Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.866
Filter
1.
Nat Commun ; 15(1): 4820, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844479

ABSTRACT

Chondrocyte differentiation controls skeleton development and stature. Here we provide a comprehensive map of chondrocyte-specific enhancers and show that they provide a mechanistic framework through which non-coding genetic variants can influence skeletal development and human stature. Working with fetal chondrocytes isolated from mice bearing a Col2a1 fluorescent regulatory sensor, we identify 780 genes and 2'704 putative enhancers specifically active in chondrocytes using a combination of RNA-seq, ATAC-seq and H3K27ac ChIP-seq. Most of these enhancers (74%) show pan-chondrogenic activity, with smaller populations being restricted to limb (18%) or trunk (8%) chondrocytes only. Notably, genetic variations overlapping these enhancers better explain height differences than those overlapping non-chondrogenic enhancers. Finally, targeted deletions of identified enhancers at the Fgfr3, Col2a1, Hhip and, Nkx3-2 loci confirm their role in regulating cognate genes. This enhancer map provides a framework for understanding how genes and non-coding variations influence bone development and diseases.


Subject(s)
Chondrocytes , Chondrogenesis , Enhancer Elements, Genetic , Receptor, Fibroblast Growth Factor, Type 3 , Animals , Enhancer Elements, Genetic/genetics , Humans , Chondrocytes/metabolism , Chondrocytes/cytology , Mice , Chondrogenesis/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Collagen Type II/genetics , Collagen Type II/metabolism , Gene Expression Regulation, Developmental , Bone Development/genetics , Extremities/embryology , Male , Cell Differentiation/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Female
2.
Dev Biol ; 512: 1-10, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38657748

ABSTRACT

Precise regulation of gene expression is of utmost importance during cell fate specification. DNA methylation is a key epigenetic mechanism that plays a significant role in the regulation of cell fate by recruiting repression proteins or inhibiting the binding of transcription factors to DNA to regulate gene expression. Limb development is a well-established model for understanding cell fate decisions, and the formation of skeletal elements is coordinated through a sequence of events that control chondrogenesis spatiotemporally. It has been established that epigenetic control participates in cartilage maturation. However, further investigation is required to determine its role in the earliest stages of chondrocyte differentiation. This study investigates how the DNA methylation environment affects cell fate divergence during the early chondrogenic events. Our research has shown for the first time that inhibiting DNA methylation in interdigital tissue with 5-azacytidine results in the formation of an ectopic digit. This discovery suggested that DNA methylation dynamics could regulate the fate of cells between chondrogenesis and cell death during autopod development. Our in vitro findings indicate that DNA methylation at the early stages of chondrogenesis is integral in regulating condensation by controlling cell adhesion and proapoptotic genes. As a result, the dynamics of methylation and demethylation are crucial in governing chondrogenesis and cell death during different stages of limb chondrogenesis.


Subject(s)
Cell Differentiation , Chondrocytes , Chondrogenesis , DNA Methylation , Extremities , DNA Methylation/genetics , Chondrogenesis/genetics , Animals , Extremities/embryology , Cell Differentiation/genetics , Chondrocytes/metabolism , Chondrocytes/cytology , Azacitidine/pharmacology , Gene Expression Regulation, Developmental , Chick Embryo , Epigenesis, Genetic , Apoptosis/genetics
3.
Nature ; 626(7997): 151-159, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38233525

ABSTRACT

Enhancers control the location and timing of gene expression and contain the majority of variants associated with disease1-3. The ZRS is arguably the most well-studied vertebrate enhancer and mediates the expression of Shh in the developing limb4. Thirty-one human single-nucleotide variants (SNVs) within the ZRS are associated with polydactyly4-6. However, how this enhancer encodes tissue-specific activity, and the mechanisms by which SNVs alter the number of digits, are poorly understood. Here we show that the ETS sites within the ZRS are low affinity, and identify a functional ETS site, ETS-A, with extremely low affinity. Two human SNVs and a synthetic variant optimize the binding affinity of ETS-A subtly from 15% to around 25% relative to the strongest ETS binding sequence, and cause polydactyly with the same penetrance and severity. A greater increase in affinity results in phenotypes that are more penetrant and more severe. Affinity-optimizing SNVs in other ETS sites in the ZRS, as well as in ETS, interferon regulatory factor (IRF), HOX and activator protein 1 (AP-1) sites within a wide variety of enhancers, cause gain-of-function gene expression. The prevalence of binding sites with suboptimal affinity in enhancers creates a vulnerability in genomes whereby SNVs that optimize affinity, even slightly, can be pathogenic. Searching for affinity-optimizing SNVs in genomes could provide a mechanistic approach to identify causal variants that underlie enhanceropathies.


