosis , Oocytes/cytology , Plasmids/genetics , Pregnancy , Superovulation
11.
PLoS One ; 15(9): e0233072, 2020.
Article in English | MEDLINE | ID: mdl-32911495

ABSTRACT

In mammals, the fusion of two gametes, an oocyte and a spermatozoon, during fertilization forms a totipotent zygote. There has been no reported case of adult mammal development by natural parthenogenesis, in which embryos develop from unfertilized oocytes. The genome and epigenetic information of haploid gametes are crucial for mammalian development. Haploid embryonic stem cells (haESCs) can be established from uniparental blastocysts and possess only one set of chromosomes. Previous studies have shown that sperm or oocyte genome can be replaced by haESCs with or without manipulation of genomic imprinting for generation of mice. Recently, these remarkable semi-cloning methods have been applied for screening of key factors of mouse embryonic development. While haESCs have been applied as substitutes of gametic genomes, the fundamental mechanism how haESCs contribute to the genome of totipotent embryos is unclear. Here, we show the generation of fertile semi-cloned mice by injection of parthenogenetic haESCs (phaESCs) into oocytes after deletion of two differentially methylated regions (DMRs), the IG-DMR and H19-DMR. For characterizing the genome of semi-cloned embryos further, we establish ESC lines from semi-cloned blastocysts. We report that polyploid karyotypes are observed in semi-cloned ESCs (scESCs). Our results confirm that mitotically arrested phaESCs yield semi-cloned embryos and mice when the IG-DMR and H19-DMR are deleted. In addition, we highlight the occurrence of polyploidy that needs to be considered for further improving the development of semi-cloned embryos derived by haESC injection.


Subject(s)
Cloning, Organism/methods , Embryonic Development , Haploidy , Parthenogenesis , Polyploidy , Animals , Cell Line , Embryonic Stem Cells/cytology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA
13.
Development ; 147(11)2020 06 11.
Article in English | MEDLINE | ID: mdl-32439758

ABSTRACT

We previously identified the cyclin dependent kinase Cdk8 as a putative silencing factor for Xist To investigate its role in X inactivation, we engineered a Cdk8 mutation in mouse embryonic stem cells (ESCs) carrying an inducible system for studying Xist function. We found that Xist repressed X-linked genes at half of the expression level in Cdk8 mutant cells, whereas they were almost completely silenced in the controls. Lack of Cdk8 impaired Ezh2 recruitment and the establishment of histone H3 lysine 27 tri-methylation but not PRC1 recruitment by Xist Transgenic expression of wild-type but not catalytically inactive Cdk8 restored efficient gene repression and PRC2 recruitment. Mutation of the paralogous kinase Cdk19 did not affect Xist function, and combined mutations of Cdk8 and Cdk19 resembled the Cdk8 mutation. In mice, a Cdk8 mutation caused post-implantation lethality. We observed that homozygous Cdk8 mutant female embryos showed a greater developmental delay than males on day 10.5. Together with the inefficient repression of X-linked genes in differentiating Cdk8 mutant female ESCs, these data show a requirement for Cdk8 in the initiation of X inactivation.


Subject(s)
Cyclin-Dependent Kinase 8/metabolism , Histones/metabolism , RNA, Long Noncoding/metabolism , Animals , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Embryo, Mammalian , Embryonic Development , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Gene Editing , Gene Expression Regulation, Developmental , Male , Methylation , Mice , Mice, Transgenic , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Mutagenesis , Polycomb Repressive Complex 2/metabolism , RNA, Guide, Kinetoplastida/metabolism , RNA, Long Noncoding/genetics , SOXB1 Transcription Factors/deficiency , SOXB1 Transcription Factors/genetics
14.
F1000Res ; 92020.
Article in English | MEDLINE | ID: mdl-32047616

ABSTRACT

Female mammals express the long noncoding X inactivation-specific transcript ( Xist) RNA to initiate X chromosome inactivation (XCI) that eventually results in the formation of the Barr body. Xist encompasses half a dozen repeated sequence stretches containing motifs for RNA-binding proteins that recruit effector complexes with functions for silencing genes and establishing a repressive chromatin configuration. Functional characterization of these effector proteins unveils the cooperation of a number of pathways to repress genes on the inactive X chromosome. Mechanistic insights can be extended to other noncoding RNAs with similar structure and open avenues for the design of new therapies to switch off gene expression. Here we review recent advances in the understanding of Xist and on this basis try to synthesize a model for the initiation of XCI.


