Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Development ; 151(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38828852

ABSTRACT

The cellular and genetic networks that contribute to the development of the zeugopod (radius and ulna of the forearm, tibia and fibula of the leg) are not well understood, although these bones are susceptible to loss in congenital human syndromes and to the action of teratogens such as thalidomide. Using a new fate-mapping approach with the Chameleon transgenic chicken line, we show that there is a small contribution of SHH-expressing cells to the posterior ulna, posterior carpals and digit 3. We establish that although the majority of the ulna develops in response to paracrine SHH signalling in both the chicken and mouse, there are differences in the contribution of SHH-expressing cells between mouse and chicken as well as between the chicken ulna and fibula. This is evidence that, although zeugopod bones are clearly homologous according to the fossil record, the gene regulatory networks that contribute to their development and evolution are not fixed.


Subject(s)
Animals, Genetically Modified , Chickens , Hedgehog Proteins , Animals , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Chickens/genetics , Mice , Biological Evolution , Chick Embryo , Ulna , Gene Expression Regulation, Developmental , Fibula/metabolism , Radius/metabolism , Humans , Extremities/embryology
2.
Cell ; 149(5): 1008-22, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22579044

ABSTRACT

The presence of ribonucleotides in genomic DNA is undesirable given their increased susceptibility to hydrolysis. Ribonuclease (RNase) H enzymes that recognize and process such embedded ribonucleotides are present in all domains of life. However, in unicellular organisms such as budding yeast, they are not required for viability or even efficient cellular proliferation, while in humans, RNase H2 hypomorphic mutations cause the neuroinflammatory disorder Aicardi-Goutières syndrome. Here, we report that RNase H2 is an essential enzyme in mice, required for embryonic growth from gastrulation onward. RNase H2 null embryos accumulate large numbers of single (or di-) ribonucleotides embedded in their genomic DNA (>1,000,000 per cell), resulting in genome instability and a p53-dependent DNA-damage response. Our findings establish RNase H2 as a key mammalian genome surveillance enzyme required for ribonucleotide removal and demonstrate that ribonucleotides are the most commonly occurring endogenous nucleotide base lesion in replicating cells.


Subject(s)
DNA Replication , Embryo, Mammalian/metabolism , Ribonuclease H/genetics , Ribonuclease H/metabolism , Ribonucleotides/metabolism , Animals , Chromosomal Instability , DNA-Directed DNA Polymerase/metabolism , Embryonic Stem Cells/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
3.
Development ; 146(19)2019 09 30.
Article in English | MEDLINE | ID: mdl-31511252

ABSTRACT

Topologically associating domains (TADs) have been proposed to both guide and constrain enhancer activity. Shh is located within a TAD known to contain all its enhancers. To investigate the importance of chromatin conformation and TAD integrity on developmental gene regulation, we have manipulated the Shh TAD - creating internal deletions, deleting CTCF sites, and deleting and inverting sequences at TAD boundaries. Chromosome conformation capture and fluorescence in situ hybridisation assays were used to investigate the changes in chromatin conformation that result from these manipulations. Our data suggest that these substantial alterations in TAD structure have no readily detectable effect on Shh expression patterns or levels of Shh expression during development - except where enhancers are deleted - and result in no detectable phenotypes. Only in the case of a larger deletion at one TAD boundary could ectopic influence of the Shh limb enhancer be detected on a gene (Mnx1) in the neighbouring TAD. Our data suggests that, contrary to expectations, the developmental regulation of Shh expression is remarkably robust to TAD perturbations.