Subject(s)
Enhancer Elements, Genetic , Extremities , Polydactyly , Proto-Oncogene Proteins c-ets , Humans , Enhancer Elements, Genetic/genetics , Extremities/embryology , Extremities/pathology , Gain of Function Mutation , Homeodomain Proteins/metabolism , Interferon Regulatory Factors/metabolism , Organ Specificity/genetics , Penetrance , Phenotype , Polydactyly/embryology , Polydactyly/genetics , Polydactyly/pathology , Polymorphism, Single Nucleotide , Protein Binding , Proto-Oncogene Proteins c-ets/metabolism , Transcription Factor AP-1/metabolism
4.
Proc Natl Acad Sci U S A ; 119(27): e2119297119, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35776546

ABSTRACT

Regenerating animals have the ability to reproduce body parts that were originally made in the embryo and subsequently lost due to injury. Understanding whether regeneration mirrors development is an open question in most regenerative species. Here, we take a transcriptomics approach to examine whether leg regeneration shows similar temporal patterns of gene expression as leg development in the embryo, in the crustacean Parhyale hawaiensis. We find that leg development in the embryo shows stereotypic temporal patterns of gene expression. In contrast, the dynamics of gene expression during leg regeneration show a higher degree of variation related to the physiology of individual animals. A major driver of this variation is the molting cycle. We dissect the transcriptional signals of individual physiology and regeneration to obtain clearer temporal signals marking distinct phases of leg regeneration. Comparing the transcriptional dynamics of development and regeneration we find that, although the two processes use similar sets of genes, the temporal patterns in which these genes are deployed are different and cannot be systematically aligned.


Subject(s)
Amphipoda , Extremities , Regeneration , Amphipoda/embryology , Amphipoda/genetics , Animals , Embryo, Nonmammalian , Extremities/embryology , Gene Expression , Regeneration/genetics
5.
Development ; 149(9)2022 05 01.
Article in English | MEDLINE | ID: mdl-35552394

ABSTRACT

In central nervous system vascular endothelial cells, signaling via the partially redundant ligands WNT7A and WNT7B requires two co-activator proteins, GPR124 and RECK. WNT7A and RECK have been shown previously to play a role in limb development, but the mechanism of RECK action in this context is unknown. The roles of WNT7B and GPR124 in limb development have not been investigated. Using combinations of conventional and/or conditional loss-of-function alleles for mouse Wnt7a, Wnt7b, Gpr124 and Reck, including a Reck allele that codes for a protein that is specifically defective in WNT7A/WNT7B signaling, we show that reductions in ligand and/or co-activator function synergize to cause reduced and dysmorphic limb bone growth. Two additional limb phenotypes - loss of distal Lmx1b expression and ectopic growth of nail-like structures - occur with reduced Wnt7a/Wnt7b gene copy number and, respectively, with Reck mutations and with combined Reck and Gpr124 mutations. A third limb phenotype - bleeding into a digit - occurs with the most severe combinations of Wnt7a/Wnt7b, Reck and Gpr124 mutations. These data imply that the WNT7A/WNT7B-FRIZZLED-LRP5/LRP6-GPR124-RECK signaling system functions as an integral unit in limb development.


Subject(s)
Embryo, Mammalian/metabolism , Extremities/embryology , GPI-Linked Proteins/metabolism , Receptors, G-Protein-Coupled , Wnt Proteins , Animals , Central Nervous System/metabolism , Endothelial Cells/metabolism , Ligands , Mice , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Wnt Proteins/genetics , Wnt Proteins/metabolism
6.
Nat Commun ; 13(1): 304, 2022 01 13.
Article in English | MEDLINE | ID: mdl-35027568

ABSTRACT

The evolution of uniquely human traits likely entailed changes in developmental gene regulation. Human Accelerated Regions (HARs), which include transcriptional enhancers harboring a significant excess of human-specific sequence changes, are leading candidates for driving gene regulatory modifications in human development. However, insight into whether HARs alter the level, distribution, and timing of endogenous gene expression remains limited. We examined the role of the HAR HACNS1 (HAR2) in human evolution by interrogating its molecular functions in a genetically humanized mouse model. We find that HACNS1 maintains its human-specific enhancer activity in the mouse embryo and modifies expression of Gbx2, which encodes a transcription factor, during limb development. Using single-cell RNA-sequencing, we demonstrate that Gbx2 is upregulated in the limb chondrogenic mesenchyme of HACNS1 homozygous embryos, supporting that HACNS1 alters gene expression in cell types involved in skeletal patterning. Our findings illustrate that humanized mouse models provide mechanistic insight into how HARs modified gene expression in human evolution.


Subject(s)
Gene Expression Regulation , Genome , Models, Genetic , Animals , Base Sequence , Cell Differentiation/genetics , Chondrocytes/cytology , Chondrogenesis/genetics , Embryo, Mammalian/metabolism , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Extremities/embryology , Gene Expression Profiling , Gene Knock-In Techniques , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Homozygote , Humans , Mesoderm/embryology , Mesoderm/metabolism , Mice, Inbred C57BL , Pan troglodytes , Promoter Regions, Genetic/genetics , Time Factors
7.
Dev Biol ; 483: 76-88, 2022 03.
Article in English | MEDLINE | ID: mdl-34973174

ABSTRACT

The chick limb bud has plasticity to reconstruct a normal skeletal pattern after a part of mesenchymal mass is excised to make a hole in its early stage of development. To understand the details of hole closure and re-establishment of normal limb axes to reconstruct a normal limb skeleton, we focused on cellular and molecular changes during hole repair and limb restoration. We excised a cube-shaped mass of mesenchymal cells from the medial region of chick hindlimb bud (stage 23) and observed the following morphogenesis. The hole had closed by 15 â€‹h after excision, followed by restoration of the limb bud morphology, and the cartilage pattern was largely restored by 48 â€‹h. Lineage analysis of the mesenchymal cells showed that cells at the anterior and posterior margins of the hole were adjoined at the hole closure site, whereas cells at the proximal and distal margins were not. To investigate cell polarity during hole repair, we analyzed intracellular positioning of the Golgi apparatus relative to the nuclei. We found that the Golgi apparatus tended to be directed toward the hole among cells at the anterior and posterior margins but not among cells at identical positions in normal limb buds or cells at the proximal and distal hole margins. In the manipulated limb buds, the frequency of cell proliferation was maintained compared with the control side. Tbx3 expression, which was usually restricted to anterior and posterior margins of the limb bud, was temporarily expanded medially and then reverted to a normal pattern as limb reconstruction proceeded, with Tbx3 negative cells reappearing in the medial regions of the limb buds. Thus, mesenchymal hole closure and limb reconstruction are mainly mediated by cells at the anterior and posterior hole margins. These results suggest that adjustment of cellular properties along the anteroposterior axis is crucial to restore limb damage and reconstruct normal skeletal patterns.


Subject(s)
Body Patterning/physiology , Limb Buds/cytology , Limb Buds/embryology , Mesenchymal Stem Cells/metabolism , Mesoderm/cytology , Mesoderm/embryology , Skeleton/embryology , Animals , Avian Proteins/metabolism , Cell Nucleus/metabolism , Cell Polarity/physiology , Cell Proliferation/physiology , Chick Embryo , Extremities/embryology , Golgi Apparatus/metabolism , Hindlimb/embryology , Signal Transduction/physiology , Skeleton/cytology , Skeleton/metabolism , T-Box Domain Proteins/metabolism
8.
Dev Biol ; 482: 124-134, 2022 02.
Article in English | MEDLINE | ID: mdl-34942194

ABSTRACT

Leg axis formation in Drosophila is organized by Wingless (Wg) and Decapentaplegic (Dpp) that control a number of downstream factors to pattern the dorsoventral (DV) and proximodistal (PD) axis. The T-box genes are important downstream factors mainly involved in dorsoventral leg axis formation. The ventral side is specified by H15 and midline, whereas optomotor-blind (omb) and Dorsocross (Doc1) are factors to specify dorsal cell fates. We show here that omb also organizes PD leg axis patterning in the beetle Tribolium castaneum. In the legs, Tc-omb is expressed along the dorsal side and represses ventral factors like wg and H15. Intriguingly, removing Tc-omb function leads to the activation of the Dpp pathway along the dorsal side of the legs, thus mimicking normal dpp expression in Drosophila. Dpp activity along the dorsal side leads to altered expression of proximal-distal patterning genes such as Distal-less (Dll) and dachshund (dac). Our results indicate a cell-autonomous activation of Dll and repression of dac by dpp. These findings are compatible with the cross-regulatory "cascade model" of proximal-distal leg imaginal disc patterning of Drosophila.


