Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.376
Filter
1.
Nat Commun ; 15(1): 8153, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300061

ABSTRACT

Limb reduction has occurred multiple times in tetrapod history. Among ratites, wing reductions range from mild vestigialization to complete loss, with emus (Dromaius novaehollandiae) serving as a model for studying the genetic mechanisms behind limb reduction. Here, we explore the developmental mechanisms underlying wing reduction in emu. Our analyses reveal that immobilization resulting from the absence of distal muscles contributes to skeletal shortening, fusion and left-right intraindividual variation. Expression analysis and single cell-RNA sequencing identify muscle progenitors displaying a dual lateral plate mesodermal and myogenic signature. These cells aggregate at the proximal region of wing buds and undergo cell death. We propose that this cell death, linked to the lack of distal muscle masses, underlines the morphological features and variability in skeletal elements due to reduced mechanical loading. Our results demonstrate that differential mobility during embryonic development may drive morphological diversification in vestigial structures.


Subject(s)
Cell Death , Dromaiidae , Gene Expression Regulation, Developmental , Wings, Animal , Animals , Wings, Animal/metabolism , Dromaiidae/genetics , Cell Death/genetics , Mesoderm/metabolism , Muscle, Skeletal/metabolism , Body Patterning/genetics , Myoblasts/metabolism , Myoblasts/cytology
2.
Development ; 151(16)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39225402

ABSTRACT

Adult mammalian lungs exhibit a fractal pattern, as each successive generation of airways is a fraction of the size of the parental branch. Achieving this structure likely requires precise control of airway length and diameter, as the embryonic airways initially lack the fractal scaling observed in the adult. In monolayers and tubes, directional growth can be regulated by the planar cell polarity (PCP) complex. Here, we characterized the roles of PCP complex components in airway initiation, elongation and widening during branching morphogenesis of the lung. Using tissue-specific knockout mice, we surprisingly found that branching morphogenesis proceeds independently of PCP complex function in the lung epithelium. Instead, we found a previously unreported Celsr1-independent role for the PCP complex components Vangl1 and Vangl2 in the pulmonary mesenchyme, where they are required for branch initiation, elongation and widening. Our data thus reveal an explicit function for Vangl1 and Vangl2 that is independent of the core PCP complex, suggesting a functional diversification of PCP complex components in vertebrate development. These data also reveal an essential role for the embryonic mesenchyme in generating the fractal structure of airways in the mature lung.


Subject(s)
Cell Polarity , Lung , Membrane Proteins , Mesoderm , Nerve Tissue Proteins , Animals , Mice , Lung/embryology , Lung/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mesoderm/metabolism , Mesoderm/embryology , Mice, Knockout , Morphogenesis , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Receptors, G-Protein-Coupled
3.
Development ; 151(18)2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39315665

ABSTRACT

The intricate dynamics of Hes expression across diverse cell types in the developing vertebrate embryonic tail have remained elusive. To address this, we have developed an endogenously tagged Hes1-Achilles mouse line, enabling precise quantification of dynamics at the single-cell resolution across various tissues. Our findings reveal striking disparities in Hes1 dynamics between presomitic mesoderm (PSM) and preneural tube (pre-NT) cells. While pre-NT cells display variable, low-amplitude oscillations, PSM cells exhibit synchronized, high-amplitude oscillations. Upon the induction of differentiation, the oscillation amplitude increases in pre-NT cells. Additionally, our study of Notch inhibition on Hes1 oscillations unveils distinct responses in PSM and pre-NT cells, corresponding to differential Notch ligand expression dynamics. These findings suggest the involvement of separate mechanisms driving Hes1 oscillations. Thus, Hes1 demonstrates dynamic behaviour across adjacent tissues of the embryonic tail, yet the varying oscillation parameters imply differences in the information that can be transmitted by these dynamics.


