Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 16.553
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Curr Top Dev Biol ; 159: 232-271, 2024.
Article in English | MEDLINE | ID: mdl-38729677

ABSTRACT

The anterior-to-posterior (head-to-tail) body axis is extraordinarily diverse among vertebrates but conserved within species. Body axis development requires a population of axial progenitors that resides at the posterior of the embryo to sustain elongation and is then eliminated once axis extension is complete. These progenitors occupy distinct domains in the posterior (tail-end) of the embryo and contribute to various lineages along the body axis. The subset of axial progenitors with neuromesodermal competency will generate both the neural tube (the precursor of the spinal cord), and the trunk and tail somites (producing the musculoskeleton) during embryo development. These axial progenitors are called Neuromesodermal Competent cells (NMCs) and Neuromesodermal Progenitors (NMPs). NMCs/NMPs have recently attracted interest beyond the field of developmental biology due to their clinical potential. In the mouse, the maintenance of neuromesodermal competency relies on a fine balance between a trio of known signals: Wnt/ß-catenin, FGF signalling activity and suppression of retinoic acid signalling. These signals regulate the relative expression levels of the mesodermal transcription factor Brachyury and the neural transcription factor Sox2, permitting the maintenance of progenitor identity when co-expressed, and either mesoderm or neural lineage commitment when the balance is tilted towards either Brachyury or Sox2, respectively. Despite important advances in understanding key genes and cellular behaviours involved in these fate decisions, how the balance between mesodermal and neural fates is achieved remains largely unknown. In this chapter, we provide an overview of signalling and gene regulatory networks in NMCs/NMPs. We discuss mutant phenotypes associated with axial defects, hinting at the potential significant role of lesser studied proteins in the maintenance and differentiation of the progenitors that fuel axial elongation.


Subject(s)
Body Patterning , Mesoderm , Animals , Body Patterning/genetics , Mesoderm/metabolism , Mesoderm/cytology , Mesoderm/embryology , Gene Expression Regulation, Developmental , Humans , Signal Transduction , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/genetics , Cell Differentiation , Head/embryology
7.
Nat Commun ; 15(1): 4550, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38811547

ABSTRACT

The emergence of new structures can often be linked to the evolution of novel cell types that follows the rewiring of developmental gene regulatory subnetworks. Vertebrates are characterized by a complex body plan compared to the other chordate clades and the question remains of whether and how the emergence of vertebrate morphological innovations can be related to the appearance of new embryonic cell populations. We previously proposed, by studying mesoderm development in the cephalochordate amphioxus, a scenario for the evolution of the vertebrate head mesoderm. To further test this scenario at the cell population level, we used scRNA-seq to construct a cell atlas of the amphioxus neurula, stage at which the main mesodermal compartments are specified. Our data allowed us to validate the presence of a prechordal-plate like territory in amphioxus. Additionally, the transcriptomic profile of somite cell populations supports the homology between specific territories of amphioxus somites and vertebrate cranial/pharyngeal and lateral plate mesoderm. Finally, our work provides evidence that the appearance of the specific mesodermal structures of the vertebrate head was associated to both segregation of pre-existing cell populations, and co-option of new genes for the control of myogenesis.


Subject(s)
Gene Expression Regulation, Developmental , Head , Lancelets , Mesoderm , Vertebrates , Animals , Mesoderm/cytology , Mesoderm/embryology , Lancelets/embryology , Lancelets/genetics , Head/embryology , Vertebrates/embryology , Vertebrates/genetics , Somites/embryology , Somites/cytology , Somites/metabolism , Biological Evolution , Transcriptome
8.
Development ; 151(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38757779

ABSTRACT

Collective migration of caudal visceral mesoderm (CVM) cells in Drosophila embryos helps form the longitudinal muscles of the larval gut. In their study, Angelike Stathopoulos and colleagues reveal that cell division coordinates two gene expression programmes in migrating CVM cells. To know more about their work, we spoke to the first author, Jingjing Sun, and the corresponding author, Angelike Stathopoulos, Professor in the Division of Biology at the California Institute of Technology, USA.


Subject(s)
Developmental Biology , Animals , Developmental Biology/history , History, 20th Century , History, 21st Century , Mesoderm/metabolism , Drosophila/embryology , Cell Movement , Humans
9.
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
10.
Development ; 151(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38742434

ABSTRACT

During mouse development, presomitic mesoderm cells synchronize Wnt and Notch oscillations, creating sequential phase waves that pattern somites. Traditional somitogenesis models attribute phase waves to a global modulation of the oscillation frequency. However, increasing evidence suggests that they could arise in a self-organizing manner. Here, we introduce the Sevilletor, a novel reaction-diffusion system that serves as a framework to compare different somitogenesis patterning hypotheses. Using this framework, we propose the Clock and Wavefront Self-Organizing model that considers an excitable self-organizing region where phase waves form independent of global frequency gradients. The model recapitulates the change in relative phase of Wnt and Notch observed during mouse somitogenesis and provides a theoretical basis for understanding the excitability of mouse presomitic mesoderm cells in vitro.


Subject(s)
Receptors, Notch , Somites , Animals , Mice , Somites/embryology , Somites/metabolism , Receptors, Notch/metabolism , Receptors, Notch/genetics , Mesoderm/embryology , Mesoderm/metabolism , Models, Biological , Body Patterning/genetics , Wnt Proteins/metabolism , Wnt Proteins/genetics , Embryonic Development/genetics , Embryonic Development/physiology , Biological Clocks/physiology
11.
Article in Chinese | MEDLINE | ID: mdl-38563180

ABSTRACT

Cartilage mesenchyme hamartoma originates from the mesoderm and contains a blend of interstitium and cartilage, which is mostly benign tumor and is a non-neoplastic cartilage lesion with self-limiting hyperplasia. This article reports a infant with cervical chondromesenchymal hamartoma in the neck, the main clinical manifestations of which are asphyxia and acute respiratory distress, and the imaging features are often similar to those of malignant tumors.Radical resection operation under general anesthesia is the main treatment method, and the postoperative pathological diagnosis was cartilage mesenchyme, and immunohistochemistry showed Catenin(-),MDM2(+),CDK4(-),H3K36M(+),Myogenin (-),SMA (-).The clinical characteristics and diagnosis and treatment process of this case are reported and related literature is reviewed.


Subject(s)
Cartilage , Hamartoma , Humans , Infant, Newborn , Immunohistochemistry , Mesoderm/pathology
12.
PLoS One ; 19(4): e0297853, 2024.
Article in English | MEDLINE | ID: mdl-38635504

ABSTRACT

During vertebrate embryo development, the body is progressively segmented along the anterior-posterior (A-P) axis early in development. The rate of somite formation is controlled by the somitogenesis embryo clock (EC), which was first described as gene expression oscillations of hairy1 (hes4) in the presomitic mesoderm of chick embryos with 15-20 somites. Here, the EC displays the same periodicity as somite formation, 90 min, whereas the posterior-most somites (44-52) only arise every 150 minutes, matched by a corresponding slower pace of the EC. Evidence suggests that the rostral-most somites are formed faster, however, their periodicity and the EC expression dynamics in these early stages are unknown. In this study, we used time-lapse imaging of chicken embryos from primitive streak to somitogenesis stages with high temporal resolution (3-minute intervals). We measured the length between the anterior-most and the last formed somitic clefts in each captured frame and developed a simple algorithm to automatically infer both the length and time of formation of each somite. We found that the occipital somites (up to somite 5) form at an average rate of 75 minutes, while somites 6 onwards are formed approximately every 90 minutes. We also assessed the expression dynamics of hairy1 using half-embryo explants cultured for different periods of time. This showed that EC hairy1 expression is highly dynamic prior to somitogenesis and assumes a clear oscillatory behaviour as the first somites are formed. Importantly, using ex ovo culture and live-imaging techniques, we showed that the hairy1 expression pattern recapitulates with the formation of each new pair of somites, indicating that somite segmentation is coupled with EC oscillations since the onset of somitogenesis.


Subject(s)
Avian Proteins , Somites , Animals , Chick Embryo , Chickens , Basic Helix-Loop-Helix Transcription Factors/genetics , Avian Proteins/genetics , Mesoderm/metabolism
13.
Int J Oral Sci ; 16(1): 33, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38654018

ABSTRACT

Precise orchestration of cell fate determination underlies the success of scaffold-based skeletal regeneration. Despite extensive studies on mineralized parenchymal tissue rebuilding, regenerating and maintaining undifferentiated mesenchyme within calvarial bone remain very challenging with limited advances yet. Current knowledge has evidenced the indispensability of rebuilding suture mesenchymal stem cell niches to avoid severe brain or even systematic damage. But to date, the absence of promising therapeutic biomaterials/scaffolds remains. The reason lies in the shortage of fundamental knowledge and methodological evidence to understand the cellular fate regulations of scaffolds. To address these issues, in this study, we systematically investigated the cellular fate determinations and transcriptomic mechanisms by distinct types of commonly used calvarial scaffolds. Our data elucidated the natural processes without scaffold transplantation and demonstrated how different scaffolds altered in vivo cellular responses. A feasible scaffold, polylactic acid electrospinning membrane (PLA), was next identified to precisely control mesenchymal ingrowth and self-renewal to rebuild non-osteogenic suture-like tissue at the defect center, meanwhile supporting proper osteointegration with defect bony edges. Especially, transcriptome analysis and cellular mechanisms underlying the well-orchestrated cell fate determination of PLA were deciphered. This study for the first time cellularly decoded the fate regulations of scaffolds in suture-bony composite defect healing, offering clinicians potential choices for regenerating such complicated injuries.


Subject(s)
Bone Regeneration , Tissue Scaffolds , Transcriptome , Animals , Bone Regeneration/physiology , Polyesters , Skull/surgery , Mesenchymal Stem Cells , Mesoderm/cytology , Cell Differentiation , Tissue Engineering/methods , Cranial Sutures , Biocompatible Materials
14.
PLoS Biol ; 22(4): e3002590, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38683849

ABSTRACT

Brain pericytes are one of the critical cell types that regulate endothelial barrier function and activity, thus ensuring adequate blood flow to the brain. The genetic pathways guiding undifferentiated cells into mature pericytes are not well understood. We show here that pericyte precursor populations from both neural crest and head mesoderm of zebrafish express the transcription factor nkx3.1 develop into brain pericytes. We identify the gene signature of these precursors and show that an nkx3.1-, foxf2a-, and cxcl12b-expressing pericyte precursor population is present around the basilar artery prior to artery formation and pericyte recruitment. The precursors later spread throughout the brain and differentiate to express canonical pericyte markers. Cxcl12b-Cxcr4 signaling is required for pericyte attachment and differentiation. Further, both nkx3.1 and cxcl12b are necessary and sufficient in regulating pericyte number as loss inhibits and gain increases pericyte number. Through genetic experiments, we have defined a precursor population for brain pericytes and identified genes critical for their differentiation.


Subject(s)
Brain , Pericytes , Transcription Factors , Zebrafish Proteins , Animals , Brain/metabolism , Brain/embryology , Cell Differentiation , Chemokine CXCL12/metabolism , Chemokine CXCL12/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Mesoderm/metabolism , Mesoderm/cytology , Neural Crest/metabolism , Neural Crest/cytology , Pericytes/metabolism , Pericytes/cytology , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Signal Transduction , Transcription Factors/metabolism , Transcription Factors/genetics , Zebrafish/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics
15.
Development ; 151(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38646822