Subject(s)
Body Patterning/genetics , Extremities/embryology , Nerve Tissue Proteins/genetics , T-Box Domain Proteins/genetics , Tribolium/embryology , Animals , Tribolium/genetics
9.
Cells ; 10(9)2021 09 17.
Article in English | MEDLINE | ID: mdl-34572097

ABSTRACT

MT1-MMP/MMP14 belongs to a subgroup of the matrix metalloproteinases family that presents a transmembrane domain, with a cytosolic tail and the catalytic site exposed to the extracellular space. Deficient mice for this enzyme result in early postnatal death and display severe defects in skeletal, muscle and lung development. By using a transgenic line expressing the LacZ reporter under the control of the endogenous Mt1-mmp promoter, we reported a dynamic spatiotemporal expression pattern for Mt1-mmp from early embryonic to perinatal stages during cardiovascular development and brain formation. Thus, Mt1-mmp shows expression in the endocardium of the heart and the truncus arteriosus by E8.5, and is also strongly detected during vascular system development as well as in endothelial cells. In the brain, LacZ reporter expression was detected in the olfactory bulb, the rostral cerebral cortex and the caudal mesencephalic tectum. LacZ-positive cells were observed in neural progenitors of the spinal cord, neural crest cells and the intersomitic region. In the limb, Mt1-mmp expression was restricted to blood vessels, cartilage primordium and muscles. Detection of the enzyme was confirmed by Western blot and immunohistochemical analysis. We suggest novel functions for this metalloproteinase in angiogenesis, endocardial formation and vascularization during organogenesis. Moreover, Mt1-mmp expression revealed that the enzyme may contribute to heart, muscle and brain throughout development.


Subject(s)
Cardiovascular System/metabolism , Embryo, Mammalian/metabolism , Embryonic Development , Eye/metabolism , Matrix Metalloproteinase 14/metabolism , Morphogenesis , Nervous System/metabolism , Animals , Cardiovascular System/embryology , Cells, Cultured , Embryo, Mammalian/cytology , Extremities/embryology , Extremities/physiology , Eye/embryology , Matrix Metalloproteinase 14/genetics , Mice , Mice, Inbred C57BL , Nervous System/embryology
10.
Dev Cell ; 56(14): 2089-2102.e11, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34242585

ABSTRACT

In ribosomopathies, perturbed expression of ribosome components leads to tissue-specific phenotypes. What accounts for such tissue-selective manifestations as a result of mutations in the ribosome, a ubiquitous cellular machine, has remained a mystery. Combining mouse genetics and in vivo ribosome profiling, we observe limb-patterning phenotypes in ribosomal protein (RP) haploinsufficient embryos, and we uncover selective translational changes of transcripts that controlling limb development. Surprisingly, both loss of p53, which is activated by RP haploinsufficiency, and augmented protein synthesis rescue these phenotypes. These findings are explained by the finding that p53 functions as a master regulator of protein synthesis, at least in part, through transcriptional activation of 4E-BP1. 4E-BP1, a key translational regulator, in turn, facilitates selective changes in the translatome downstream of p53, and this thereby explains how RP haploinsufficiency may elicit specificity to gene expression. These results provide an integrative model to help understand how in vivo tissue-specific phenotypes emerge in ribosomopathies.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Extremities/embryology , Haploinsufficiency , Protein Biosynthesis , Protein Processing, Post-Translational , Ribosomal Proteins/physiology , Tumor Suppressor Protein p53/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Body Patterning , Cell Cycle Proteins/genetics , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Phenotype , Ribosomes/metabolism
11.
J Autoimmun ; 123: 102709, 2021 09.
Article in English | MEDLINE | ID: mdl-34304080

ABSTRACT

Chromatin remodeling, and a persistent histone 3 lysine 27 acetylation (H3K27ac) in particular, are associated with a sustained inflammatory response of synovial fibroblasts (SF) in rheumatoid arthritis (RA). Here we investigated individual functions of the writers of H3K27ac marks, the homologues histone acetyl transferases (HAT) CBP and p300, in controlling the constitutive and inflammatory gene expression in RA SF. We applied a silencing strategy, followed by RNA-sequencing and pathway analysis, complemented with the treatment of SF with inhibitors targeting the HAT (C646) or bromo domains (I-CBP) of CBP and p300. We showed that CBP and p300 undertook overlapping and, in particular at gene levels, distinct regulatory functions in SF. p300 is the major HAT for H3K27ac in SF and regulated more diverse pathways than CBP. Whereas both factors regulated genes associated with extracellular matrix remodeling, adhesion and proliferation, p300 specifically controlled developmental genes associated with limb development. Silencing of CBP specifically down regulated the TNF-induced expression of interferon-signature genes. In contrast, silencing of p300 resulted in anti- and pro-inflammatory effects. Integration of data sets derived from RNA-sequencing and chromatin immunoprecipitation sequencing for H3K27ac revealed that changes in gene expression after CBP or p300 silencing could be only partially explained by changes in levels of H3K27ac. Inhibition of CBP/p300 using HAT and bromo domain inhibitors strongly mirrored effects obtained by silencing of p300, including anti- and pro-inflammatory effects, indicating that such inhibitors are not sufficient to be used as anti-inflammatory drugs.


Subject(s)
CREB-Binding Protein/physiology , Inflammation/etiology , p300-CBP Transcription Factors/physiology , Aged , Aged, 80 and over , Animals , CREB-Binding Protein/antagonists & inhibitors , Cell Proliferation , Chromatin Assembly and Disassembly , Extracellular Matrix/physiology , Extremities/embryology , Female , Fibroblasts/physiology , Humans , Male , Middle Aged , Synoviocytes/physiology , p300-CBP Transcription Factors/antagonists & inhibitors
12.
Nat Commun ; 12(1): 3851, 2021 06 22.
Article in English | MEDLINE | ID: mdl-34158501

ABSTRACT

Positional information driving limb muscle patterning is contained in connective tissue fibroblasts but not in myogenic cells. Limb muscles originate from somites, while connective tissues originate from lateral plate mesoderm. With cell and genetic lineage tracing we challenge this model and identify an unexpected contribution of lateral plate-derived fibroblasts to the myogenic lineage, preferentially at the myotendinous junction. Analysis of single-cell RNA-sequencing data from whole limbs at successive developmental stages identifies a population displaying a dual muscle and connective tissue signature. BMP signalling is active in this dual population and at the tendon/muscle interface. In vivo and in vitro gain- and loss-of-function experiments show that BMP signalling regulates a fibroblast-to-myoblast conversion. These results suggest a scenario in which BMP signalling converts a subset of lateral plate mesoderm-derived cells to a myogenic fate in order to create a boundary of fibroblast-derived myonuclei at the myotendinous junction that controls limb muscle patterning.


Subject(s)
Body Patterning/genetics , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Muscle, Skeletal/metabolism , Somites/metabolism , Animals , Cell Lineage/genetics , Cells, Cultured , Chick Embryo , Extremities/embryology , Fibroblasts/cytology , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/metabolism , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Muscle Development/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/embryology , Reverse Transcriptase Polymerase Chain Reaction , Somites/cytology , Somites/embryology
13.
Dev Biol ; 477: 49-63, 2021 09.
Article in English | MEDLINE | ID: mdl-34010606

ABSTRACT

Previous studies on mouse embryo limbs have established that interzone mesenchymal progenitor cells emerging at each prescribed joint site give rise to joint tissues over fetal time. These incipient tissues undergo structural maturation and morphogenesis postnatally, but underlying mechanisms of regulation remain unknown. Hox11 genes dictate overall zeugopod musculoskeletal patterning and skeletal element identities during development. Here we asked where these master regulators are expressed in developing limb joints and whether they are maintained during postnatal zeugopod joint morphogenesis. We found that Hoxa11 was predominantly expressed and restricted to incipient wrist and ankle joints in E13.5 mouse embryos, and became apparent in medial and central regions of knees by E14.5, though remaining continuously dormant in elbow joints. Closer examination revealed that Hoxa11 initially characterized interzone and neighboring cells and was then restricted to nascent articular cartilage, intra joint ligaments and structures such as meniscal horns over prenatal time. Postnatally, articular cartilage progresses from a nondescript cell-rich, matrix-poor tissue to a highly structured, thick, zonal and mechanically competent tissue with chondrocyte columns over time, most evident at sites such as the tibial plateau. Indeed, Hox11 expression (primarily Hoxa11) was intimately coupled to such morphogenetic processes and, in particular, to the topographical rearrangement of chondrocytes into columns within the intermediate and deep zones of tibial plateau that normally endures maximal mechanical loads. Revealingly, these expression patterns were maintained even at 6 months of age. In sum, our data indicate that Hox11 genes remain engaged well beyond embryonic synovial joint patterning and are specifically tied to postnatal articular cartilage morphogenesis into a zonal and resilient tissue. The data demonstrate that Hox11 genes characterize adult, terminally differentiated, articular chondrocytes and maintain region-specificity established in the embryo.


Subject(s)
Cartilage, Articular/embryology , Chondrogenesis/genetics , Genes, Homeobox , Synovial Membrane/embryology , Animals , Chondrogenesis/physiology , Extremities/embryology , Gene Expression Regulation, Developmental , Genes, Reporter , Green Fluorescent Proteins/genetics , Mice
14.
Cells ; 10(4)2021 04 15.
Article in English | MEDLINE | ID: mdl-33921015

ABSTRACT

During limb formation in vertebrates with free digits, the interdigital mesoderm is eliminated by a massive degeneration process that involves apoptosis and cell senescence. The degradation process is preceded by intense DNA damage in zones located close to methylated DNA, accompanied by the activation of the DNA repair response. In this study, we show that trimethylated histone 3 (H3K4me3, H3K9me3, and H3K27me3) overlaps with zones positive for 5mC in the nuclei of interdigital cells. This pattern contrasts with the widespread distribution of acetylated histones (H3K9ac and H4ac) and the histone variant H3.3 throughout the nucleoplasm. Consistent with the intense labeling of acetylated histones, the histone deacetylase genes Hdac1, Hdac2, Hdac3, and Hdac8, and at a more reduced level, Hdac10, are expressed in the interdigits. Furthermore, local treatments with the histone deacetylase inhibitor trichostatin A, which promotes an open chromatin state, induces massive cell death and transcriptional changes reminiscent of, but preceding, the physiological process of interdigit remodeling. Together, these findings suggest that the epigenetic profile of the interdigital mesoderm contributes to the sensitivity to DNA damage that precedes apoptosis during tissue regression.


Subject(s)
Epigenesis, Genetic , Extremities/embryology , Histones/metabolism , Animals , Cell Death/drug effects , Chick Embryo , DNA Damage/genetics , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Developmental/drug effects , Histone Deacetylases/metabolism , Histones/genetics , Hydroxamic Acids/pharmacology , Microsurgery , Protein Processing, Post-Translational/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism
15.
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
16.
Dis Model Mech ; 14(4)2021 04 01.
Article in English | MEDLINE | ID: mdl-33771841

ABSTRACT

Fetal activity in utero is a normal part of pregnancy and reduced or absent movement can lead to long-term skeletal defects, such as Fetal Akinesia Deformation Sequence, joint dysplasia and arthrogryposis. A variety of animal models with decreased or absent embryonic movements show a consistent set of developmental defects, providing insight into the aetiology of congenital skeletal abnormalities. At developing joints, defects include reduced joint interzones with frequent fusion of cartilaginous skeletal rudiments across the joint. At the spine, defects include shortening and a spectrum of curvature deformations. An important question, with relevance to possible therapeutic interventions for human conditions, is the capacity for recovery with resumption of movement following short-term immobilisation. Here, we use the well-established chick model to compare the effects of sustained immobilisation from embryonic day (E)4-10 to two different recovery scenarios: (1) natural recovery from E6 until E10 and (2) the addition of hyperactive movement stimulation during the recovery period. We demonstrate partial recovery of movement and partial recovery of joint development under both recovery conditions, but no improvement in spine defects. The joints examined (elbow, hip and knee) showed better recovery in hindlimb than forelimb, with hyperactive mobility leading to greater recovery in the knee and hip. The hip joint showed the best recovery with improved rudiment separation, tissue organisation and commencement of cavitation. This work demonstrates that movement post paralysis can partially recover specific aspects of joint development, which could inform therapeutic approaches to ameliorate the effects of human fetal immobility. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Embryo, Nonmammalian/pathology , Joints/embryology , Movement , Paralysis/embryology , Animals , Bone Development , Chick Embryo , Extremities/embryology , Extremities/pathology
17.
Cell ; 184(4): 899-911.e13, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33545089