Subject(s)
Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Animals , Base Pairing/genetics , CCCTC-Binding Factor , Chromatin/metabolism , Embryo, Mammalian/metabolism , Extremities/embryology , Genome , Hedgehog Proteins/metabolism , Mice , Organ Specificity/genetics , Phenotype , Sequence Deletion/genetics
4.
EMBO J ; 35(8): 831-44, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26903602

ABSTRACT

Aicardi-Goutières syndrome (AGS) provides a monogenic model of nucleic acid-mediated inflammation relevant to the pathogenesis of systemic autoimmunity. Mutations that impair ribonuclease (RNase) H2 enzyme function are the most frequent cause of this autoinflammatory disorder of childhood and are also associated with systemic lupus erythematosus. Reduced processing of eitherRNA:DNAhybrid or genome-embedded ribonucleotide substrates is thought to lead to activation of a yet undefined nucleic acid-sensing pathway. Here, we establishRnaseh2b(A174T/A174T)knock-in mice as a subclinical model of disease, identifying significant interferon-stimulated gene (ISG) transcript upregulation that recapitulates theISGsignature seen inAGSpatients. The inflammatory response is dependent on the nucleic acid sensor cyclicGMP-AMPsynthase (cGAS) and its adaptorSTINGand is associated with reduced cellular ribonucleotide excision repair activity and increasedDNAdamage. This suggests thatcGAS/STINGis a key nucleic acid-sensing pathway relevant toAGS, providing additional insight into disease pathogenesis relevant to the development of therapeutics for this childhood-onset interferonopathy and adult systemic autoimmune disorders.


Subject(s)
Autoimmune Diseases of the Nervous System/genetics , Immunity, Innate/genetics , Membrane Proteins/immunology , Mutation, Missense , Nervous System Malformations/genetics , Nucleotidyltransferases/immunology , Ribonuclease H/genetics , Ribonucleases/genetics , Animals , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/metabolism , Autoimmunity/genetics , DNA Damage , Gene Expression Regulation , Humans , Interferons/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Mutant Strains , Nervous System Malformations/immunology , Nervous System Malformations/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Ribonuclease H/metabolism
5.
Development ; 143(16): 2994-3001, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27402708

ABSTRACT

Limb-specific Shh expression is regulated by the (∼1 Mb distant) ZRS enhancer. In the mouse, limb bud-restricted spatiotemporal Shh expression occurs from ∼E10 to E11.5 at the distal posterior margin and is essential for correct autopod formation. Here, we have analysed the higher-order chromatin conformation of Shh in expressing and non-expressing tissues, both by fluorescence in situ hybridisation (FISH) and by chromosome conformation capture (5C). Conventional and super-resolution light microscopy identified significantly elevated frequencies of Shh/ZRS colocalisation only in the Shh-expressing regions of the limb bud, in a conformation consistent with enhancer-promoter loop formation. However, in all tissues and at all developmental stages analysed, Shh-ZRS spatial distances were still consistently shorter than those to a neural enhancer located between Shh and ZRS in the genome. 5C identified a topologically associating domain (TAD) over the Shh/ZRS genomic region and enriched interactions between Shh and ZRS throughout E11.5 embryos. Shh/ZRS colocalisation, therefore, correlates with the spatiotemporal domain of limb bud-specific Shh expression, but close Shh and ZRS proximity in the nucleus occurs regardless of whether the gene or enhancer is active. We suggest that this constrained chromatin configuration optimises the opportunity for the active enhancer to locate and instigate the expression of Shh.


Subject(s)
Enhancer Elements, Genetic/genetics , Hedgehog Proteins/metabolism , Animals , Chromosomes/genetics , Chromosomes/metabolism , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , In Situ Hybridization, Fluorescence , Limb Buds/metabolism , Mice
7.
PLoS Genet ; 11(12): e1005673, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26646717

