Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.364
Filter
1.
Nat Commun ; 15(1): 6948, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138165

ABSTRACT

Cranial sutures separate neighboring skull bones and are sites of bone growth. A key question is how osteogenic activity is controlled to promote bone growth while preventing aberrant bone fusions during skull expansion. Using single-cell transcriptomics, lineage tracing, and mutant analysis in zebrafish, we uncover key developmental transitions regulating bone formation at sutures during skull expansion. In particular, we identify a subpopulation of mesenchyme cells in the mid-suture region that upregulate a suite of genes including BMP antagonists (e.g. grem1a) and pro-angiogenic factors. Lineage tracing with grem1a:nlsEOS reveals that this mid-suture subpopulation is largely non-osteogenic. Moreover, combinatorial mutation of BMP antagonists enriched in this mid-suture subpopulation results in increased BMP signaling in the suture, misregulated bone formation, and abnormal suture morphology. These data reveal establishment of a non-osteogenic mesenchyme population in the mid-suture region that restricts bone formation through local BMP antagonism, thus ensuring proper suture morphology.


Subject(s)
Bone Morphogenetic Proteins , Cranial Sutures , Mesoderm , Osteogenesis , Zebrafish Proteins , Zebrafish , Animals , Zebrafish/embryology , Zebrafish/genetics , Cranial Sutures/metabolism , Cranial Sutures/embryology , Cranial Sutures/growth & development , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/genetics , Mesoderm/metabolism , Mesoderm/embryology , Mesoderm/cytology , Gene Expression Regulation, Developmental , Signal Transduction , Skull/embryology , Single-Cell Analysis , Mutation
2.
Dev Cell ; 59(16): 2118-2133.e8, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39106861

ABSTRACT

Pluripotent embryonic stem cells (ESCs) can develop into any cell type in the body. Yet, the regulatory mechanisms that govern cell fate decisions during embryogenesis remain largely unknown. We now demonstrate that mouse ESCs (mESCs) display large natural variations in mitochondrial reactive oxygen species (mitoROS) levels that individualize their nuclear redox state, H3K4me3 landscape, and cell fate. While mESCs with high mitoROS levels (mitoROSHIGH) differentiate toward mesendoderm and form the primitive streak during gastrulation, mESCs, which generate less ROS, choose the alternative neuroectodermal fate. Temporal studies demonstrated that mesendodermal (ME) specification of mitoROSHIGH mESCs is mediated by a Nrf2-controlled switch in the nuclear redox state, triggered by the accumulation of redox-sensitive H3K4me3 marks, and executed by a hitherto unknown ROS-dependent activation process of the Wnt signaling pathway. In summary, our study explains how ESC heterogeneity is generated and used by individual cells to decide between distinct cellular fates.


Subject(s)
Cell Differentiation , Mitochondria , Mouse Embryonic Stem Cells , Oxidation-Reduction , Reactive Oxygen Species , Wnt Signaling Pathway , Animals , Mice , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/cytology , Cell Differentiation/physiology , Reactive Oxygen Species/metabolism , Mitochondria/metabolism , NF-E2-Related Factor 2/metabolism , Histones/metabolism , Cell Lineage , Mesoderm/cytology , Mesoderm/metabolism
3.
Development ; 151(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38975838

ABSTRACT

Cohesin, a chromatin-associated protein complex with four core subunits (Smc1a, Smc3, Rad21 and either Stag1 or 2), has a central role in cell proliferation and gene expression in metazoans. Human developmental disorders termed 'cohesinopathies' are characterized by germline variants of cohesin or its regulators that do not entirely eliminate cohesin function. However, it is not clear whether mutations in individual cohesin subunits have independent developmental consequences. Here, we show that zebrafish rad21 or stag2b mutants independently influence embryonic tailbud development. Both mutants have altered mesoderm induction, but only homozygous or heterozygous rad21 mutation affects cell cycle gene expression. stag2b mutants have narrower notochords and reduced Wnt signaling in neuromesodermal progenitors as revealed by single-cell RNA sequencing. Stimulation of Wnt signaling rescues transcription and morphology in stag2b, but not rad21, mutants. Our results suggest that mutations altering the quantity versus composition of cohesin have independent developmental consequences, with implications for the understanding and management of cohesinopathies.