ABSTRACT

The precise assembly of tissues and organs relies on spatiotemporal regulation of gene expression to coordinate the collective behavior of cells. In Drosophila embryos, the midgut musculature is formed through collective migration of caudal visceral mesoderm (CVM) cells, but how gene expression changes as cells migrate is not well understood. Here, we have focused on ten genes expressed in the CVM and the cis-regulatory sequences controlling their expression. Although some genes are continuously expressed, others are expressed only early or late during migration. Late expression relates to cell cycle progression, as driving string/Cdc25 causes earlier division of CVM cells and accelerates the transition to late gene expression. In particular, we found that the cell cycle effector transcription factor E2F1 is a required input for the late gene CG5080. Furthermore, whereas late genes are broadly expressed in all CVM cells, early gene transcripts are polarized to the anterior or posterior ends of the migrating collective. We show this polarization requires transcription factors Snail, Zfh1 and Dorsocross. Collectively, these results identify two sequential gene expression programs bridged by cell division that support long-distance directional migration of CVM cells.


Subject(s)
Cell Division , Cell Movement , Drosophila Proteins , Gene Expression Regulation, Developmental , Animals , Cell Movement/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Cell Division/genetics , Mesoderm/metabolism , Mesoderm/cytology , Transcription Factors/metabolism , Transcription Factors/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/embryology , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/genetics , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/cytology , Drosophila/genetics , Drosophila/metabolism , Drosophila/embryology , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics
17.
Dev Cell ; 59(10): 1302-1316.e5, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38569553

ABSTRACT

The planar cell polarity (PCP) complex is speculated to function in murine lung development, where branching morphogenesis generates an epithelial tree whose distal tips expand dramatically during sacculation. Here, we show that PCP is dispensable in the airway epithelium for sacculation. Rather, we find a Celsr1-independent role for the PCP component Vangl in the pulmonary mesenchyme: loss of Vangl1/2 inhibits mesenchymal thinning and expansion of the saccular epithelium. Further, loss of mesenchymal Wnt5a mimics sacculation defects observed in Vangl2-mutant lungs, implicating mesenchymal Wnt5a/Vangl signaling as a key regulator of late lung morphogenesis. A computational model predicts that sacculation requires a fluid mesenchymal compartment. Lineage-tracing and cell-shape analyses are consistent with the mesenchyme acting as a fluid tissue, suggesting that loss of Vangl1/2 impacts the ability of mesenchymal cells to exchange neighbors. Our data thus identify an explicit function for Vangl and the pulmonary mesenchyme in actively shaping the saccular epithelium.


Subject(s)
Cell Polarity , Lung , Mesoderm , Morphogenesis , Nerve Tissue Proteins , Animals , Mesoderm/metabolism , Mice , Lung/metabolism , Lung/pathology , Lung/embryology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Wnt-5a Protein/metabolism , Wnt-5a Protein/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Signal Transduction , Organogenesis/genetics , Receptors, G-Protein-Coupled
18.
PLoS Biol ; 22(4): e3002611, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38683880

ABSTRACT

As tissues grow and change shape during animal development, they physically pull and push on each other, and these mechanical interactions can be important for morphogenesis. During Drosophila gastrulation, mesoderm invagination temporally overlaps with the convergence and extension of the ectodermal germband; the latter is caused primarily by Myosin II-driven polarised cell intercalation. Here, we investigate the impact of mesoderm invagination on ectoderm extension, examining possible mechanical and mechanotransductive effects on Myosin II recruitment and polarised cell intercalation. We find that the germband ectoderm is deformed by the mesoderm pulling in the orthogonal direction to germband extension (GBE), showing mechanical coupling between these tissues. However, we do not find a significant change in Myosin II planar polarisation in response to mesoderm invagination, nor in the rate of junction shrinkage leading to neighbour exchange events. We conclude that the main cellular mechanism of axis extension, polarised cell intercalation, is robust to the mesoderm invagination pull. We find, however, that mesoderm invagination slows down the rate of anterior-posterior cell elongation that contributes to axis extension, counteracting the tension from the endoderm invagination, which pulls along the direction of GBE.