ABSTRACT

Changes in appendage structure underlie key transitions in vertebrate evolution. Addition of skeletal elements along the proximal-distal axis facilitated critical transformations, including the fin-to-limb transition that permitted generation of diverse modes of locomotion. Here, we identify zebrafish mutants that form supernumerary long bones in their pectoral fins. These new bones integrate into musculature, form joints, and articulate with neighboring elements. This phenotype is caused by activating mutations in previously unrecognized regulators of appendage patterning, vav2 and waslb, that function in a common pathway. This pathway is required for appendage development across vertebrates, and loss of Wasl in mice causes defects similar to those seen in murine Hox mutants. Concordantly, formation of supernumerary bones requires Hox11 function, and mutations in the vav2/wasl pathway drive enhanced expression of hoxa11b, indicating developmental homology with the forearm. Our findings reveal a latent, limb-like pattern ability in fins that is activated by simple genetic perturbation.


Subject(s)
Bone and Bones/embryology , Extremities/embryology , Zebrafish/embryology , Actins/metabolism , Animal Fins/embryology , Animals , Base Sequence , Body Patterning , CRISPR-Cas Systems/genetics , Cell Lineage , Epistasis, Genetic , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Genes, Reporter , HeLa Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Mutation/genetics , Phenotype , Phylogeny , Signal Transduction/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
18.
Am J Med Genet A ; 185(5): 1379-1387, 2021 05.
Article in English | MEDLINE | ID: mdl-33522143

ABSTRACT

Decades of clinical, pathological, and epidemiological study and the recent application of advanced microarray and gene sequencing technologies have led to an understanding of the causes and pathogenesis of most recognized patterns of malformation. Still, there remain a number of patterns of malformation whose pathogenesis has not been established. Six such patterns of malformation are sirenomelia, VACTERL association, OEIS complex, limb-body wall defect (LBWD), urorectal septum malformation (URSM) sequence, and MURCS association, all of which predominantly affect caudal structures. On the basis of the overlap of the component malformations, the co-occurrence in individual fetuses, and the findings on fetal examination, a common pathogenesis is proposed for these patterns of malformation. The presence of a single artery in the umbilical cord provides a visible clue to the pathogenesis of all cases of sirenomelia and 30%-50% of cases of VACTERL association, OEIS complex, URSM sequence, and LBWD. The single artery is formed by a coalescence of arteries that supply the yolk sac, arises from the descending aorta high in the abdominal cavity, and redirects blood flow from the developing caudal structures of the embryo to the placenta. This phenomenon during embryogenesis is termed vitelline vascular steal.


Subject(s)
46, XX Disorders of Sex Development/diagnosis , Abnormalities, Multiple/diagnosis , Anal Canal/abnormalities , Congenital Abnormalities/diagnosis , Ectromelia/diagnosis , Esophagus/abnormalities , Heart Defects, Congenital/physiopathology , Kidney/abnormalities , Limb Deformities, Congenital/physiopathology , Mullerian Ducts/abnormalities , Spine/abnormalities , Trachea/abnormalities , 46, XX Disorders of Sex Development/physiopathology , Abnormalities, Multiple/physiopathology , Anal Canal/blood supply , Anal Canal/physiopathology , Anus, Imperforate/physiopathology , Aorta/pathology , Arteries/pathology , Congenital Abnormalities/physiopathology , Ectromelia/physiopathology , Embryo, Mammalian , Esophagus/blood supply , Esophagus/physiopathology , Extremities/blood supply , Extremities/embryology , Extremities/growth & development , Female , Fetus , Hernia, Umbilical/physiopathology , Humans , Kidney/blood supply , Kidney/physiopathology , Mullerian Ducts/blood supply , Mullerian Ducts/physiopathology , Pregnancy , Scoliosis/physiopathology , Spine/blood supply , Spine/physiopathology , Torso/blood supply , Torso/physiopathology , Trachea/blood supply , Trachea/physiopathology , Umbilical Cord/blood supply , Umbilical Cord/physiopathology , Urogenital Abnormalities/physiopathology
19.
Elife ; 102021 02 09.
Article in English | MEDLINE | ID: mdl-33560225

ABSTRACT

How genetic changes are linked to morphological novelties and developmental constraints remains elusive. Here, we investigate genetic apparatuses that distinguish fish fins from tetrapod limbs by analyzing transcriptomes and open-chromatin regions (OCRs). Specifically, we compared mouse forelimb buds with the pectoral fin buds of an elasmobranch, the brown-banded bamboo shark (Chiloscyllium punctatum). A transcriptomic comparison with an accurate orthology map revealed both a mass heterochrony and hourglass-shaped conservation of gene expression between fins and limbs. Furthermore, open-chromatin analysis suggested that access to conserved regulatory sequences is transiently increased during mid-stage limb development. During this stage, stage-specific and tissue-specific OCRs were also enriched. Together, early and late stages of fin/limb development are more permissive to mutations than middle stages, which may have contributed to major morphological changes during the fin-to-limb evolution. We hypothesize that the middle stages are constrained by regulatory complexity that results from dynamic and tissue-specific transcriptional controls.


Animals come in all shapes and sizes. This diversity arose through genetic mutations during evolution, but it is unclear exactly how these variations led to the formation of new shapes. There is increasing evidence to suggest that not all shapes are possible and that variability between animals is limited by a phenomenon known as "developmental constraint". These limitations direct parts of the body towards a specific shape as they develop in the embryo. Therefore, understanding the mechanisms underlying these developmental constraints could help explain how different body shapes evolved. The limbs of humans and other mammals evolved from the fins of fish, and this transition is often used to study the role developmental constraints play in evolution. This is an ideal model as there is already a detailed fossil record mapping this evolutionary event, and data pinpointing some of the genes involved in the development of limbs and fins. But this data is incomplete, and a full comparison between the genes activated in the fin and the limb during embryonic development had not been achieved. This is because most fish used for research have undergone recent genetic changes, making it hard to spot which genetic differences are linked to the evolution of the limb. To overcome this barrier, Onimaru et al. compared genetic data from the developing limbs of mice to the developing fins of the brown-banded bamboo shark, which evolves much slower than other fish. This revealed that although many genes commonly played a role in the development of the fin and the limb in the embryo, the activity of these shared genes was not the same. For example, genes that switched on in the late stages of limb development, switched off in the late stages of fin development. But in the middle of development, those differences were relatively small and both species activated very similar sets of genes. Many of these genes were pleiotropic, which means they have important roles in other tissues and therefore mutate less often. This suggests that the mid-stage of limb development is under the strongest level of constraint. Darwin's theory of natural selection explains that mutations drive evolution. But the theory cannot predict what kinds of new body shapes new mutations will produce. Understanding how the activity levels of different genes affect development could help to fill this knowledge gap. This has potential medical applications, for example, understanding why some genetic changes cause more serious problems than others. This work suggests that mutations in genes that are active during the mid-stage of limb development may have the most serious impact.


Subject(s)
Animal Fins/embryology , Biological Evolution , Embryo, Mammalian/embryology , Embryo, Nonmammalian/embryology , Limb Buds/embryology , Sharks/embryology , Animals , Extremities/embryology , Mice , Phylogeny
20.
Nat Commun ; 12(1): 439, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33469032

ABSTRACT

Developmental genes are often regulated by multiple elements with overlapping activity. Yet, in most cases, the relative function of those elements and their contribution to endogenous gene expression remain poorly characterized. An example of this phenomenon is that distinct sets of enhancers have been proposed to direct Fgf8 in the limb apical ectodermal ridge and the midbrain-hindbrain boundary. Using in vivo CRISPR/Cas9 genome engineering, we functionally dissect this complex regulatory ensemble and demonstrate two distinct regulatory logics. In the apical ectodermal ridge, the control of Fgf8 expression appears distributed between different enhancers. In contrast, we find that in the midbrain-hindbrain boundary, one of the three active enhancers is essential while the other two are dispensable. We further dissect the essential midbrain-hindbrain boundary enhancer to reveal that it is also composed by a mixture of essential and dispensable modules. Cross-species transgenic analysis of this enhancer suggests that its composition may have changed in the vertebrate lineage.


Subject(s)
Embryonic Development/genetics , Enhancer Elements, Genetic/genetics , Fibroblast Growth Factor 8/genetics , Gene Expression Regulation, Developmental , Genetic Engineering/methods , Animals , CRISPR-Cas Systems/genetics , Ectoderm/embryology , Embryo, Mammalian , Extremities/embryology , Feasibility Studies , Female , Fibroblast Growth Factor 8/metabolism , Gene Regulatory Networks , Male , Mesencephalon/embryology , Mice , Mice, Transgenic , Rhombencephalon/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...