Subject(s)
Embryo, Mammalian , Gene Expression Regulation, Developmental , Mesoderm , Single-Cell Analysis , Transcription Factor HES-1 , Animals , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics , Mice , Mesoderm/metabolism , Mesoderm/cytology , Mesoderm/embryology , Embryo, Mammalian/metabolism , Receptors, Notch/metabolism , Cell Differentiation , Body Patterning , Somites/metabolism , Somites/embryology , Embryonic Development/genetics , Tail/embryology
4.
Development ; 151(17)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39221968

ABSTRACT

The lymphatic system is formed during embryonic development by the commitment of specialized lymphatic endothelial cells (LECs) and their subsequent assembly in primary lymphatic vessels. Although lymphatic cells are in continuous contact with mesenchymal cells during development and in adult tissues, the role of mesenchymal cells in lymphatic vasculature development remains poorly characterized. Here, we show that a subpopulation of mesenchymal cells expressing the transcription factor Osr1 are in close association with migrating LECs and established lymphatic vessels in mice. Lineage tracing experiments revealed that Osr1+ cells precede LEC arrival during lymphatic vasculature assembly in the back of the embryo. Using Osr1-deficient embryos and functional in vitro assays, we show that Osr1 acts in a non-cell-autonomous manner controlling proliferation and early migration of LECs to peripheral tissues. Thereby, mesenchymal Osr1+ cells control, in a bimodal manner, the production of extracellular matrix scaffold components and signal ligands crucial for lymphatic vessel formation.


Subject(s)
Endothelial Cells , Lymphangiogenesis , Lymphatic Vessels , Transcription Factors , Animals , Lymphatic Vessels/embryology , Lymphatic Vessels/metabolism , Lymphatic Vessels/cytology , Mice , Lymphangiogenesis/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Endothelial Cells/metabolism , Endothelial Cells/cytology , Cell Movement/genetics , Cell Proliferation , Embryo, Mammalian/metabolism , Embryo, Mammalian/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mesoderm/metabolism , Mesoderm/cytology , Gene Expression Regulation, Developmental , Cell Lineage
5.
Stem Cell Res Ther ; 15(1): 273, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39218985

ABSTRACT

BACKGROUND: Chronic lung disease of prematurity, called bronchopulmonary dysplasia (BPD), lacks effective therapies, stressing the need for preclinical testing systems that reflect human pathology for identifying causal pathways and testing novel compounds. Alveolar organoids derived from human pluripotent stem cells (hPSC) are promising test platforms for studying distal airway diseases like BPD, but current protocols do not accurately replicate the distal niche environment of the native lung. Herein, we investigated the contributions of cellular constituents of the alveolus and fetal respiratory movements on hPSC-derived alveolar organoid formation. METHODS: Human PSCs were differentiated in 2D culture into lung progenitor cells (LPC) which were then further differentiated into alveolar organoids before and after removal of co-developing mesodermal cells. LPCs were also differentiated in Transwell® co-cultures with and without human fetal lung fibroblast. Forming organoids were subjected to phasic mechanical strain using a Flexcell® system. Differentiation within organoids and Transwell® cultures was assessed by flow cytometry, immunofluorescence, and qPCR for lung epithelial and alveolar markers of differentiation including GATA binding protein 6 (GATA 6), E-cadherin (CDH1), NK2 Homeobox 1 (NKX2-1), HT2-280, surfactant proteins B (SFTPB) and C (SFTPC). RESULTS: We observed that co-developing mesenchymal progenitors promote alveolar epithelial type 2 cell (AEC2) differentiation within hPSC-derived lung organoids. This mesenchymal effect on AEC2 differentiation was corroborated by co-culturing hPSC-NKX2-1+ lung progenitors with human embryonic lung fibroblasts. The stimulatory effect did not require direct contact between fibroblasts and NKX2-1+ lung progenitors. Additionally, we demonstrate that episodic mechanical deformation of hPSC-derived lung organoids, mimicking in situ fetal respiratory movements, increased AEC2 differentiation without affecting proximal epithelial differentiation. CONCLUSION: Our data suggest that biophysical and mesenchymal components promote AEC2 differentiation within hPSC-derived distal organoids in vitro.