Subject(s)
Cell Cycle Proteins , Chromosomal Proteins, Non-Histone , Cohesins , Mutation , Zebrafish Proteins , Zebrafish , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Mutation/genetics , Gene Expression Regulation, Developmental , Wnt Signaling Pathway/genetics , Embryonic Development/genetics , Gene Dosage , Mesoderm/metabolism , Mesoderm/embryology
4.
Sci Adv ; 10(29): eadl6366, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39028807

ABSTRACT

Physical processes ultimately shape tissue during development. Two emerging proposals are that cells migrate toward stiffer tissue (durotaxis) and that the extent of cell rearrangements reflects tissue phase, but it is unclear whether and how these concepts are related. Here, we identify fibronectin-dependent tissue stiffness as a control variable that underlies and unifies these phenomena in vivo. In murine limb bud mesoderm, cells are either caged, move directionally, or intercalate as a function of their location along a stiffness gradient. A modified Landau phase equation that incorporates tissue stiffness accurately predicts cell diffusivity upon loss or gain of fibronectin. Fibronectin is regulated by WNT5A-YAP feedback that controls cell movements, tissue shape, and skeletal pattern. The results identify a key determinant of phase transition and show how fibronectin-dependent directional cell movement emerges in a mixed-phase environment in vivo.


Subject(s)
Cell Movement , Fibronectins , Mesoderm , Fibronectins/metabolism , Animals , Mesoderm/metabolism , Mesoderm/cytology , Mice , Wnt-5a Protein/metabolism
5.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-39000154

ABSTRACT

Putatively, tooth agenesis was attributed to the initiation failure of tooth germs, though little is known about the histological and molecular alterations. To address if constitutively active FGF signaling is associated with tooth agenesis, we activated Fgf8 in dental mesenchyme with Osr-cre knock-in allele in mice (Osr2-creKI; Rosa26R-Fgf8) and found incisor agenesis and molar microdontia. The cell survival assay showed tremendous apoptosis in both the Osr2-creKI; Rosa26R-Fgf8 incisor epithelium and mesenchyme, which initiated incisor regression from cap stage. In situ hybridization displayed vanished Shh transcription, and immunostaining exhibited reduced Runx2 expression and enlarged mesenchymal Lef1 domain in Osr2-creKI; Rosa26R-Fgf8 incisors, both of which were suggested to enhance apoptosis. In contrast, Osr2-creKI; Rosa26R-Fgf8 molar germs displayed mildly suppressed Shh transcription, and the increased expression of Ectodin, Runx2 and Lef1. Although mildly smaller than WT controls prenatally, the Osr2-creKI; Rosa26R-Fgf8 molar germs produced a miniature tooth with impaired mineralization after a 6-week sub-renal culture. Intriguingly, the implanted Osr2-creKI; Rosa26R-Fgf8 molar germs exhibited delayed odontoblast differentiation and accelerated ameloblast maturation. Collectively, the ectopically activated Fgf8 in dental mesenchyme caused incisor agenesis by triggering incisor regression and postnatal molar microdontia. Our findings reported tooth agenesis resulting from the regression from the early bell stage and implicated a correlation between tooth agenesis and microdontia.


Subject(s)
Fibroblast Growth Factor 8 , Incisor , Mesoderm , Molar , Animals , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/metabolism , Mice , Incisor/abnormalities , Incisor/metabolism , Mesoderm/metabolism , Mesoderm/pathology , Molar/abnormalities , Molar/metabolism , Anodontia/genetics , Anodontia/metabolism , Anodontia/pathology , Apoptosis , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Lymphoid Enhancer-Binding Factor 1/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Signal Transduction , Gene Expression Regulation, Developmental , Odontogenesis/genetics , Mice, Transgenic
6.
Genes Dev ; 38(9-10): 393-414, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38834239

ABSTRACT

The fibroblast growth factor (FGF) pathway is a conserved signaling pathway required for embryonic development. Activated FGF receptor 1 (FGFR1) drives multiple intracellular signaling cascade pathways, including ERK/MAPK and PI3K/AKT, collectively termed canonical signaling. However, unlike Fgfr1-null embryos, embryos containing hypomorphic mutations in Fgfr1 lacking the ability to activate canonical downstream signals are still able to develop to birth but exhibit severe defects in all mesodermal-derived tissues. The introduction of an additional signaling mutation further reduces the activity of Fgfr1, leading to earlier lethality, reduced somitogenesis, and more severe changes in transcriptional outputs. Genes involved in migration, ECM interaction, and phosphoinositol signaling were significantly downregulated, proteomic analysis identified changes in interactions with endocytic pathway components, and cells expressing mutant receptors show changes in endocytic trafficking. Together, we identified processes regulating early mesoderm development by mechanisms involving both canonical and noncanonical Fgfr1 pathways, including direct interaction with cell adhesion components and endocytic regulation.