Subject(s)
X Chromosome Inactivation , X Chromosome , Animals , Chromatin , Female , Gene Silencing , RNA, Long Noncoding/genetics
15.
Cell Stem Cell ; 25(5): 587-589, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31703767

ABSTRACT

In mammals, both parents make unique contributions to the offspring and maternal and paternal genomes are required for development. Two recent papers in Cell Stem Cell (Leng et al., 2019; Sagi et al., 2019) study uniparental embryos and uniparental embryonic stem cells to interrogate parent-of-origin effects in human embryogenesis.


Subject(s)
Embryonic Stem Cells , Fathers , Animals , Cell Differentiation , Embryo, Mammalian , Female , Humans , Male , Mothers
16.
Sci Adv ; 5(9): eaaw6490, 2019 09.
Article in English | MEDLINE | ID: mdl-31555730

ABSTRACT

Hedgehog signaling is central in embryonic development and tissue regeneration. Disruption of the pathway is linked to genetic diseases and cancer. Binding of the secreted ligand, Sonic hedgehog (ShhN) to its receptor Patched (PTCH1) activates the signaling pathway. Here, we describe a 3.4-Å cryo-EM structure of the human PTCH1 bound to ShhNC24II, a modified hedgehog ligand mimicking its palmitoylated form. The membrane-embedded part of PTCH1 is surrounded by 10 sterol molecules at the inner and outer lipid bilayer portion of the protein. The annular sterols interact at multiple sites with both the sterol-sensing domain (SSD) and the SSD-like domain (SSDL), which are located on opposite sides of PTCH1. The structure reveals a possible route for sterol translocation across the lipid bilayer by PTCH1 and homologous transporters.


Subject(s)
Hedgehog Proteins/chemistry , Lipid Bilayers/chemistry , Patched-1 Receptor/chemistry , Sterols/chemistry , Biological Transport , Cryoelectron Microscopy , Hedgehog Proteins/metabolism , Hedgehog Proteins/ultrastructure , Humans , Lipid Bilayers/metabolism , Patched-1 Receptor/metabolism , Patched-1 Receptor/ultrastructure , Protein Domains , Sterols/metabolism
17.
Transgenic Res ; 28(5-6): 525-535, 2019 12.
Article in English | MEDLINE | ID: mdl-31482512

ABSTRACT

CRISPR-associated (Cas) nucleases are established tools for engineering of animal genomes. These programmable RNA-guided nucleases have been introduced into zygotes using expression vectors, mRNA, or directly as ribonucleoprotein (RNP) complexes by different delivery methods. Whereas microinjection techniques are well established, more recently developed electroporation methods simplify RNP delivery but can provide less consistent efficiency. Previously, we have designed Cas12a-crRNA pairs to introduce large genomic deletions in the Ubn1, Ubn2, and Rbm12 genes in mouse embryonic stem cells (ESC). Here, we have optimized the conditions for electroporation of the same Cas12a RNP pairs into mouse zygotes. Using our protocol, large genomic deletions can be generated efficiently by electroporation of zygotes with or without an intact zona pellucida. Electroporation of as few as ten zygotes is sufficient to obtain a gene deletion in mice suggesting potential applicability of this method for species with limited availability of zygotes.


Subject(s)
Bacterial Proteins/genetics , CRISPR-Associated Proteins/genetics , CRISPR-Cas Systems/genetics , Endodeoxyribonucleases/genetics , Gene Deletion , Gene Transfer Techniques , Animals , Electroporation , Genome/genetics , Mice , Mouse Embryonic Stem Cells/metabolism , Mutation/genetics , RNA, Guide, Kinetoplastida/genetics , Zona Pellucida/metabolism , Zygote/growth & development
18.
J Mol Biol ; 431(19): 3920-3932, 2019 09 06.
Article in English | MEDLINE | ID: mdl-31306665

ABSTRACT

Modifications by kinases are a fast and reversible mechanism to diversify the function of the targeted proteins. The OCT4 transcription factor is essential for preimplantation development and pluripotency of embryonic stem cells (ESC), and its activity is tightly regulated by post-transcriptional modifications. Several phosphorylation sites have been identified by systemic approaches and their functions proposed. Here, we combined molecular and cellular biology with CRISPR/Cas9-mediated genome engineering to pinpoint the function of serine 12 of OCT4 in ESCs. Using chemical inhibitors and an antibody specific to OCT4 phosphorylated on S12, we identified cyclin-dependent kinase (CDK) 7 as upstream kinase. Surprisingly, generation of isogenic mESCs that endogenously ablate S12 revealed no effects on pluripotency and self-renewal, potentially due to compensation by other phosphorylation events. Our approach reveals that modification of distinct amino acids by precise genome engineering can help to clarify the functions of post-translational modifications on proteins encoded by essential gene in an endogenous context.


Subject(s)
Genetic Engineering , Genome , Molecular Biology , Protein Processing, Post-Translational , Amino Acid Sequence , Animals , Cell Self Renewal , Humans , Mice , Mouse Embryonic Stem Cells/metabolism , Mutation/genetics , Octamer Transcription Factor-3/chemistry , Octamer Transcription Factor-3/metabolism , Phosphorylation , Phosphoserine/metabolism , Pluripotent Stem Cells/metabolism
19.
Methods Enzymol ; 616: 241-263, 2019.
Article in English | MEDLINE | ID: mdl-30691645

ABSTRACT

CRISPR-Cas12a is a bacterial RNA-guided deoxyribonuclease that has been adopted for genetic engineering in a broad variety of organisms. Here, we describe protocols for the preparation and application of AsCas12a-guide RNA ribonucleoprotein (RNP) complexes for engineering gene deletions in mouse embryonic stem (ES) cells. We provide detailed protocols for purification of an NLS-containing AsCas12a-eGFP fusion protein, design of guide RNAs, assembly of RNP complexes, and transfection of mouse ES cells by electroporation. In addition, we present data illustrating the use of pairs of Cas12a nucleases for engineering large genetic deletions and outline experimental considerations for applications of Cas12a nucleases in ES cells.


Subject(s)
CRISPR-Cas Systems , Gene Deletion , Gene Editing/methods , Mouse Embryonic Stem Cells/metabolism , Animals , CRISPR-Associated Proteins/genetics , Electroporation/methods , Genetic Engineering/methods , Mice , RNA, Guide, Kinetoplastida/genetics
20.
Methods Mol Biol ; 1861: 1-18, 2018.
Article in English | MEDLINE | ID: mdl-30218355

ABSTRACT

Forward genetics can provide insight into molecular pathways as has been demonstrated by advances in cell biology from comprehensive genetic studies in simple organisms. Recently, techniques have become available that promise a similar potential for understanding developmental pathways in mammals. Here we describe a genetic forward screening approach for identifying factors involved in X chromosome inactivation that is based on haploid mouse embryonic stem cells. Using a genetically encoded selection system screening of large mutant pools can identify candidate silencing factors with high confidence.


Subject(s)
Genetic Techniques , Mouse Embryonic Stem Cells , X Chromosome Inactivation , Animals , Cell Line , Epigenomics/methods , Female , HEK293 Cells , Haploidy , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...