ABSTRACT

Human ß-defensin 3 (hBD3) is a cationic host defence peptide and is part of the innate immune response. HBD3 is present on a highly copy number variable block of six ß-defensin genes, and increased copy number is associated with the autoimmune disease psoriasis. It is not known how this increase influences disease development, but psoriasis is a T cell-mediated disease and activation of the innate immune system is required for the initial trigger that leads to the amplification stage. We investigated the effect of hBD3 on the response of primary macrophages to various TLR agonists. HBD3 exacerbated the production of type I Interferon-ß in response to the viral ligand mimic polyinosinic:polycytidylic acid (polyI:C) in both human and mouse primary cells, although production of the chemokine CXCL10 was suppressed. Compared to polyI:C alone, mice injected with both hBD3 peptide and polyI:C also showed an enhanced increase in Interferon-ß. Mice expressing a transgene encoding hBD3 had elevated basal levels of Interferon-ß, and challenge with polyI:C further increased this response. HBD3 peptide increased uptake of polyI:C by macrophages, however the cellular response and localisation of polyI:C in cells treated contemporaneously with hBD3 or cationic liposome differed. Immunohistochemistry showed that hBD3 and polyI:C do not co-localise, but in the presence of hBD3 less polyI:C localises to the early endosome. Using bone marrow derived macrophages from knockout mice we demonstrate that hBD3 suppresses the polyI:C-induced TLR3 response mediated by TICAM1 (TRIF), while exacerbating the cytoplasmic response through MDA5 (IFIH1) and MAVS (IPS1/CARDIF). Thus, hBD3, a highly copy number variable gene in human, influences cellular responses to the viral mimic polyI:C implying that copy number may have a significant phenotypic effect on the response to viral infection and development of autoimmunity in humans.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Vesicular Transport/genetics , DEAD-box RNA Helicases/genetics , Psoriasis/genetics , Toll-Like Receptor 3/genetics , beta-Defensins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Bone Marrow , Chemokine CXCL10/genetics , DEAD-box RNA Helicases/metabolism , Humans , Immunity, Innate/genetics , Interferon-Induced Helicase, IFIH1 , Liposomes/metabolism , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Poly I-C/administration & dosage , Psoriasis/pathology , Toll-Like Receptor 3/antagonists & inhibitors , beta-Defensins/metabolism
8.
Development ; 141(20): 3934-43, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25252942

ABSTRACT

Coordinated gene expression controlled by long-distance enhancers is orchestrated by DNA regulatory sequences involving transcription factors and layers of control mechanisms. The Shh gene and well-established regulators are an example of genomic composition in which enhancers reside in a large desert extending into neighbouring genes to control the spatiotemporal pattern of expression. Exploiting the local hopping activity of the Sleeping Beauty transposon, the lacZ reporter gene was dispersed throughout the Shh region to systematically map the genomic features responsible for expression activity. We found that enhancer activities are retained inside a genomic region that corresponds to the topological associated domain (TAD) defined by Hi-C. This domain of approximately 900 kb is in an open conformation over its length and is generally susceptible to all Shh enhancers. Similar to the distal enhancers, an enhancer residing within the Shh second intron activates the reporter gene located at distances of hundreds of kilobases away, suggesting that both proximal and distal enhancers have the capacity to survey the Shh topological domain to recognise potential promoters. The widely expressed Rnf32 gene lying within the Shh domain evades enhancer activities by a process that may be common among other housekeeping genes that reside in large regulatory domains. Finally, the boundaries of the Shh TAD do not represent the absolute expression limits of enhancer activity, as expression activity is lost stepwise at a number of genomic positions at the verges of these domains.


Subject(s)
Gene Expression Regulation, Developmental , Hedgehog Proteins/physiology , Animals , Blastocyst/cytology , DNA Transposable Elements , Enhancer Elements, Genetic , Gene Expression Profiling , Genes, Reporter , Genetic Complementation Test , Hedgehog Proteins/genetics , Heterozygote , Introns , Mice , Mice, Transgenic , Models, Genetic , Promoter Regions, Genetic , Protein Structure, Tertiary , Transgenes
9.
Development ; 141(8): 1715-25, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24715461