Subject(s)
Drosophila melanogaster , Ectoderm , Gastrulation , Mesoderm , Myosin Type II , Animals , Mesoderm/embryology , Mesoderm/cytology , Gastrulation/physiology , Ectoderm/cytology , Ectoderm/embryology , Ectoderm/metabolism , Myosin Type II/metabolism , Drosophila melanogaster/embryology , Cell Polarity , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Embryo, Nonmammalian , Morphogenesis , Body Patterning/physiology , Drosophila/embryology
19.
Stem Cell Reports ; 19(5): 618-628, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38579708

ABSTRACT

SOX2 is a transcription factor involved in the regulatory network maintaining the pluripotency of embryonic stem cells in culture as well as in early embryos. In addition, SOX2 plays a pivotal role in neural stem cell formation and neurogenesis. How SOX2 can serve both processes has remained elusive. Here, we identified a set of SOX2-dependent neural-associated enhancers required for neural lineage priming. They form a distinct subgroup (1,898) among 8,531 OCT4/SOX2/NANOG-bound enhancers characterized by enhanced SOX2 binding and chromatin accessibility. Activation of these enhancers is triggered by neural induction of wild-type cells or by default in Smad4-ablated cells resistant to mesoderm induction and is antagonized by mesodermal transcription factors via Sox2 repression. Our data provide mechanistic insight into the transition from the pluripotency state to the early neural fate and into the regulation of early neural versus mesodermal specification in embryonic stem cells and embryos.


Subject(s)
Enhancer Elements, Genetic , Mesoderm , Neural Stem Cells , SOXB1 Transcription Factors , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/genetics , Animals , Mice , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Mesoderm/cytology , Mesoderm/metabolism , Neurogenesis , Gene Expression Regulation, Developmental , Octamer Transcription Factor-3/metabolism , Octamer Transcription Factor-3/genetics , Cell Differentiation/genetics , Nanog Homeobox Protein/metabolism , Nanog Homeobox Protein/genetics , Cell Lineage/genetics , Smad4 Protein/metabolism , Smad4 Protein/genetics , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/cytology , Chromatin/metabolism , Protein Binding
20.
FASEB J ; 38(9): e23632, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38686936

ABSTRACT

The upper Müllerian duct (MD) is patterned and specified into two morphologically and functionally distinct organs, the oviduct and uterus. It is known that this regionalization process is instructed by inductive signals from the adjacent mesenchyme. However, the interaction landscape between epithelium and mesenchyme during upper MD development remains largely unknown. Here, we performed single-cell transcriptomic profiling of mouse neonatal oviducts and uteri at the initiation of MD epithelial differentiation (postnatal day 3). We identified major cell types including epithelium, mesenchyme, pericytes, mesothelium, endothelium, and immune cells in both organs with established markers. Moreover, we uncovered region-specific epithelial and mesenchymal subpopulations and then deduced region-specific ligand-receptor pairs mediating mesenchymal-epithelial interactions along the craniocaudal axis. Unexpectedly, we discovered a mesenchymal subpopulation marked by neurofilaments with specific localizations at the mesometrial pole of both the neonatal oviduct and uterus. Lastly, we analyzed and revealed organ-specific signature genes of pericytes and mesothelial cells. Taken together, our study enriches our knowledge of upper MD development, and provides a manageable list of potential genes, pathways, and region-specific cell subtypes for future functional studies.


Subject(s)
Mullerian Ducts , Oviducts , Single-Cell Analysis , Transcriptome , Uterus , Animals , Female , Mice , Uterus/metabolism , Uterus/cytology , Mullerian Ducts/metabolism , Oviducts/metabolism , Oviducts/cytology , Gene Expression Profiling , Animals, Newborn , Cell Differentiation , Mesoderm/metabolism , Mesoderm/cytology , Epithelial Cells/metabolism , Mice, Inbred C57BL , Gene Expression Regulation, Developmental
SELECTION OF CITATIONS
SEARCH DETAIL
...