Subject(s)
Endocytosis , Gene Expression Regulation, Developmental , Mesoderm , Receptor, Fibroblast Growth Factor, Type 1 , Signal Transduction , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Animals , Mesoderm/embryology , Mesoderm/metabolism , Signal Transduction/genetics , Endocytosis/genetics , Gene Expression Regulation, Developmental/genetics , Mice , Embryonic Development/genetics , Protein Transport , Mutation
7.
Int J Mol Sci ; 25(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38891790

ABSTRACT

Derived from axial structures, Sonic Hedgehog (Shh) is secreted into the paraxial mesoderm, where it plays crucial roles in sclerotome induction and myotome differentiation. Through conditional loss-of-function in quail embryos, we investigate the timing and impact of Shh activity during early formation of sclerotome-derived vertebrae and ribs, and of lateral mesoderm-derived sternum. To this end, Hedgehog interacting protein (Hhip) was electroporated at various times between days 2 and 5. While the vertebral body and rib primordium showed consistent size reduction, rib expansion into the somatopleura remained unaffected, and the sternal bud developed normally. Additionally, we compared these effects with those of locally inhibiting BMP activity. Transfection of Noggin in the lateral mesoderm hindered sternal bud formation. Unlike Hhip, BMP inhibition via Noggin or Smad6 induced myogenic differentiation of the lateral dermomyotome lip, while impeding the growth of the myotome/rib complex into the somatic mesoderm, thus affirming the role of the lateral dermomyotome epithelium in rib guidance. Overall, these findings underscore the continuous requirement for opposing gradients of Shh and BMP activity in the morphogenesis of proximal and distal flank skeletal structures, respectively. Future research should address the implications of these early interactions to the later morphogenesis and function of the musculo-skeletal system and of possible associated malformations.


Subject(s)
Hedgehog Proteins , Ribs , Spine , Animals , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Ribs/metabolism , Ribs/embryology , Spine/metabolism , Spine/embryology , Gene Expression Regulation, Developmental , Mesoderm/metabolism , Mesoderm/embryology , Quail , Somites/metabolism , Somites/embryology , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/genetics , Carrier Proteins
8.
Nat Commun ; 15(1): 5233, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898031

ABSTRACT

Mutations in the FOXF1 gene, a key transcriptional regulator of pulmonary vascular development, cause Alveolar Capillary Dysplasia with Misalignment of Pulmonary Veins, a lethal lung disease affecting newborns and infants. Identification of new FOXF1 upstream regulatory elements is critical to explain why frequent non-coding FOXF1 deletions are linked to the disease. Herein, we use multiome single-nuclei RNA and ATAC sequencing of mouse and human patient lungs to identify four conserved endothelial and mesenchymal FOXF1 enhancers. We demonstrate that endothelial FOXF1 enhancers are autoactivated, whereas mesenchymal FOXF1 enhancers are regulated by EBF1 and GLI1. The cell-specificity of FOXF1 enhancers is validated by disrupting these enhancers in mouse embryonic stem cells using CRISPR/Cpf1 genome editing followed by lineage-tracing of mutant embryonic stem cells in mouse embryos using blastocyst complementation. This study resolves an important clinical question why frequent non-coding FOXF1 deletions that interfere with endothelial and mesenchymal enhancers can lead to the disease.


Subject(s)
Enhancer Elements, Genetic , Forkhead Transcription Factors , Mesoderm , Persistent Fetal Circulation Syndrome , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Animals , Humans , Persistent Fetal Circulation Syndrome/genetics , Persistent Fetal Circulation Syndrome/pathology , Persistent Fetal Circulation Syndrome/metabolism , Mice , Enhancer Elements, Genetic/genetics , Mesoderm/metabolism , Mesoderm/embryology , Lung/pathology , Endothelial Cells/metabolism , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism , Embryonic Stem Cells/metabolism , Pulmonary Alveoli/abnormalities
9.
Dev Growth Differ ; 66(5): 320-328, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38925637

ABSTRACT

During the formation of the neural tube, the primordium of the vertebrate central nervous system, the actomyosin activity of cells in different regions drives neural plate bending. However, how the stiffness of the neural plate and surrounding tissues is regulated and mechanically influences neural plate bending has not been elucidated. Here, we used atomic force microscopy to reveal the relationship between the stiffness of the neural plate and the mesoderm during Xenopus neural tube formation. Measurements with intact embryos revealed that the stiffness of the neural plate was consistently higher compared with the non-neural ectoderm and that it increased in an actomyosin activity-dependent manner during neural plate bending. Interestingly, measurements of isolated tissue explants also revealed that the relationship between the stiffness of the apical and basal sides of the neural plate was reversed during bending and that the stiffness of the mesoderm was lower than that of the basal side of the neural plate. The experimental elevation of mesoderm stiffness delayed neural plate bending, suggesting that low mesoderm stiffness mechanically supports neural tube closure. This study provides an example of mechanical interactions between tissues during large-scale morphogenetic movements.


Subject(s)
Neural Plate , Neural Tube , Xenopus laevis , Animals , Neural Tube/embryology , Neural Tube/cytology , Neural Tube/metabolism , Neural Plate/embryology , Neural Plate/metabolism , Neural Plate/cytology , Xenopus laevis/embryology , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/metabolism , Ectoderm/cytology , Ectoderm/metabolism , Microscopy, Atomic Force , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/embryology
10.
Cell ; 187(12): 2898-2900, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38848672

ABSTRACT

Epithelial folding is a fundamental biological process that requires epithelial interactions with the underlying mesenchyme. In this issue of Cell, Huycke et al. investigate intestinal villus formation. They discover that water-droplet-like behavior of mesenchymal cells drives their coalescence into uniformly patterned aggregates, which generate forces on the epithelium to initiate folding.


Subject(s)
Epithelium , Mesoderm , Animals , Humans , Epithelial Cells/metabolism , Epithelial Cells/cytology , Intestinal Mucosa/metabolism , Intestinal Mucosa/cytology , Mesoderm/metabolism , Mesoderm/cytology , Epithelium/metabolism
11.
Curr Top Dev Biol ; 160: 1-30, 2024.
Article in English | MEDLINE | ID: mdl-38937029

ABSTRACT

The salivary gland undergoes branching morphogenesis to elaborate into a tree-like structure with numerous saliva-secreting acinar units, all joined by a hierarchical ductal system. The expansive epithelial surface generated by branching morphogenesis serves as the structural basis for the efficient production and delivery of saliva. Here, we elucidate the process of salivary gland morphogenesis, emphasizing the role of mechanics. Structurally, the developing salivary gland is characterized by a stratified epithelium tightly encased by the basement membrane, which is in turn surrounded by a mesenchyme consisting of a dense network of interstitial matrix and mesenchymal cells. Diverse cell types and extracellular matrices bestow this developing organ with organized, yet spatially varied mechanical properties. For instance, the surface epithelial sheet of the bud is highly fluidic due to its high cell motility and weak cell-cell adhesion, rendering it highly pliable. In contrast, the inner core of the bud is more rigid, characterized by reduced cell motility and strong cell-cell adhesion, which likely provide structural support for the tissue. The interactions between the surface epithelial sheet and the inner core give rise to budding morphogenesis. Furthermore, the basement membrane and the mesenchyme offer mechanical constraints that could play a pivotal role in determining the higher-order architecture of a fully mature salivary gland.