ABSTRACT

Conservation within intergenic DNA often highlights regulatory elements that control gene expression from a long range. How conservation within a single element relates to regulatory information and how internal composition relates to function is unknown. Here, we examine the structural features of the highly conserved ZRS (also called MFCS1) cis-regulator responsible for the spatiotemporal control of Shh in the limb bud. By systematically dissecting the ZRS, both in transgenic assays and within in the endogenous locus, we show that the ZRS is, in effect, composed of two distinct domains of activity: one domain directs spatiotemporal activity but functions predominantly from a short range, whereas a second domain is required to promote long-range activity. We show further that these two domains encode activities that are highly integrated and that the second domain is crucial in promoting the chromosomal conformational changes correlated with gene activity. During limb bud development, these activities encoded by the ZRS are interpreted differently by the fore limbs and the hind limbs; in the absence of the second domain there is no Shh activity in the fore limb, and in the hind limb low levels of Shh lead to a variant digit pattern ranging from two to four digits. Hence, in the embryo, the second domain stabilises the developmental programme providing a buffer for SHH morphogen activity and this ensures that five digits form in both sets of limbs.


Subject(s)
Limb Buds/embryology , Limb Buds/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Animals , Base Sequence , Chromosomes, Mammalian/chemistry , DNA Mutational Analysis , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Hindlimb/embryology , Hindlimb/metabolism , In Situ Hybridization, Fluorescence , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nucleic Acid Conformation , Phenotype , Point Mutation/genetics , Sequence Deletion/genetics
10.
PLoS Genet ; 9(10): e1003826, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24204287

ABSTRACT

ß-defensin peptides are a family of antimicrobial peptides present at mucosal surfaces, with the main site of expression under normal conditions in the male reproductive tract. Although they kill microbes in vitro and interact with immune cells, the precise role of these genes in vivo remains uncertain. We show here that homozygous deletion of a cluster of nine ß-defensin genes (DefbΔ9) in the mouse results in male sterility. The sperm derived from the mutants have reduced motility and increased fragility. Epididymal sperm isolated from the cauda should require capacitation to induce the acrosome reaction but sperm from the mutants demonstrate precocious capacitation and increased spontaneous acrosome reaction compared to wild-types but have reduced ability to bind the zona pellucida of oocytes. Ultrastructural examination reveals a defect in microtubule structure of the axoneme with increased disintegration in mutant derived sperm present in the epididymis cauda region, but not in caput region or testes. Consistent with premature acrosome reaction, sperm from mutant animals have significantly increased intracellular calcium content. Thus we demonstrate in vivo that ß-defensins are essential for successful sperm maturation, and their disruption leads to alteration in intracellular calcium, inappropriate spontaneous acrosome reaction and profound male infertility.


Subject(s)
Chromosome Deletion , Infertility, Male/genetics , Spermatozoa/metabolism , beta-Defensins/genetics , Animals , Chromosomes/genetics , Chromosomes/metabolism , Infertility, Male/pathology , Male , Mice , Sperm Maturation/genetics , Spermatozoa/pathology
11.
Hum Mol Genet ; 22(25): 5083-95, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-23900076

ABSTRACT

The embryonic epicardium is an important source of cardiovascular precursor cells and paracrine factors that are required for adequate heart formation. Signaling pathways regulated by WT1 that promote heart development have started to be described; however, there is little information on signaling pathways regulated by WT1 that could act in a negative manner. Transcriptome analysis of Wt1KO epicardial cells reveals an unexpected role for WT1 in repressing the expression of interferon-regulated genes that could be involved in a negative regulation of heart morphogenesis. Here, we showed that WT1 is required to repress the expression of the chemokines Ccl5 and Cxcl10 in epicardial cells. We observed an inverse correlation of Wt1 and the expression of Cxcl10 and Ccl5 during epicardium development. Chemokine receptor analyses of hearts from Wt1(gfp/+) mice demonstrate the differential expression of their chemokine receptors in GFP(+) epicardial enriched cells and GFP(-) cells. Functional assays demonstrate that CXCL10 and CCL5 inhibit epicardial cells migration and the proliferation of cardiomyocytes respectively. WT1 regulates the expression levels of Cxcl10 and Ccl5 in epicardial cells directly and indirectly through increasing the levels of IRF7. As epicardial cell reactivation after a myocardial damage is linked with WT1 expression, the present work has potential implications in adult heart repair.