Subject(s)
Cell Differentiation , Lung , Organoids , Humans , Organoids/cytology , Organoids/metabolism , Lung/cytology , Lung/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Coculture Techniques/methods , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism
6.
Commun Biol ; 7(1): 1100, 2024 Sep 07.
Article in English | MEDLINE | ID: mdl-39244636

ABSTRACT

PHD2 is essential in modulating HIF-1α levels upon oxygen fluctuations. Hypoxia, a hallmark of uterus, and HIF-1α have recently emerged as opposing regulators of mesendoderm specification, suggesting a role for PHD2 therein. We found that PHD2 expression initially covered the epiblast and gradually receded from the primitive streak, which was identical to hypoxia and exclusive to HIF-1α. The investigations performed in mESCs, embryoids, and mouse embryos together demonstrated that PHD2 negatively regulated mesendoderm specification. Single-cell RNA sequencing revealed that PHD2 governed the transition from epiblast to mesendoderm. The downstream effect of PHD2 relied on the HIF-1α regulated Wnt/ß-catenin pathway, while it was regulated upstream by miR-429. In summary, our research highlights PHD2's essential role in mesendoderm specification and its interactions with hypoxia and HIF-1α.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Hypoxia-Inducible Factor-Proline Dioxygenases , Animals , Mice , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mesoderm/metabolism , Mesoderm/embryology , Gene Expression Regulation, Developmental , Wnt Signaling Pathway , Endoderm/metabolism , Endoderm/embryology , MicroRNAs/metabolism , MicroRNAs/genetics
7.
Stem Cell Reports ; 19(9): 1304-1319, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39178847

ABSTRACT

Effective lineage-specific differentiation is essential to fulfilling the great potentials of human pluripotent stem cells (hPSCs). In this report, we investigate how modulation of medium pH and associated metabolic changes influence mesendoderm differentiation from hPSCs. We show that daily medium pH fluctuations are critical for the heterogeneity of cell fates in the absence of exogenous inducers. Acidic environment alone leads to cardiomyocyte generation without other signaling modulators. In contrast, medium alkalinization is inhibitory to cardiac fate even in the presence of classic cardiac inducers. We then demonstrate that acidic environment suppresses glycolysis to facilitate cardiac differentiation, while alkaline condition promotes glycolysis and diverts the differentiation toward other cell types. We further show that glycolysis inhibition or AMPK activation can rescue cardiac differentiation under alkalinization, and glycolysis inhibition alone can drive cardiac cell fate. This study highlights that pH changes remodel metabolic patterns and modulate signaling pathways to control cell fate.


Subject(s)
Cell Differentiation , Glycolysis , Myocytes, Cardiac , Pluripotent Stem Cells , Humans , Cell Differentiation/drug effects , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Hydrogen-Ion Concentration , Acidosis/metabolism , Endoderm/cytology , Endoderm/metabolism , Cell Lineage/drug effects , Mesoderm/cytology , Mesoderm/metabolism , Culture Media/pharmacology , Culture Media/chemistry , Signal Transduction/drug effects , Cell Line , AMP-Activated Protein Kinases/metabolism
8.
Nature ; 632(8027): 1101-1109, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39112711

ABSTRACT

The mouse small intestine shows profound variability in gene expression along the crypt-villus axis1,2. Whether similar spatial heterogeneity exists in the adult human gut remains unclear. Here we use spatial transcriptomics, spatial proteomics and single-molecule fluorescence in situ hybridization to reconstruct a comprehensive spatial expression atlas of the adult human proximal small intestine. We describe zonated expression and cell type representation for epithelial, mesenchymal and immune cell types. We find that migrating enterocytes switch from lipid droplet assembly and iron uptake at the villus bottom to chylomicron biosynthesis and iron release at the tip. Villus tip cells are pro-immunogenic, recruiting γδ T cells and macrophages to the tip, in contrast to their immunosuppressive roles in mouse. We also show that the human small intestine contains abundant serrated and branched villi that are enriched at the tops of circular folds. Our study presents a detailed resource for understanding the biology of the adult human small intestine.