Subject(s)
Morphogenesis , Salivary Glands , Salivary Glands/embryology , Salivary Glands/cytology , Salivary Glands/metabolism , Animals , Humans , Basement Membrane/metabolism , Cell Movement , Biomechanical Phenomena , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/metabolism , Cell Adhesion
12.
Elife ; 122024 Jun 10.
Article in English | MEDLINE | ID: mdl-38856718

ABSTRACT

Abnormal lung development can cause congenital pulmonary cysts, the mechanisms of which remain largely unknown. Although the cystic lesions are believed to result directly from disrupted airway epithelial cell growth, the extent to which developmental defects in lung mesenchymal cells contribute to abnormal airway epithelial cell growth and subsequent cystic lesions has not been thoroughly examined. In the present study using genetic mouse models, we dissected the roles of bone morphogenetic protein (BMP) receptor 1a (Bmpr1a)-mediated BMP signaling in lung mesenchyme during prenatal lung development and discovered that abrogation of mesenchymal Bmpr1a disrupted normal lung branching morphogenesis, leading to the formation of prenatal pulmonary cystic lesions. Severe deficiency of airway smooth muscle cells and subepithelial elastin fibers were found in the cystic airways of the mesenchymal Bmpr1a knockout lungs. In addition, ectopic mesenchymal expression of BMP ligands and airway epithelial perturbation of the Sox2-Sox9 proximal-distal axis were detected in the mesenchymal Bmpr1a knockout lungs. However, deletion of Smad1/5, two major BMP signaling downstream effectors, from the lung mesenchyme did not phenocopy the cystic abnormalities observed in the mesenchymal Bmpr1a knockout lungs, suggesting that a Smad-independent mechanism contributes to prenatal pulmonary cystic lesions. These findings reveal for the first time the role of mesenchymal BMP signaling in lung development and a potential pathogenic mechanism underlying congenital pulmonary cysts.


Congenital disorders are medical conditions that are present from birth. Although many congenital disorders are rare, they can have a severe impact on the quality of life of those affected. For example, congenital pulmonary airway malformation (CPAM) is a rare congenital disorder that occurs in around 1 out of every 25,000 pregnancies. In CPAM, abnormal, fluid-filled sac-like pockets of tissue, known as cysts, form within the lungs of unborn babies. After birth, these cysts become air-filled and do not behave like normal lung tissue and stop a baby's lungs from working properly. In severe cases, babies with CPAM need surgery immediately after birth. We still do not understand exactly what the underlying causes of CPAM might be. CPAM is not considered to be hereditary ­ that is, it does not appear to be passed down in families ­ nor is it obviously linked to any environmental factors. CPAM is also very difficult to study, because researchers cannot access tissue samples during the critical early stages of the disease. To overcome these difficulties, Luo et al. wanted to find a way to study CPAM in the laboratory. First, they developed a non-human animal 'model' that naturally forms CPAM-like lung cysts, using genetically modified mice where the gene for the signaling molecule Bmpr1a had been deleted in lung cells. Normally, Bmpr1a is part of a set of the molecular instructions, collectively termed BMP signaling, which guide healthy lung development early in life. However, mouse embryos lacking Bmpr1a developed abnormal lung cysts that were similar to those found in CPAM patients, suggesting that problems with BMP signalling might also trigger CPAM in humans. Luo et al. also identified several other genes in the Bmpr1a-deficient mouse lungs that had abnormal patterns of activity. All these genes were known to be controlled by BMP signaling, and to play a role in the development and organisation of lung tissue. This suggests that when these genes are not controlled properly, they could drive formation of CPAM cysts when BMP signaling is compromised. This work is a significant advance in the tools available to study CPAM. Luo et al.'s results also shed new light on the molecular mechanisms underpinning this rare disorder. In the future, Luo et al. hope this knowledge will help us develop better treatments for CPAM, or even help to prevent it altogether.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I , Lung , Mesoderm , Mice, Knockout , Signal Transduction , Animals , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Protein Receptors, Type I/deficiency , Mice , Lung/embryology , Lung/metabolism , Lung/pathology , Mesoderm/embryology , Mesoderm/metabolism , Cysts/metabolism , Cysts/pathology , Cysts/genetics , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/genetics , Lung Diseases/metabolism , Lung Diseases/pathology , Lung Diseases/genetics , Disease Models, Animal
13.
Nat Commun ; 15(1): 5210, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890321

ABSTRACT

Cell-fate decisions during mammalian gastrulation are poorly understood outside of rodent embryos. The embryonic disc of pig embryos mirrors humans, making them a useful proxy for studying gastrulation. Here we present a single-cell transcriptomic atlas of pig gastrulation, revealing cell-fate emergence dynamics, as well as conserved and divergent gene programs governing early porcine, primate, and murine development. We highlight heterochronicity in extraembryonic cell-types, despite the broad conservation of cell-type-specific transcriptional programs. We apply these findings in combination with functional investigations, to outline conserved spatial, molecular, and temporal events during definitive endoderm specification. We find early FOXA2 + /TBXT- embryonic disc cells directly form definitive endoderm, contrasting later-emerging FOXA2/TBXT+ node/notochord progenitors. Unlike mesoderm, none of these progenitors undergo epithelial-to-mesenchymal transition. Endoderm/Node fate hinges on balanced WNT and hypoblast-derived NODAL, which is extinguished upon endodermal differentiation. These findings emphasise the interplay between temporal and topological signalling in fate determination during gastrulation.


Subject(s)
Embryo, Mammalian , Endoderm , Gastrulation , Gene Expression Regulation, Developmental , Single-Cell Analysis , Animals , Endoderm/cytology , Endoderm/metabolism , Endoderm/embryology , Swine , Mice , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Cell Differentiation , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/metabolism , Transcriptome , Hepatocyte Nuclear Factor 3-beta/metabolism , Hepatocyte Nuclear Factor 3-beta/genetics , Cell Lineage , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/genetics , Epithelial-Mesenchymal Transition/genetics
14.
Development ; 151(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38828908

ABSTRACT

During limb bud formation, axis polarities are established as evidenced by the spatially restricted expression of key regulator genes. In particular, the mutually antagonistic interaction between the GLI3 repressor and HAND2 results in distinct and non-overlapping anterior-distal Gli3 and posterior Hand2 expression domains. This is a hallmark of the establishment of antero-posterior limb axis polarity, together with spatially restricted expression of homeodomain and other transcriptional regulators. Here, we show that TBX3 is required for establishment of the posterior expression boundary of anterior genes in mouse limb buds. ChIP-seq and differential gene expression analysis of wild-type and mutant limb buds identifies TBX3-specific and shared TBX3-HAND2 target genes. High sensitivity fluorescent whole-mount in situ hybridisation shows that the posterior expression boundaries of anterior genes are positioned by TBX3-mediated repression, which excludes anterior genes such as Gli3, Alx4, Hand1 and Irx3/5 from the posterior limb bud mesenchyme. This exclusion delineates the posterior mesenchymal territory competent to establish the Shh-expressing limb bud organiser. In turn, HAND2 is required for Shh activation and cooperates with TBX3 to upregulate shared posterior identity target genes in early limb buds.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Gene Expression Regulation, Developmental , Limb Buds , T-Box Domain Proteins , Animals , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/genetics , Limb Buds/metabolism , Limb Buds/embryology , Mice , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Zinc Finger Protein Gli3/metabolism , Zinc Finger Protein Gli3/genetics , Up-Regulation/genetics , Body Patterning/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Mesoderm/metabolism , Mesoderm/embryology
15.
Cell Syst ; 15(5): 445-461.e4, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38692274

ABSTRACT

BMP signaling is essential for mammalian gastrulation, as it initiates a cascade of signals that control self-organized patterning. As development is highly dynamic, it is crucial to understand how time-dependent combinatorial signaling affects cellular differentiation. Here, we show that BMP signaling duration is a crucial control parameter that determines cell fates upon the exit from pluripotency through its interplay with the induced secondary signal WNT. BMP signaling directly converts cells from pluripotent to extraembryonic fates while simultaneously upregulating Wnt signaling, which promotes primitive streak and mesodermal specification. Using live-cell imaging of signaling and cell fate reporters together with a simple mathematical model, we show that this circuit produces a temporal morphogen effect where, once BMP signal duration is above a threshold for differentiation, intermediate and long pulses of BMP signaling produce specification of mesoderm and extraembryonic fates, respectively. Our results provide a systems-level picture of how these signaling pathways control the landscape of early human development.


Subject(s)
Bone Morphogenetic Proteins , Cell Differentiation , Primitive Streak , Signal Transduction , Primitive Streak/metabolism , Primitive Streak/embryology , Bone Morphogenetic Proteins/metabolism , Humans , Signal Transduction/physiology , Animals , Mesoderm/metabolism , Mesoderm/embryology , Wnt Signaling Pathway/physiology , Wnt Proteins/metabolism , Gene Expression Regulation, Developmental , Gastrulation/physiology
16.
Curr Top Dev Biol ; 159: 372-405, 2024.
Article in English | MEDLINE | ID: mdl-38729682

ABSTRACT

The Segmentation Clock is a tissue-level patterning system that enables the segmentation of the vertebral column precursors into transient multicellular blocks called somites. This patterning system comprises a set of elements that are essential for correct segmentation. Under the so-called "Clock and Wavefront" model, the system consists of two elements, a genetic oscillator that manifests itself as traveling waves of gene expression, and a regressing wavefront that transforms the temporally periodic signal encoded in the oscillations into a permanent spatially periodic pattern of somite boundaries. Over the last twenty years, every new discovery about the Segmentation Clock has been tightly linked to the nomenclature of the "Clock and Wavefront" model. This constrained allocation of discoveries into these two elements has generated long-standing debates in the field as what defines molecularly the wavefront and how and where the interaction between the two elements establishes the future somite boundaries. In this review, we propose an expansion of the "Clock and Wavefront" model into three elements, "Clock", "Wavefront" and signaling gradients. We first provide a detailed description of the components and regulatory mechanisms of each element, and we then examine how the spatiotemporal integration of the three elements leads to the establishment of the presumptive somite boundaries. To be as exhaustive as possible, we focus on the Segmentation Clock in zebrafish. Furthermore, we show how this three-element expansion of the model provides a better understanding of the somite formation process and we emphasize where our current understanding of this patterning system remains obscure.


Subject(s)
Body Patterning , Gene Expression Regulation, Developmental , Mesoderm , Somites , Animals , Body Patterning/genetics , Somites/embryology , Somites/metabolism , Mesoderm/embryology , Mesoderm/metabolism , Mesoderm/cytology , Zebrafish/embryology , Zebrafish/genetics , Signal Transduction , Biological Clocks/genetics
17.
Differentiation ; 138: 100782, 2024.
Article in English | MEDLINE | ID: mdl-38810379

ABSTRACT

The mandible is composed of several musculoskeletal tissues including bone, cartilage, and tendon that require precise patterning to ensure structural and functional integrity. Interestingly, most of these tissues are derived from one multipotent cell population called cranial neural crest cells (CNCCs). How CNCCs are properly instructed to differentiate into various tissue types remains nebulous. To better understand the mechanisms necessary for the patterning of mandibular musculoskeletal tissues we utilized the avian mutant talpid2 (ta2) which presents with several malformations of the facial skeleton including dysplastic tendons, mispatterned musculature, and bilateral ectopic cartilaginous processes extending off Meckel's cartilage. We found an ectopic epithelial BMP signaling domain in the ta2 mandibular prominence (MNP) that correlated with the subsequent expansion of SOX9+ cartilage precursors. These findings were validated with conditional murine models suggesting an evolutionarily conserved mechanism for CNCC-derived musculoskeletal patterning. Collectively, these data support a model in which cilia are required to define epithelial signal centers essential for proper musculoskeletal patterning of CNCC-derived mesenchyme.


Subject(s)
Mandible , Neural Crest , Animals , Chick Embryo , Mice , Avian Proteins/genetics , Avian Proteins/metabolism , Body Patterning/genetics , Cartilage/metabolism , Cartilage/growth & development , Cartilage/cytology , Cell Differentiation , Chickens/genetics , Cilia/metabolism , Cilia/genetics , Gene Expression Regulation, Developmental , Mandible/growth & development , Mandible/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mesoderm/growth & development , Neural Crest/cytology , Neural Crest/metabolism , Signal Transduction , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics
18.
Cell ; 187(12): 3072-3089.e20, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38781967

ABSTRACT

Tissue folds are structural motifs critical to organ function. In the intestine, bending of a flat epithelium into a periodic pattern of folds gives rise to villi, finger-like protrusions that enable nutrient absorption. However, the molecular and mechanical processes driving villus morphogenesis remain unclear. Here, we identify an active mechanical mechanism that simultaneously patterns and folds the intestinal epithelium to initiate villus formation. At the cellular level, we find that PDGFRA+ subepithelial mesenchymal cells generate myosin II-dependent forces sufficient to produce patterned curvature in neighboring tissue interfaces. This symmetry-breaking process requires altered cell and extracellular matrix interactions that are enabled by matrix metalloproteinase-mediated tissue fluidization. Computational models, together with in vitro and in vivo experiments, revealed that these cellular features manifest at the tissue level as differences in interfacial tensions that promote mesenchymal aggregation and interface bending through a process analogous to the active dewetting of a thin liquid film.


Subject(s)
Extracellular Matrix , Intestinal Mucosa , Animals , Mice , Intestinal Mucosa/metabolism , Intestinal Mucosa/cytology , Extracellular Matrix/metabolism , Myosin Type II/metabolism , Mesoderm/metabolism , Mesoderm/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Morphogenesis , Matrix Metalloproteinases/metabolism
19.
Development ; 151(12)2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38814743

ABSTRACT

Apical expansion of calvarial osteoblast progenitors from the cranial mesenchyme (CM) above the eye is integral to calvarial growth and enclosure of the brain. The cellular behaviors and signals underlying the morphogenetic process of calvarial expansion are unknown. Time-lapse light-sheet imaging of mouse embryos revealed calvarial progenitors intercalate in 3D in the CM above the eye, and exhibit protrusive and crawling activity more apically. CM cells express non-canonical Wnt/planar cell polarity (PCP) core components and calvarial osteoblasts are bidirectionally polarized. We found non-canonical ligand Wnt5a-/- mutants have less dynamic cell rearrangements and protrusive activity. Loss of CM-restricted Wntless (CM-Wls), a gene required for secretion of all Wnt ligands, led to diminished apical expansion of Osx+ calvarial osteoblasts in the frontal bone primordia in a non-cell autonomous manner without perturbing proliferation or survival. Calvarial osteoblast polarization, progressive cell elongation and enrichment for actin along the baso-apical axis were dependent on CM-Wnts. Thus, CM-Wnts regulate cellular behaviors during calvarial morphogenesis for efficient apical expansion of calvarial osteoblasts. These findings also offer potential insights into the etiologies of calvarial dysplasias.


Subject(s)
Mesoderm , Morphogenesis , Osteoblasts , Skull , Wnt Proteins , Animals , Osteoblasts/metabolism , Osteoblasts/cytology , Skull/embryology , Mice , Mesoderm/cytology , Mesoderm/metabolism , Wnt Proteins/metabolism , Wnt Proteins/genetics , Cell Polarity , Wnt-5a Protein/metabolism , Wnt-5a Protein/genetics , Cell Movement , Cell Proliferation
20.
EMBO J ; 43(12): 2308-2336, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38760574

ABSTRACT

How cells coordinate morphogenetic cues and fate specification during development remains a fundamental question in organogenesis. The mammary gland arises from multipotent stem cells (MaSCs), which are progressively replaced by unipotent progenitors by birth. However, the lack of specific markers for early fate specification has prevented the delineation of the features and spatial localization of MaSC-derived lineage-committed progenitors. Here, using single-cell RNA sequencing from E13.5 to birth, we produced an atlas of matched mouse mammary epithelium and mesenchyme and reconstructed the differentiation trajectories of MaSCs toward basal and luminal fate. We show that murine MaSCs exhibit lineage commitment just prior to the first sprouting events of mammary branching morphogenesis at E15.5. We identify early molecular markers for committed and multipotent MaSCs and define their spatial distribution within the developing tissue. Furthermore, we show that the mammary embryonic mesenchyme is composed of two spatially restricted cell populations, and that dermal mesenchyme-produced FGF10 is essential for embryonic mammary branching morphogenesis. Altogether, our data elucidate the spatiotemporal signals underlying lineage specification of multipotent MaSCs, and uncover the signals from mesenchymal cells that guide mammary branching morphogenesis.


Subject(s)
Cell Lineage , Epithelial Cells , Mammary Glands, Animal , Mesenchymal Stem Cells , Animals , Mice , Mammary Glands, Animal/cytology , Mammary Glands, Animal/embryology , Mammary Glands, Animal/metabolism , Female , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Cell Differentiation , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Fibroblast Growth Factor 10/metabolism , Fibroblast Growth Factor 10/genetics , Morphogenesis , Single-Cell Analysis , Mesoderm/cytology , Mesoderm/metabolism , Mesoderm/embryology
SELECTION OF CITATIONS
SEARCH DETAIL