Subject(s)
Chemokine CCL5/biosynthesis , Chemokine CXCL10/biosynthesis , Heart/growth & development , Pericardium/growth & development , WT1 Proteins/genetics , Animals , Chemokine CCL5/genetics , Chemokine CXCL10/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Interferon Regulatory Factor-7/metabolism , Mice , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Pericardium/cytology , Receptors, Chemokine/antagonists & inhibitors , Receptors, Chemokine/metabolism , Signal Transduction , WT1 Proteins/biosynthesis
12.
Trends Genet ; 28(8): 364-73, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22534646

ABSTRACT

Human hands and feet contain bones of a particular size and shape arranged in a precise pattern. The secreted factor sonic hedgehog (SHH) acts through the conserved hedgehog (Hh) signaling pathway to regulate the digital pattern in the limbs of tetrapods (i.e. land-based vertebrates). Genetic analysis is now uncovering a remarkable set of pathogenetic mutations that alter the Hh pathway, thus compromising both digit number and identity. Several of these are regulatory mutations that have the surprising attribute of misdirecting expression of Hh ligands to ectopic sites in the developing limb buds. In addition, other mutations affect a fundamental structural property of the embryonic cell that is essential to Hh signaling. In this review, we focus on the role that the Hh pathway plays in limb development, and how the many human genetic defects in this pathway are providing clues to the mechanisms that regulate limb development.


Subject(s)
Extremities , Limb Deformities, Congenital/metabolism , Signal Transduction , Animals , Body Patterning , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Limb Deformities, Congenital/genetics
13.
Development ; 139(17): 3157-67, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22872084

ABSTRACT

A late phase of HoxD activation is crucial for the patterning and growth of distal structures across the anterior-posterior (A-P) limb axis of mammals. Polycomb complexes and chromatin compaction have been shown to regulate Hox loci along the main body axis in embryonic development, but the extent to which they have a role in limb-specific HoxD expression, an evolutionary adaptation defined by the activity of distal enhancer elements that drive expression of 5' Hoxd genes, has yet to be fully elucidated. We reveal two levels of chromatin topology that differentiate distal limb A-P HoxD activity. Using both immortalised cell lines derived from posterior and anterior regions of distal E10.5 mouse limb buds, and analysis in E10.5 dissected limb buds themselves, we show that there is a loss of polycomb-catalysed H3K27me3 histone modification and a chromatin decompaction over HoxD in the distal posterior limb compared with anterior. Moreover, we show that the global control region (GCR) long-range enhancer spatially colocalises with the 5' HoxD genomic region specifically in the distal posterior limb. This is consistent with the formation of a chromatin loop between 5' HoxD and the GCR regulatory module at the time and place of distal limb bud development when the GCR participates in initiating Hoxd gene quantitative collinearity and Hoxd13 expression. This is the first example of A-P differences in chromatin compaction and chromatin looping in the development of the mammalian secondary body axis (limb).


Subject(s)
Body Patterning/physiology , Chromatin Assembly and Disassembly/physiology , Extremities/embryology , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Blotting, Western , Cell Line , Chromatin Immunoprecipitation , DNA Primers/genetics , Gene Expression Regulation, Developmental/genetics , Histones/metabolism , Image Processing, Computer-Assisted , In Situ Hybridization, Fluorescence , Mice , Microscopy, Fluorescence , Polycomb-Group Proteins , Real-Time Polymerase Chain Reaction , Repressor Proteins/metabolism
14.
Development ; 138(6): 1093-7, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21343363