Subject(s)
Cell Biology , Gene Expression Profiling , Intestine, Small , Adult , Animals , Female , Humans , Male , Mice , Cell Movement , Chylomicrons/biosynthesis , Enterocytes/metabolism , Enterocytes/cytology , Epithelial Cells , In Situ Hybridization, Fluorescence , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestine, Small/cytology , Intestine, Small/immunology , Intestine, Small/metabolism , Iron/metabolism , Lipid Droplets/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Proteomics , Single Molecule Imaging , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transcriptome
9.
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
10.
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
11.
Cell Stem Cell ; 31(9): 1298-1314.e8, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39146934

ABSTRACT

Endogenous retroviruses (ERVs) occupy a significant part of the human genome, with some encoding proteins that influence the immune system or regulate cell-cell fusion in early extra-embryonic development. However, whether ERV-derived proteins regulate somatic development is unknown. Here, we report a somatic developmental function for the primate-specific ERVH48-1 (SUPYN/Suppressyn). ERVH48-1 encodes a fragment of a viral envelope that is expressed during early embryonic development. Loss of ERVH48-1 led to impaired mesoderm and cardiomyocyte commitment and diverted cells to an ectoderm-like fate. Mechanistically, ERVH48-1 is localized to sub-cellular membrane compartments through a functional N-terminal signal peptide and binds to the WNT antagonist SFRP2 to promote its polyubiquitination and degradation, thus limiting SFRP2 secretion and blocking repression of WNT/ß-catenin signaling. Knockdown of SFRP2 or expression of a chimeric SFRP2 with the ERVH48-1 signal peptide rescued cardiomyocyte differentiation. This study demonstrates how ERVH48-1 modulates WNT/ß-catenin signaling and cell type commitment in somatic development.


Subject(s)
Cell Differentiation , Endogenous Retroviruses , Membrane Proteins , Myocytes, Cardiac , Wnt Signaling Pathway , Humans , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Membrane Proteins/metabolism , Membrane Proteins/genetics , Endogenous Retroviruses/metabolism , Endogenous Retroviruses/genetics , Animals , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Primates , HEK293 Cells , Mesoderm/metabolism
12.
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
13.
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
14.
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
15.
Development ; 151(16)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39082789

ABSTRACT

Skeletal muscles of the head and trunk originate in distinct lineages with divergent regulatory programmes converging on activation of myogenic determination factors. Branchiomeric head and neck muscles share a common origin with cardiac progenitor cells in cardiopharyngeal mesoderm (CPM). The retinoic acid (RA) signalling pathway is required during a defined early time window for normal deployment of cells from posterior CPM to the heart. Here, we show that blocking RA signalling in the early mouse embryo also results in selective loss of the trapezius neck muscle, without affecting other skeletal muscles. RA signalling is required for robust expression of myogenic determination factors in posterior CPM and subsequent expansion of the trapezius primordium. Lineage-specific activation of a dominant-negative RA receptor reveals that trapezius development is not regulated by direct RA signalling to myogenic progenitor cells in CPM, or through neural crest cells, but indirectly through the somitic lineage, closely apposed with posterior CPM in the early embryo. These findings suggest that trapezius development is dependent on precise spatiotemporal interactions between cranial and somitic mesoderm at the head/trunk interface.


Subject(s)
Head , Mesoderm , Muscle Development , Neck Muscles , Signal Transduction , Tretinoin , Animals , Tretinoin/metabolism , Mice , Neck Muscles/embryology , Mesoderm/metabolism , Mesoderm/embryology , Head/embryology , Gene Expression Regulation, Developmental , Somites/metabolism , Somites/embryology , Receptors, Retinoic Acid/metabolism
16.
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
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL