Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Cell ; 185(20): 3689-3704.e21, 2022 09 29.
Article in English | MEDLINE | ID: mdl-36179666

ABSTRACT

Regulatory landscapes drive complex developmental gene expression, but it remains unclear how their integrity is maintained when incorporating novel genes and functions during evolution. Here, we investigated how a placental mammal-specific gene, Zfp42, emerged in an ancient vertebrate topologically associated domain (TAD) without adopting or disrupting the conserved expression of its gene, Fat1. In ESCs, physical TAD partitioning separates Zfp42 and Fat1 with distinct local enhancers that drive their independent expression. This separation is driven by chromatin activity and not CTCF/cohesin. In contrast, in embryonic limbs, inactive Zfp42 shares Fat1's intact TAD without responding to active Fat1 enhancers. However, neither Fat1 enhancer-incompatibility nor nuclear envelope-attachment account for Zfp42's unresponsiveness. Rather, Zfp42's promoter is rendered inert to enhancers by context-dependent DNA methylation. Thus, diverse mechanisms enabled the integration of independent Zfp42 regulation in the Fat1 locus. Critically, such regulatory complexity appears common in evolution as, genome wide, most TADs contain multiple independently expressed genes.


Subject(s)
Chromatin , Placenta , Animals , CCCTC-Binding Factor/metabolism , Chromatin Assembly and Disassembly , Enhancer Elements, Genetic , Evolution, Molecular , Female , Genome , Mammals/metabolism , Placenta/metabolism , Pregnancy , Promoter Regions, Genetic , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Cell ; 161(5): 1012-1025, 2015 May 21.
Article in English | MEDLINE | ID: mdl-25959774

ABSTRACT

Mammalian genomes are organized into megabase-scale topologically associated domains (TADs). We demonstrate that disruption of TADs can rewire long-range regulatory architecture and result in pathogenic phenotypes. We show that distinct human limb malformations are caused by deletions, inversions, or duplications altering the structure of the TAD-spanning WNT6/IHH/EPHA4/PAX3 locus. Using CRISPR/Cas genome editing, we generated mice with corresponding rearrangements. Both in mouse limb tissue and patient-derived fibroblasts, disease-relevant structural changes cause ectopic interactions between promoters and non-coding DNA, and a cluster of limb enhancers normally associated with Epha4 is misplaced relative to TAD boundaries and drives ectopic limb expression of another gene in the locus. This rewiring occurred only if the variant disrupted a CTCF-associated boundary domain. Our results demonstrate the functional importance of TADs for orchestrating gene expression via genome architecture and indicate criteria for predicting the pathogenicity of human structural variants, particularly in non-coding regions of the human genome.


Subject(s)
Disease Models, Animal , Enhancer Elements, Genetic , Gene Expression Regulation , Animals , Extremities/anatomy & histology , Extremities/growth & development , Humans , Limb Deformities, Congenital/genetics , Mice , Promoter Regions, Genetic , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Receptor, EphA4/genetics
3.
Nature ; 623(7988): 772-781, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37968388

ABSTRACT

Mouse models are a critical tool for studying human diseases, particularly developmental disorders1. However, conventional approaches for phenotyping may fail to detect subtle defects throughout the developing mouse2. Here we set out to establish single-cell RNA sequencing of the whole embryo as a scalable platform for the systematic phenotyping of mouse genetic models. We applied combinatorial indexing-based single-cell RNA sequencing3 to profile 101 embryos of 22 mutant and 4 wild-type genotypes at embryonic day 13.5, altogether profiling more than 1.6 million nuclei. The 22 mutants represent a range of anticipated phenotypic severities, from established multisystem disorders to deletions of individual regulatory regions4,5. We developed and applied several analytical frameworks for detecting differences in composition and/or gene expression across 52 cell types or trajectories. Some mutants exhibit changes in dozens of trajectories whereas others exhibit changes in only a few cell types. We also identify differences between widely used wild-type strains, compare phenotyping of gain- versus loss-of-function mutants and characterize deletions of topological associating domain boundaries. Notably, some changes are shared among mutants, suggesting that developmental pleiotropy might be 'decomposable' through further scaling of this approach. Overall, our findings show how single-cell profiling of whole embryos can enable the systematic molecular and cellular phenotypic characterization of mouse mutants with unprecedented breadth and resolution.


Subject(s)
Developmental Disabilities , Embryo, Mammalian , Mutation , Phenotype , Single-Cell Gene Expression Analysis , Animals , Mice , Cell Nucleus/genetics , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Gain of Function Mutation , Genotype , Loss of Function Mutation , Models, Genetic , Disease Models, Animal
4.
Genome Res ; 34(4): 572-589, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38719471

ABSTRACT

Dormancy is a key feature of stem cell function in adult tissues as well as in embryonic cells in the context of diapause. The establishment of dormancy is an active process that involves extensive transcriptional, epigenetic, and metabolic rewiring. How these processes are coordinated to successfully transition cells to the resting dormant state remains unclear. Here we show that microRNA activity, which is otherwise dispensable for preimplantation development, is essential for the adaptation of early mouse embryos to the dormant state of diapause. In particular, the pluripotent epiblast depends on miRNA activity, the absence of which results in the loss of pluripotent cells. Through the integration of high-sensitivity small RNA expression profiling of individual embryos and protein expression of miRNA targets with public data of protein-protein interactions, we constructed the miRNA-mediated regulatory network of mouse early embryos specific to diapause. We find that individual miRNAs contribute to the combinatorial regulation by the network, and the perturbation of the network compromises embryo survival in diapause. We further identified the nutrient-sensitive transcription factor TFE3 as an upstream regulator of diapause-specific miRNAs, linking cytoplasmic MTOR activity to nuclear miRNA biogenesis. Our results place miRNAs as a critical regulatory layer for the molecular rewiring of early embryos to establish dormancy.


Subject(s)
Cell Proliferation , MicroRNAs , Pluripotent Stem Cells , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Mice , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Embryonic Development/genetics , Germ Layers/metabolism , Germ Layers/cytology , Blastocyst/metabolism , Blastocyst/cytology , Female
5.
Nature ; 592(7852): 93-98, 2021 04.
Article in English | MEDLINE | ID: mdl-33568816

ABSTRACT

Long non-coding RNAs (lncRNAs) can be important components in gene-regulatory networks1, but the exact nature and extent of their involvement in human Mendelian disease is largely unknown. Here we show that genetic ablation of a lncRNA locus on human chromosome 2 causes a severe congenital limb malformation. We identified homozygous 27-63-kilobase deletions located 300 kilobases upstream of the engrailed-1 gene (EN1) in patients with a complex limb malformation featuring mesomelic shortening, syndactyly and ventral nails (dorsal dimelia). Re-engineering of the human deletions in mice resulted in a complete loss of En1 expression in the limb and a double dorsal-limb phenotype that recapitulates the human disease phenotype. Genome-wide transcriptome analysis in the developing mouse limb revealed a four-exon-long non-coding transcript within the deleted region, which we named Maenli. Functional dissection of the Maenli locus showed that its transcriptional activity is required for limb-specific En1 activation in cis, thereby fine-tuning the gene-regulatory networks controlling dorso-ventral polarity in the developing limb bud. Its loss results in the En1-related dorsal ventral limb phenotype, a subset of the full En1-associated phenotype. Our findings demonstrate that mutations involving lncRNA loci can result in human Mendelian disease.


Subject(s)
Extremities , Homeodomain Proteins/genetics , Limb Deformities, Congenital/genetics , RNA, Long Noncoding/genetics , Sequence Deletion/genetics , Transcription, Genetic , Transcriptional Activation/genetics , Animals , Cell Line , Chromatin/genetics , Disease Models, Animal , Female , Humans , Mice , Mice, Transgenic
6.
Development ; 150(10)2023 05 15.
Article in English | MEDLINE | ID: mdl-37082965

ABSTRACT

Cardiac lineage specification in the mouse is controlled by TGFß and WNT signaling. From fly to fish, BMP has been identified as an indispensable heart inducer. A detailed analysis of the role of Bmp4 and its effectors Smad1/5, however, was still missing. We show that Bmp4 induces cardiac mesoderm formation in murine embryonic stem cells in vitro. Bmp4 first activates Wnt3 and upregulates Nodal. pSmad1/5 and the WNT effector Tcf3 form a complex, and together with pSmad2/3 activate mesoderm enhancers and Eomes. They then cooperate with Eomes to consolidate the expression of many mesoderm factors, including T. Eomes and T form a positive- feedback loop and open additional enhancers regulating early mesoderm genes, including the transcription factor Mesp1, establishing the cardiac mesoderm lineage. In parallel, the neural fate is suppressed. Our data confirm the pivotal role of Bmp4 in cardiac mesoderm formation in the mouse. We describe in detail the consecutive and cooperative actions of three signaling pathways, BMP, WNT and Nodal, and their effector transcription factors, during cardiac mesoderm specification.


Subject(s)
Heart , Transcription Factors , Mice , Animals , Cell Differentiation/genetics , Transcription Factors/metabolism , Mesoderm/metabolism , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway/genetics , Bone Morphogenetic Protein 4/metabolism
7.
Development ; 150(22)2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37882764

ABSTRACT

The node and notochord are important signaling centers organizing the dorso-ventral patterning of cells arising from neuro-mesodermal progenitors forming the embryonic body anlage. Owing to the scarcity of notochord progenitors and notochord cells, a comprehensive identification of regulatory elements driving notochord-specific gene expression has been lacking. Here, we have used ATAC-seq analysis of FACS-purified notochord cells from Theiler stage 12-13 mouse embryos to identify 8921 putative notochord enhancers. In addition, we established a new model for generating notochord-like cells in culture, and found 3728 of these enhancers occupied by the essential notochord control factors brachyury (T) and/or Foxa2. We describe the regulatory landscape of the T locus, comprising ten putative enhancers occupied by these factors, and confirmed the regulatory activity of three of these elements. Moreover, we characterized seven new elements by knockout analysis in embryos and identified one new notochord enhancer, termed TNE2. TNE2 cooperates with TNE in the trunk notochord, and is essential for notochord differentiation in the tail. Our data reveal an essential role of Foxa2 in directing T-expressing cells towards the notochord lineage.


Subject(s)
Enhancer Elements, Genetic , Notochord , Mice , Animals , Enhancer Elements, Genetic/genetics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Fetal Proteins/genetics , Fetal Proteins/metabolism , Gene Expression Regulation, Developmental/genetics
8.
Development ; 150(17)2023 09 01.
Article in English | MEDLINE | ID: mdl-37519269

ABSTRACT

Changes in gene expression represent an important source of phenotypic innovation. Yet how such changes emerge and impact the evolution of traits remains elusive. Here, we explore the molecular mechanisms associated with the development of masculinizing ovotestes in female moles. By performing integrative analyses of epigenetic and transcriptional data in mole and mouse, we identified the co-option of SALL1 expression in mole ovotestes formation. Chromosome conformation capture analyses highlight a striking conservation of the 3D organization at the SALL1 locus, but an evolutionary divergence of enhancer activity. Interspecies reporter assays support the capability of mole-specific enhancers to activate transcription in urogenital tissues. Through overexpression experiments in transgenic mice, we further demonstrate the capability of SALL1 to induce kidney-related gene programs, which are a signature of mole ovotestes. Our results highlight the co-option of gene expression, through changes in enhancer activity, as a plausible mechanism for the evolution of traits.


Subject(s)
Kidney , Moles , Animals , Female , Mice , Kidney/metabolism , Mice, Transgenic , Moles/genetics
9.
Nature ; 584(7819): 102-108, 2020 08.
Article in English | MEDLINE | ID: mdl-32728215

ABSTRACT

During ontogeny, proliferating cells become restricted in their fate through the combined action of cell-type-specific transcription factors and ubiquitous epigenetic machinery, which recognizes universally available histone residues or nucleotides in a context-dependent manner1,2. The molecular functions of these regulators are generally well understood, but assigning direct developmental roles to them is hampered by complex mutant phenotypes that often emerge after gastrulation3,4. Single-cell RNA sequencing and analytical approaches have explored this highly conserved, dynamic period across numerous model organisms5-8, including mouse9-18. Here we advance these strategies using a combined zygotic perturbation and single-cell RNA-sequencing platform in which many mutant mouse embryos can be assayed simultaneously, recovering robust  morphological and transcriptional information across a panel of ten essential regulators. Deeper analysis of central Polycomb repressive complex (PRC) 1 and 2 components indicates substantial cooperativity, but distinguishes a dominant role for PRC2 in restricting the germline. Moreover, PRC mutant phenotypes emerge after gross epigenetic and transcriptional changes within the initial conceptus prior to gastrulation. Our experimental framework may eventually lead to a fully quantitative view of how cellular diversity emerges using an identical genetic template and from a single totipotent cell.


Subject(s)
Epigenesis, Genetic , Gastrula/embryology , Gastrula/metabolism , Gastrulation/genetics , Animals , Cell Lineage , Female , Gastrula/cytology , Gene Expression Regulation, Developmental , Male , Mice , Mutation , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 2/metabolism , Single-Cell Analysis , Transcription, Genetic
10.
Proc Natl Acad Sci U S A ; 119(22): e2201883119, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35617427

ABSTRACT

Polycomb-group proteins play critical roles in gene silencing through the deposition of histone H3 lysine 27 trimethylation (H3K27me3) and chromatin compaction. This process is essential for embryonic stem cell (ESC) pluripotency, differentiation, and development. Polycomb repressive complex 2 (PRC2) can both read and write H3K27me3, enabling progressive spreading of H3K27me3 on the linear genome. Long-range Polycomb-associated DNA contacts have also been described, but their regulation and role in gene silencing remain unclear. Here, we apply H3K27me3 HiChIP, a protein-directed chromosome conformation method, and optical reconstruction of chromatin architecture to profile long-range Polycomb-associated DNA loops that span tens to hundreds of megabases across multiple topological associated domains in mouse ESCs and human induced pluripotent stem cells. We find that H3K27me3 loop anchors are enriched for Polycomb nucleation points and coincide with key developmental genes. Genetic deletion of H3K27me3 loop anchors results in disruption of spatial contact between distant loci and altered H3K27me3 in cis, both locally and megabases away on the same chromosome. In mouse embryos, loop anchor deletion leads to ectopic activation of the partner gene, suggesting that Polycomb-associated loops control gene silencing during development. Further, we find that alterations in PRC2 occupancy resulting from an RNA binding­deficient EZH2 mutant are accompanied by loss of Polycomb-associated DNA looping. Together, these results suggest PRC2 uses RNA binding to enhance long-range chromosome folding and H3K27me3 spreading. Developmental gene loci have unique roles in Polycomb spreading, emerging as important architectural elements of the epigenome.


Subject(s)
Chromosomes , Gene Expression Regulation, Developmental , Gene Silencing , Histones , Polycomb Repressive Complex 2 , Animals , Chromatin Immunoprecipitation/methods , Chromosomes/chemistry , Chromosomes/metabolism , Embryo, Mammalian , Enhancer of Zeste Homolog 2 Protein/genetics , Histones/genetics , Histones/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Lysine/metabolism , Methylation , Mice , Nucleic Acid Conformation , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism
11.
Development ; 148(23)2021 12 01.
Article in English | MEDLINE | ID: mdl-34822716

ABSTRACT

The node-streak border region comprising notochord progenitor cells (NPCs) at the posterior node and neuro-mesodermal progenitor cells (NMPs) in the adjacent epiblast is the prime organizing center for axial elongation in mouse embryos. The T-box transcription factor brachyury (T) is essential for both formation of the notochord and maintenance of NMPs, and thus is a key regulator of trunk and tail development. The T promoter controlling T expression in NMPs and nascent mesoderm has been characterized in detail; however, control elements for T expression in the notochord have not been identified yet. We have generated a series of deletion alleles by CRISPR/Cas9 genome editing in mESCs, and analyzed their effects in mutant mouse embryos. We identified a 37 kb region upstream of T that is essential for notochord function and tailbud outgrowth. Within that region, we discovered a T-binding enhancer required for notochord cell specification and differentiation. Our data reveal a complex regulatory landscape controlling cell type-specific expression and function of T in NMP/nascent mesoderm and node/notochord, allowing proper trunk and tail development.


Subject(s)
Embryonic Development/genetics , Enhancer Elements, Genetic/genetics , Fetal Proteins/genetics , T-Box Domain Proteins/genetics , Tail/growth & development , Amino Acid Sequence/genetics , Animals , CRISPR-Cas Systems/genetics , Gene Editing/methods , Gene Expression Regulation, Developmental/genetics , Mesoderm/growth & development , Mesoderm/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Notochord/growth & development , Notochord/metabolism , Promoter Regions, Genetic/genetics , Regulatory Sequences, Nucleic Acid/genetics , Tail/metabolism
12.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Article in English | MEDLINE | ID: mdl-33402532

ABSTRACT

Pathogenic germline mutations in PIGV lead to glycosylphosphatidylinositol biosynthesis deficiency (GPIBD). Individuals with pathogenic biallelic mutations in genes of the glycosylphosphatidylinositol (GPI)-anchor pathway exhibit cognitive impairments, motor delay, and often epilepsy. Thus far, the pathophysiology underlying the disease remains unclear, and suitable rodent models that mirror all symptoms observed in human patients have not been available. Therefore, we used CRISPR-Cas9 to introduce the most prevalent hypomorphic missense mutation in European patients, Pigv:c.1022C > A (p.A341E), at a site that is conserved in mice. Mirroring the human pathology, mutant Pigv341E mice exhibited deficits in motor coordination, cognitive impairments, and alterations in sociability and sleep patterns, as well as increased seizure susceptibility. Furthermore, immunohistochemistry revealed reduced synaptophysin immunoreactivity in Pigv341E mice, and electrophysiology recordings showed decreased hippocampal synaptic transmission that could underlie impaired memory formation. In single-cell RNA sequencing, Pigv341E-hippocampal cells exhibited changes in gene expression, most prominently in a subtype of microglia and subicular neurons. A significant reduction in Abl1 transcript levels in several cell clusters suggested a link to the signaling pathway of GPI-anchored ephrins. We also observed elevated levels of Hdc transcripts, which might affect histamine metabolism with consequences for circadian rhythm. This mouse model will not only open the doors to further investigation into the pathophysiology of GPIBD, but will also deepen our understanding of the role of GPI-anchor-related pathways in brain development.


Subject(s)
Glycosylphosphatidylinositols/genetics , Glycosylphosphatidylinositols/metabolism , Mannosyltransferases/metabolism , Abnormalities, Multiple/genetics , Amino Acid Sequence , Amino Acids/genetics , Animals , CRISPR-Cas Systems , Disease Models, Animal , Epilepsy/genetics , Glycosylphosphatidylinositols/deficiency , Hippocampus/metabolism , Intellectual Disability/genetics , Mannosyltransferases/physiology , Mice , Mice, Inbred C57BL , Mutation , Mutation, Missense , Phenotype , Protein Engineering/methods , Seizures/genetics , Seizures/physiopathology
13.
Nature ; 538(7624): 265-269, 2016 Oct 13.
Article in English | MEDLINE | ID: mdl-27706140

ABSTRACT

Chromosome conformation capture methods have identified subchromosomal structures of higher-order chromatin interactions called topologically associated domains (TADs) that are separated from each other by boundary regions. By subdividing the genome into discrete regulatory units, TADs restrict the contacts that enhancers establish with their target genes. However, the mechanisms that underlie partitioning of the genome into TADs remain poorly understood. Here we show by chromosome conformation capture (capture Hi-C and 4C-seq methods) that genomic duplications in patient cells and genetically modified mice can result in the formation of new chromatin domains (neo-TADs) and that this process determines their molecular pathology. Duplications of non-coding DNA within the mouse Sox9 TAD (intra-TAD) that cause female to male sex reversal in humans, showed increased contact of the duplicated regions within the TAD, but no change in the overall TAD structure. In contrast, overlapping duplications that extended over the next boundary into the neighbouring TAD (inter-TAD), resulted in the formation of a new chromatin domain (neo-TAD) that was isolated from the rest of the genome. As a consequence of this insulation, inter-TAD duplications had no phenotypic effect. However, incorporation of the next flanking gene, Kcnj2, in the neo-TAD resulted in ectopic contacts of Kcnj2 with the duplicated part of the Sox9 regulatory region, consecutive misexpression of Kcnj2, and a limb malformation phenotype. Our findings provide evidence that TADs are genomic regulatory units with a high degree of internal stability that can be sculptured by structural genomic variations. This process is important for the interpretation of copy number variations, as these variations are routinely detected in diagnostic tests for genetic disease and cancer. This finding also has relevance in an evolutionary setting because copy-number differences are thought to have a crucial role in the evolution of genome complexity.


Subject(s)
Chromatin Assembly and Disassembly/genetics , DNA Copy Number Variations/genetics , Disease/genetics , Gene Duplication/genetics , Animals , DNA/genetics , Facies , Female , Fibroblasts , Fingers/abnormalities , Foot Deformities, Congenital/genetics , Gene Expression , Genomics , Hand Deformities, Congenital/genetics , Male , Mice , Phenotype , SOX9 Transcription Factor/genetics
14.
Proc Natl Acad Sci U S A ; 116(25): 12390-12399, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31147463

ABSTRACT

Long-range gene regulation involves physical proximity between enhancers and promoters to generate precise patterns of gene expression in space and time. However, in some cases, proximity coincides with gene activation, whereas, in others, preformed topologies already exist before activation. In this study, we investigate the preformed configuration underlying the regulation of the Shh gene by its unique limb enhancer, the ZRS, in vivo during mouse development. Abrogating the constitutive transcription covering the ZRS region led to a shift within the Shh-ZRS contacts and a moderate reduction in Shh transcription. Deletion of the CTCF binding sites around the ZRS resulted in the loss of the Shh-ZRS preformed interaction and a 50% decrease in Shh expression but no phenotype, suggesting an additional, CTCF-independent mechanism of promoter-enhancer communication. This residual activity, however, was diminished by combining the loss of CTCF binding with a hypomorphic ZRS allele, resulting in severe Shh loss of function and digit agenesis. Our results indicate that the preformed chromatin structure of the Shh locus is sustained by multiple components and acts to reinforce enhancer-promoter communication for robust transcription.


Subject(s)
Chromatin/metabolism , Extremities/embryology , Hedgehog Proteins/genetics , Transcription, Genetic , Animals , Binding Sites , CCCTC-Binding Factor/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Down-Regulation , Enhancer Elements, Genetic , Membrane Proteins/genetics , Mice , Promoter Regions, Genetic , Cohesins
15.
EMBO Rep ; 19(1): 118-134, 2018 01.
Article in English | MEDLINE | ID: mdl-29141987

ABSTRACT

T-box transcription factors play essential roles in multiple aspects of vertebrate development. Here, we show that cooperative function of BRACHYURY (T) with histone-modifying enzymes is essential for mouse embryogenesis. A single point mutation (TY88A) results in decreased histone 3 lysine 27 acetylation (H3K27ac) at T target sites, including the T locus, suggesting that T autoregulates the maintenance of its expression and functions by recruiting permissive chromatin modifications to putative enhancers during mesoderm specification. Our data indicate that T mediates H3K27ac recruitment through a physical interaction with p300. In addition, we determine that T plays a prominent role in the specification of hematopoietic and endothelial cell types. Hematopoietic and endothelial gene expression programs are disrupted in TY88A mutant embryos, leading to a defect in the differentiation of hematopoietic progenitors. We show that this role of T is mediated, at least in part, through activation of a distal Lmo2 enhancer.


Subject(s)
Embryonic Development/genetics , Fetal Proteins/genetics , Histones/metabolism , Mesoderm/metabolism , Mouse Embryonic Stem Cells/metabolism , T-Box Domain Proteins/genetics , p300-CBP Transcription Factors/genetics , Acetylation , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Base Sequence , Cell Differentiation , Cell Lineage/genetics , Chromatin/chemistry , Chromatin/metabolism , Embryo, Mammalian , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fetal Proteins/metabolism , Gene Expression Regulation, Developmental , Genetic Loci , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Histones/genetics , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Mesoderm/cytology , Mesoderm/growth & development , Mice , Mouse Embryonic Stem Cells/cytology , Point Mutation , Protein Binding , Signal Transduction , T-Box Domain Proteins/metabolism , p300-CBP Transcription Factors/metabolism
16.
Genome Res ; 26(2): 183-91, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26755636

ABSTRACT

The CRISPR/Cas technology enables targeted genome editing and the rapid generation of transgenic animal models for the study of human genetic disorders. Here we describe an autosomal recessive human disease in two unrelated families characterized by a split-foot defect, nail abnormalities of the hands, and hearing loss, due to mutations disrupting the SAM domain of the protein kinase ZAK. ZAK is a member of the MAPKKK family with no known role in limb development. We show that Zak is expressed in the developing limbs and that a CRISPR/Cas-mediated knockout of the two Zak isoforms is embryonically lethal in mice. In contrast, a deletion of the SAM domain induces a complex hindlimb defect associated with down-regulation of Trp63, a known split-hand/split-foot malformation disease gene. Our results identify ZAK as a key player in mammalian limb patterning and demonstrate the rapid utility of CRISPR/Cas genome editing to assign causality to human mutations in the mouse in <10 wk.


Subject(s)
Limb Deformities, Congenital/genetics , MAP Kinase Kinase Kinases/genetics , Protein Kinases/genetics , Amino Acid Sequence , Animals , Bacterial Proteins , CRISPR-Associated Protein 9 , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats , Coculture Techniques , Endonucleases , Exome , Female , Humans , Lod Score , MAP Kinase Kinase Kinases/chemistry , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Site-Directed , Mutation, Missense , Pedigree , Polymorphism, Single Nucleotide , Protein Kinases/chemistry , Sequence Analysis, DNA
17.
Genet Med ; 20(6): 599-607, 2018 06.
Article in English | MEDLINE | ID: mdl-29236091

ABSTRACT

PurposeCopy-number variants (CNVs) are generally interpreted by linking the effects of gene dosage with phenotypes. The clinical interpretation of noncoding CNVs remains challenging. We investigated the percentage of disease-associated CNVs in patients with congenital limb malformations that affect noncoding cis-regulatory sequences versus genes sensitive to gene dosage effects.MethodsWe applied high-resolution copy-number analysis to 340 unrelated individuals with isolated limb malformation. To investigate novel candidate CNVs, we re-engineered human CNVs in mice using clustered regularly interspaced short palindromic repeats (CRISPR)-based genome editing.ResultsOf the individuals studied, 10% harbored CNVs segregating with the phenotype in the affected families. We identified 31 CNVs previously associated with congenital limb malformations and four novel candidate CNVs. Most of the disease-associated CNVs (57%) affected the noncoding cis-regulatory genome, while only 43% included a known disease gene and were likely to result from gene dosage effects. In transgenic mice harboring four novel candidate CNVs, we observed altered gene expression in all cases, indicating that the CNVs had a regulatory effect either by changing the enhancer dosage or altering the topological associating domain architecture of the genome.ConclusionOur findings suggest that CNVs affecting noncoding regulatory elements are a major cause of congenital limb malformations.


Subject(s)
DNA, Intergenic/genetics , Limb Deformities, Congenital/genetics , Animals , DNA Copy Number Variations/genetics , Female , Gene Dosage/genetics , Genome, Human , Genome-Wide Association Study , Humans , Male , Mice , Mice, Transgenic , Pedigree , Phenotype
18.
Development ; 141(11): 2325-30, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24803591

ABSTRACT

Differential gene expression is a prerequisite for the formation of multiple cell types from the fertilized egg during embryogenesis. Understanding the gene regulatory networks controlling cellular differentiation requires the identification of crucial differentially expressed control genes and, ideally, the determination of the complete transcriptomes of each individual cell type. Here, we have analyzed the transcriptomes of six major tissues dissected from mid-gestational (TS12) mouse embryos. Approximately one billion reads derived by RNA-seq analysis provided extended transcript lengths, novel first exons and alternative transcripts of known genes. We have identified 1375 genes showing tissue-specific expression, providing gene signatures for each of the six tissues. In addition, we have identified 1403 novel putative long noncoding RNA gene loci, 439 of which show differential expression. Our analysis provides the first complete transcriptome data for the mouse embryo. It offers a rich data source for the analysis of individual genes and gene regulatory networks controlling mid-gestational development.


Subject(s)
Embryonic Development/genetics , Gene Expression Regulation, Developmental , RNA, Untranslated/genetics , Transcriptome , Alternative Splicing , Animals , Embryo, Mammalian , Exons , Female , Gene Expression Profiling , Gene Regulatory Networks , High-Throughput Nucleotide Sequencing , Mice , Mice, Inbred C57BL , RNA, Untranslated/metabolism , Sequence Analysis, RNA , Tissue Distribution
19.
Stem Cells ; 34(7): 1790-800, 2016 07.
Article in English | MEDLINE | ID: mdl-27038343

ABSTRACT

Presomitic mesoderm (PSM) cells are the precursors of the somites, which flank both sides of the neural tube and give rise to the musculo-skeletal system shaping the vertebrate body. WNT and FGF signaling control the formation of both the PSM and the somites and show a graded distribution with highest levels in the posterior PSM. We have used reporters for the mesoderm/PSM control genes T, Tbx6, and Msgn1 to investigate the differentiation of mouse ESCs from the naïve state via EpiSCs to PSM cells. Here we show that the activation of WNT signaling by CHIR99021 (CH) in combination with FGF ligand induces embryo-like PSM at high efficiency. By varying the FGF ligand concentration, the state of PSM cells formed can be altered. High FGF concentration supports posterior PSM formation, whereas low FGF generates anterior/differentiating PSM, in line with in vivo data. Furthermore, the level of Msgn1 expression depends on the FGF ligand concentration. We also show that Activin/Nodal signaling inhibits CH-mediated PSM induction in EpiSCs, without affecting T-expression. Inversely, Activin/Nodal inhibition enhances PSM induction by WNT/high FGF signaling. The ability to generate PSM cells of either posterior or anterior PSM identity with high efficiency in vitro will promote the investigation of the gene regulatory networks controlling the formation of nascent PSM cells and their switch to differentiating/somitic paraxial mesoderm. Stem Cells 2016;34:1790-1800.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 8/metabolism , Mesoderm/embryology , Somites/embryology , Wnt Proteins/metabolism , Activins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Ligands , Mesoderm/cytology , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Somites/cytology
20.
Hum Mol Genet ; 23(20): 5536-44, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-24852367

ABSTRACT

Bladder exstrophy-epispadias complex (BEEC), the severe end of the urorectal malformation spectrum, has a profound impact on continence as well as sexual and renal functions. It is widely accepted that for the majority of cases the genetic basis appears to be multifactorial. Here, we report the first study which utilizes genome-wide association methods to analyze a cohort comprising patients presenting the most common BEEC form, classic bladder exstrophy (CBE), to identify common variation associated with risk for isolated CBE. We employed discovery and follow-up samples comprising 218 cases/865 controls and 78 trios in total, all of European descent. Our discovery sample identified a marker near SALL1, showing genome-wide significant association with CBE. However, analyses performed on follow-up samples did not add further support to these findings. We were also able to identify an association with CBE across our study samples (discovery: P = 8.88 × 10(-5); follow-up: P = 0.0025; combined: 1.09 × 10(-6)) in a highly conserved 32 kb intergenic region containing regulatory elements between WNT3 and WNT9B. Subsequent analyses in mice revealed expression for both genes in the genital region during stages relevant to the development of CBE in humans. Unfortunately, we were not able to replicate the suggestive signal for WNT3 and WNT9B in a sample that was enriched for non-CBE BEEC cases (P = 0.51). Our suggestive findings support the hypothesis that larger samples are warranted to identify association of common variation with CBE.


Subject(s)
Bladder Exstrophy/genetics , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt3 Protein/genetics , Wnt3 Protein/metabolism , Animals , Base Sequence , Bladder Exstrophy/pathology , Case-Control Studies , Conserved Sequence , Genetic Predisposition to Disease , Genitalia/embryology , Genitalia/metabolism , Genome-Wide Association Study , Humans , Mice , Polymorphism, Single Nucleotide , Transcription Factors/genetics , White People/genetics
SELECTION OF CITATIONS
SEARCH DETAIL