ABSTRACT

Epicardial-derived signals are key regulators of cardiac embryonic development. An important part of these signals is known to relate to a retinoic acid (RA) receptor-dependent mechanism. RA is a potent morphogen synthesised by Raldh enzymes, Raldh2 being the predominant one in mesodermal tissues. Despite the importance of epicardial retinoid signalling in the heart, the molecular mechanisms controlling cardiac Raldh2 transcription remain unknown. In the current study, we show that Wt1-null epicardial cells display decreased expression of Raldh2 both in vivo and in vitro. Using a RA-responsive reporter, we have confirmed that Wt1-null epicardial cells actually show reduced synthesis of RA. We also demonstrate that Raldh2 is a direct transcriptional target of Wt1 in epicardial cells. A secondary objective of this study was to identify the status of RA-related receptors previously reported to be critical to epicardial biology (PDGFRα,ß; RXRα). PDGFRα and PDGFRß mRNA and protein levels are downregulated in the absence of Wt1, but only Pdgfra expression is rescued by the addition of RA to Wt1-null epicardial cells. RXRα mRNA levels are not affected in Wt1-null epicardial cells. Taken together, our results indicate that Wt1 critically regulates epicardial RA signalling via direct activation of the Raldh2 gene, and identify a role for Wt1 in the regulation of morphogen receptors involved in the proliferation, migration, and differentiation of epicardial and epicardially-derived cells (EPDC).


Subject(s)
Aldehyde Oxidoreductases/genetics , Pericardium/embryology , Tretinoin/metabolism , WT1 Proteins/physiology , Aldehyde Oxidoreductases/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Embryo, Mammalian , Gene Expression Regulation, Developmental/drug effects , Gene Knockdown Techniques , Heart/embryology , Mice , Mice, Transgenic , Pericardium/metabolism , Signal Transduction/genetics , Transcriptional Activation/drug effects , Tretinoin/pharmacology , Tretinoin/physiology
15.
Nat Struct Mol Biol ; 29(9): 891-897, 2022 09.
Article in English | MEDLINE | ID: mdl-36097291

ABSTRACT

The regulatory landscapes of developmental genes in mammals can be complex, with enhancers spread over many hundreds of kilobases. It has been suggested that three-dimensional genome organization, particularly topologically associating domains formed by cohesin-mediated loop extrusion, is important for enhancers to act over such large genomic distances. By coupling acute protein degradation with synthetic activation by targeted transcription factor recruitment, here we show that cohesin, but not CTCF, is required for activation of the target gene Shh by distant enhancers in mouse embryonic stem cells. Cohesin is not required for activation directly at the promoter or by an enhancer located closer to the Shh gene. Our findings support the hypothesis that chromatin compaction via cohesin-mediated loop extrusion allows for genes to be activated by enhancers that are located many hundreds of kilobases away in the linear genome and suggests that cohesin is dispensable for enhancers located more proximally.


Subject(s)
Cell Cycle Proteins , Chromosomal Proteins, Non-Histone , Animals , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Enhancer Elements, Genetic/genetics , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Mammals/genetics , Mice , Transcription Factors/metabolism , Cohesins
16.
Hum Mutat ; 32(12): 1492-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21948517

ABSTRACT

Disruption of the long-range cis-regulation of developmental gene expression is increasingly recognized as a cause of human disease. Here, we report a novel type of long-range cis-regulatory mutation, in which ectopic expression of a gene is driven by an enhancer that is not its own. We have termed this gain of regulatory information as "enhancer adoption." We mapped the breakpoints of a de novo 7q inversion in a child with features of a holoprosencephaly spectrum (HPES) disorder and severe upper limb syndactyly with lower limb synpolydactyly. The HPES plausibly results from the 7q36.3 breakpoint dislocating the sonic hedgehog (SHH) gene from enhancers that are known to drive expression in the early forebrain. However, the limb phenotype cannot be explained by loss of known SHH enhancers. The SHH transcription unit is relocated to 7q22.1, ∼190 kb 3' of a highly conserved noncoding element (HCNE2) within an intron of EMID2. We show that HCNE2 functions as a limb bud enhancer in mouse embryos and drives ectopic expression of Shh in vivo recapitulating the limb phenotype in the child. This developmental genetic mechanism may explain a proportion of the novel or unexplained phenotypes associated with balanced chromosome rearrangements.


Subject(s)
Chromosome Inversion/genetics , Enhancer Elements, Genetic/genetics , Hedgehog Proteins/genetics , Holoprosencephaly/genetics , Syndactyly/genetics , Animals , Child, Preschool , Chromosomes, Human, Pair 7/genetics , Extremities/embryology , Female , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Humans , Limb Buds/embryology , Mice , Mice, Transgenic , Mutation
17.
Front Cell Dev Biol ; 9: 595744, 2021.
Article in English | MEDLINE | ID: mdl-33869166

ABSTRACT

Enhancers that are conserved deep in evolutionary time regulate characteristics held in common across taxonomic classes. Here, deletion of the highly conserved Shh enhancer SBE2 (Shh brain enhancer 2) in mouse markedly reduced Shh expression within the embryonic brain specifically in the rostral diencephalon; however, no abnormal anatomical phenotype was observed. Secondary enhancer activity was subsequently identified which likely mediates low levels of expression. In contrast, when crossing the SBE2 deletion with the Shh null allele, brain and craniofacial development were disrupted; thus, linking SBE2 regulated Shh expression to multiple defects and further enabling the study of the effects of differing levels of Shh on embryogenesis. Development of the hypothalamus, derived from the rostral diencephalon, was disrupted along both the anterior-posterior (AP) and the dorsal-ventral (DV) axes. Expression of DV patterning genes and subsequent neuronal population induction were particularly sensitive to Shh expression levels, demonstrating a novel morphogenic context for Shh. The role of SBE2, which is highlighted by DV gene expression, is to step-up expression of Shh above the minimal activity of the second enhancer, ensuring the necessary levels of Shh in a regional-specific manner. We also show that low Shh levels in the diencephalon disrupted neighbouring craniofacial development, including mediolateral patterning of the bones along the cranial floor and viscerocranium. Thus, SBE2 contributes to hypothalamic morphogenesis and ensures there is coordination with the formation of the adjacent midline cranial bones that subsequently protect the neural tissue.

18.
Nat Commun ; 12(1): 2282, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33863876

ABSTRACT

Acheiropodia, congenital limb truncation, is associated with homozygous deletions in the LMBR1 gene around ZRS, an enhancer regulating SHH during limb development. How these deletions lead to this phenotype is unknown. Using whole-genome sequencing, we fine-mapped the acheiropodia-associated region to 12 kb and show that it does not function as an enhancer. CTCF and RAD21 ChIP-seq together with 4C-seq and DNA FISH identify three CTCF sites within the acheiropodia-deleted region that mediate the interaction between the ZRS and the SHH promoter. This interaction is substituted with other CTCF sites centromeric to the ZRS in the disease state. Mouse knockouts of the orthologous 12 kb sequence have no apparent abnormalities, showcasing the challenges in modelling CTCF alterations in animal models due to inherent motif differences between species. Our results show that alterations in CTCF motifs can lead to a Mendelian condition due to altered enhancer-promoter interactions.


Subject(s)
CCCTC-Binding Factor/genetics , Extremities/embryology , Foot Deformities, Congenital/genetics , Gene Expression Regulation, Developmental , Hand Deformities, Congenital/genetics , Animals , Binding Sites/genetics , Chromatin Immunoprecipitation Sequencing , Disease Models, Animal , Embryo, Mammalian , Enhancer Elements, Genetic/genetics , Exons/genetics , Female , Genetic Loci , Genetic Testing , Hedgehog Proteins/genetics , Humans , Introns/genetics , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Promoter Regions, Genetic/genetics , Sequence Deletion , Species Specificity , Whole Genome Sequencing
19.
Hum Mol Genet ; 17(7): 978-85, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18156157

ABSTRACT

Precise spatial and temporal control of developmental genes is crucial during embryogenesis. Regulatory mutations that cause the misexpression of key developmental genes may underlie a number of developmental abnormalities. The congenital abnormality preaxial polydactyly, extra digits, is an example of this novel class of mutations and is caused by ectopic expression of the signalling molecule Sonic Hedgehog (SHH) in the developing limb bud. Mutations in the long-distant, limb-specific cis-regulator for SHH, called the ZRS, are responsible for the ectopic expression which underlies the abnormality. Here, we show that populations of domestic cats which manifest extra digits, including the celebrated polydactylous Hemingway's cats, also contain mutations within the ZRS. The polydactylous cats add significantly to the number of mutations previously reported in mouse and human and to date, all are single nucleotide substitutions. A mouse transgenic assay shows that these single nucleotide substitutions operate as gain-of-function mutations that activate Shh expression at an ectopic embryonic site; and that the sequence context of the mutation is responsible for a variable regulatory output. The plasticity of the regulatory response correlates with both the phenotypic variability and with species differences. The polydactyly mutations define a new genetic mechanism that results in human congenital abnormalities and identifies a pathogenetic mechanism that may underlie other congenital diseases.


Subject(s)
Body Patterning/genetics , Hedgehog Proteins/genetics , Point Mutation , Polydactyly/genetics , Animals , Base Sequence , Cats , Forelimb/abnormalities , Forelimb/embryology , Gene Expression Regulation, Developmental , Genes, Regulator , Hedgehog Proteins/physiology , Humans , Lac Operon , Mice , Mutagenesis, Site-Directed , Pedigree , Polydactyly/embryology , Regulatory Sequences, Nucleic Acid
20.
Hum Mol Genet ; 17(16): 2417-23, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18463159

ABSTRACT

A locus for triphalangeal thumb, variably associated with pre-axial polydactyly, was previously identified in the zone of polarizing activity regulatory sequence (ZRS), a long range limb-specific enhancer of the Sonic Hedgehog (SHH) gene at human chromosome 7q36.3. Here, we demonstrate that a 295T>C variant in the human ZRS, previously thought to represent a neutral polymorphism, acts as a dominant allele with reduced penetrance. We found this variant in three independently ascertained probands from southern England with triphalangeal thumb, demonstrated significant linkage of the phenotype to the variant (LOD = 4.1), and identified a shared microsatellite haplotype around the ZRS, suggesting that the probands share a common ancestor. An individual homozygous for the 295C allele presented with isolated bilateral triphalangeal thumb resembling the heterozygous phenotype, suggesting that the variant is largely dominant to the wild-type allele. As a functional test of the pathogenicity of the 295C allele, we utilized a mutated ZRS construct to demonstrate that it can drive ectopic anterior expression of a reporter gene in the developing mouse forelimb. We conclude that the 295T>C variant is in fact pathogenic and, in southern England, appears to be the most common cause of triphalangeal thumb. Depending on the dispersal of the founding mutation, it may play a wider role in the aetiology of this disorder.


Subject(s)
Enhancer Elements, Genetic , Extremities/growth & development , Gene Expression Regulation, Developmental , Hand Deformities, Congenital/genetics , Hedgehog Proteins/genetics , Polydactyly/genetics , Animals , Base Sequence , Chromosomes, Human, Pair 7/genetics , Cohort Studies , Extremities/embryology , Female , Genetic Linkage , Hand Deformities, Congenital/embryology , Haplotypes , Humans , Male , Mice , Mice, Transgenic , Microsatellite Repeats , Molecular Sequence Data , Mutation , Pedigree , Polydactyly/embryology , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL