Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 972
Filter
1.
Methods Mol Biol ; 2805: 187-201, 2024.
Article in English | MEDLINE | ID: mdl-39008183

ABSTRACT

Epidermal tissues are among the most striking examples of planar polarity. Insect bristles, fish scales, and mammalian fur are all uniformly oriented along an animal's body axis. The collective alignment of epidermal structures provides a valuable system to interrogate the signaling mechanisms that coordinate cellular behaviors at both local and tissue-levels. Here, we provide methods to analyze the planar organization of hair follicles within the mouse epidermis. Hair follicles are specified and bud into the underlying dermis during embryonic development. Shortly after, follicle cells dynamically rearrange to orient each follicle toward the anterior of the animal. When directional signaling is disrupted, hair follicles become misoriented. In this chapter, we describe how to create a spatial map of hair follicle orientations to reveal tissue-scale patterns in both embryonic and postnatal skin. Additionally, we provide a live imaging protocol that can be used to monitor cell movements in embryonic skin explants to reveal the cellular behaviors that polarize the hair follicle itself.


Subject(s)
Cell Polarity , Epidermis , Hair Follicle , Animals , Mice , Hair Follicle/cytology , Hair Follicle/embryology , Cell Polarity/physiology , Epidermis/embryology , Epidermis/metabolism , Epidermal Cells/cytology , Cell Movement
2.
Dev Biol ; 515: 60-66, 2024 Nov.
Article in English | MEDLINE | ID: mdl-38964706

ABSTRACT

Terminal differentiation of epithelial cells is critical for the barrier function of the skin, the growth of skin appendages, such as hair and nails, and the development of the skin of amniotes. Here, we present the hypothesis that the differentiation of cells in the embryonic periderm shares characteristic features with the differentiation of epithelial cells that support the morphogenesis of cornified skin appendages during postnatal life. The periderm prevents aberrant fusion of adjacent epithelial sites during early skin development. It is shed off when keratinocytes of the epidermis form the cornified layer, the stratum corneum. A similar role is played by epithelia that ensheath cornifying skin appendages until they disintegrate to allow the separation of the mature part of the skin appendage from the adjacent tissue. These epithelia, exemplified by the inner root sheath of hair follicles and the epithelia close to the free edge of nails or claws, are referred to as scaffolding epithelia. The periderm and scaffolding epithelia are similar with regard to their transient functions in separating tissues and the conserved expression of trichohyalin and trichohyalin-like genes in mammals and birds. Thus, we propose that parts of the peridermal differentiation program were coopted to a new postnatal function during the evolution of cornified skin appendages in amniotes.


Subject(s)
Cell Differentiation , Cell Differentiation/physiology , Animals , Skin/embryology , Skin/cytology , Skin/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelium/embryology , Epithelium/metabolism , Epidermis/embryology , Epidermis/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , Hair Follicle/embryology , Hair Follicle/cytology , Humans , Morphogenesis
3.
Dev Biol ; 511: 12-25, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38556137

ABSTRACT

During epithelial morphogenesis, the apical junctions connecting cells must remodel as cells change shape and make new connections with their neighbors. In the C. elegans embryo, new apical junctions form when epidermal cells migrate and seal with one another to encase the embryo in skin ('ventral enclosure'), and junctions remodel when epidermal cells change shape to squeeze the embryo into a worm shape ('elongation'). The junctional cadherin-catenin complex (CCC), which links epithelial cells to each other and to cortical actomyosin, is essential for C. elegans epidermal morphogenesis. RNAi genetic enhancement screens have identified several genes encoding proteins that interact with the CCC to promote epidermal morphogenesis, including the scaffolding protein Afadin (AFD-1), whose depletion alone results in only minor morphogenesis defects. Here, by creating a null mutation in afd-1, we show that afd-1 provides a significant contribution to ventral enclosure and elongation on its own. Unexpectedly, we find that afd-1 mutant phenotypes are strongly modified by diet, revealing a previously unappreciated parental nutritional input to morphogenesis. We identify functional interactions between AFD-1 and the CCC by demonstrating that E-cadherin is required for the polarized distribution of AFD-1 to cell contact sites in early embryos. Finally, we show that afd-1 promotes the enrichment of polarity regulator, and CCC-interacting protein, PAC-1/ARHGAP21 to cell contact sites, and we identify genetic interactions suggesting that afd-1 and pac-1 regulate epidermal morphogenesis at least in part through parallel mechanisms. Our findings reveal that C. elegans AFD-1 makes a significant contribution to epidermal morphogenesis and functionally interfaces with core and associated CCC proteins.


Subject(s)
Cadherins , Caenorhabditis elegans Proteins , Caenorhabditis elegans , Epidermis , Morphogenesis , Animals , Cadherins/metabolism , Cadherins/genetics , Caenorhabditis elegans/embryology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Catenins/metabolism , Catenins/genetics , Epidermal Cells/metabolism , Epidermis/metabolism , Epidermis/embryology , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/genetics , Microfilament Proteins/metabolism , Microfilament Proteins/genetics
4.
Arch Biochem Biophys ; 711: 109003, 2021 10 30.
Article in English | MEDLINE | ID: mdl-34390735

ABSTRACT

During fetal development, the barrier function of the fetal skin is developed under specific conditions for epidermis formation. In keratinocyte differentiation, the well-orchestrated production and modification of various structural proteins are induced. We assessed the epidermal barrier function in different fetal stages by evaluating the enzymatic activity of cross-linking proteins, transglutaminases, and the permeation of fluorescence dye in the stained epidermal sections. During days 15.5-17.5 in gestation, the enzymatic activities in the epidermis appeared to increase significantly; meanwhile, dye permeation was substantially decreased, suggesting the formation of a protective barrier. For the fetal epidermis formation in the earlier stage, unclarified stimulating factors in the amniotic fluid (AF) are possible to promote barrier function by stimulating keratinocyte differentiation. Thus, we performed proteomic spectrometric (MS) analysis on the components in the AF at different fetal stages. Also, we investigated the promotive ability of the components using a cultured keratinocyte differentiation system. According to the MS analysis, the AF components appeared to exhibit stage-specific variations, where possible unique functions have been identified. We also found that adding the AF from each stage to the medium for cultured keratinocytes specifically enhanced the levels of the differentiation markers. These results provide information on the possible role of AF that contains regulatory factors on keratinocyte differentiation.


Subject(s)
Amniotic Fluid/metabolism , Cell Differentiation , Keratinocytes/cytology , Animals , Cells, Cultured , Epidermal Cells/metabolism , Epidermis/embryology , Epidermis/metabolism , Fetus , Mice, Inbred ICR , Transglutaminases/metabolism
5.
Dev Biol ; 478: 25-40, 2021 10.
Article in English | MEDLINE | ID: mdl-34166654

ABSTRACT

Skin development and patterning is dependent on factors that regulate the stepwise differentiation of dermal fibroblasts concomitant with dermal-epidermal reciprocal signaling, two processes that are poorly understood. Here we show that dermal EZH2, the methyltransferase enzyme of the epigenetic Polycomb Repressive Complex 2 (PRC2), is a new coordinator of both these processes. Dermal EZH2 activity is present during dermal fibroblast differentiation and is required for spatially restricting Wnt/ß-catenin signaling to reinforce dermal fibroblast cell fate. Later in development, dermal EZH2 regulates the expression of reticular dermal markers and initiation of secondary hair follicles. Embryos lacking dermal Ezh2 have elevated epidermal proliferation and differentiation that can be rescued by small molecule inhibition of retinoic acid (RA) signaling. Together, our study reveals that dermal EZH2 is acting like a rheostat to control the levels of Wnt/ß-catenin and RA signaling to impact fibroblast differentiation cell autonomously and epidermal keratinocyte development non-cell autonomously, respectively.


Subject(s)
Dermis/cytology , Dermis/embryology , Enhancer of Zeste Homolog 2 Protein/metabolism , Epidermis/embryology , Fibroblasts/cytology , Keratinocytes/cytology , Polycomb Repressive Complex 2/metabolism , Animals , Cell Differentiation , Cell Proliferation , Dermis/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Epidermis/metabolism , Fibroblasts/metabolism , Hyperplasia , Keratinocytes/metabolism , Mice , Organogenesis , Retinoids/pharmacology , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Tretinoin/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
6.
Dev Biol ; 477: 177-190, 2021 09.
Article in English | MEDLINE | ID: mdl-34038742

ABSTRACT

Teleost fish fins, like all vertebrate limbs, comprise a series of bones laid out in characteristic pattern. Each fin's distal bony rays typically branch to elaborate skeletal networks providing form and function. Zebrafish caudal fin regeneration studies suggest basal epidermal-expressed Sonic hedgehog (Shh) promotes ray branching by partitioning pools of adjacent pre-osteoblasts. This Shh role is distinct from its well-studied Zone of Polarizing Activity role establishing paired limb positional information. Therefore, we investigated if and how Shh signaling similarly functions during developmental ray branching of both paired and unpaired fins while resolving cellular dynamics of branching by live imaging. We found shha is expressed uniquely by basal epidermal cells overlying pre-osteoblast pools at the distal aspect of outgrowing juvenile fins. Lateral splitting of each shha-expressing epidermal domain followed by the pre-osteoblast pools precedes overt ray branching. We use ptch2:Kaede fish and Kaede photoconversion to identify short stretches of shha+basal epidermis and juxtaposed pre-osteoblasts as the Shh/Smoothened (Smo) active zone. Basal epidermal distal collective movements continuously replenish each shha+domain with individual cells transiently expressing and responding to Shh. In contrast, pre-osteoblasts maintain Shh/Smo activity until differentiating. The Smo inhibitor BMS-833923 prevents branching in all fins, paired and unpaired, with surprisingly minimal effects on caudal fin initial skeletal patterning, ray outgrowth or bone differentiation. Staggered BMS-833923 addition indicates Shh/Smo signaling acts throughout the branching process. We use live cell tracking to find Shh/Smo restrains the distal movement of basal epidermal cells by apparent 'tethering' to pre-osteoblasts. We propose short-range Shh/Smo signaling promotes these heterotypic associations to couple instructive basal epidermal collective movements to pre-osteoblast repositioning as a unique mode of branching morphogenesis.


Subject(s)
Animal Fins/embryology , Epidermal Cells/physiology , Epidermis/embryology , Hedgehog Proteins/physiology , Morphogenesis , Zebrafish Proteins/physiology , Animal Fins/cytology , Animal Fins/metabolism , Animals , Benzamides/pharmacology , Cell Movement , Epidermis/metabolism , Patched-2 Receptor/metabolism , Quinazolines/pharmacology , Signal Transduction/drug effects , Smoothened Receptor/physiology , Zebrafish
7.
Genes (Basel) ; 12(2)2021 02 10.
Article in English | MEDLINE | ID: mdl-33578693

ABSTRACT

Scaffoldin, an S100 fused-type protein (SFTP) with high amino acid sequence similarity to the mammalian hair follicle protein trichohyalin, has been identified in reptiles and birds, but its functions are not yet fully understood. Here, we investigated the expression pattern of scaffoldin and cornulin, a related SFTP, in the developing beaks of birds. We determined the mRNA levels of both SFTPs by reverse transcription polymerase chain reaction (RT-PCR) in the beak and other ectodermal tissues of chicken (Gallus gallus) and quail (Coturnix japonica) embryos. Immunohistochemical staining was performed to localize scaffoldin in tissues. Scaffoldin and cornulin were expressed in the beak and, at lower levels, in other embryonic tissues of both chickens and quails. Immunohistochemistry revealed scaffoldin in the peridermal compartment of the egg tooth, a transitory cornified protuberance (caruncle) on the upper beak which breaks the eggshell during hatching. Furthermore, scaffoldin marked a multilayered peridermal structure on the lower beak. The results of this study suggest that scaffoldin plays an evolutionarily conserved role in the development of the avian beak with a particular function in the morphogenesis of the egg tooth.


Subject(s)
Avian Proteins/genetics , Beak/metabolism , Chickens/genetics , Coturnix/genetics , Feathers/metabolism , Hoof and Claw/metabolism , Animals , Avian Proteins/metabolism , Beak/cytology , Beak/embryology , Biological Evolution , Chick Embryo , Chickens/growth & development , Chickens/metabolism , Conserved Sequence , Coturnix/embryology , Coturnix/metabolism , Embryo, Nonmammalian , Epidermis/embryology , Epidermis/metabolism , Feathers/cytology , Feathers/embryology , Gene Expression Regulation, Developmental , Hoof and Claw/cytology , Hoof and Claw/embryology , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , Mammals , Morphogenesis/genetics , Zygote/growth & development , Zygote/metabolism
8.
Dev Biol ; 469: 46-53, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33065118

ABSTRACT

Ciliopathies affect a variety of tissues during development including the heart, kidneys, respiratory tract, and retina. Though an increasing number of monogenic causes of ciliopathies have been described, many remain unexplained. Recently, recessive variants in NUP93 and NUP205 encoding two proteins of the inner ring of the nuclear pore complex were implicated as causes of steroid resistant nephrotic syndrome. In addition, we previously found that the inner ring nucleoporins NUP93 and NUP188 function in proper left-right patterning in developing embryos via a role at the cilium. Here, we describe the role of an additional inner ring nucleoporin NUP205 in cilia biology and establishment of normal organ situs. Using knockdown in Xenopus, we show that Nup205 depletion results in loss of cilia and abnormal cardiac morphology. Furthermore, by transmission electron microscopy, we observe a loss of cilia and mispositioning of intracellular ciliary structures such as basal bodies and rootlets upon depleting inner ring nucleoporins. We describe a model wherein NUP93 interacting with either NUP188 or NUP205 is necessary for cilia. We thus provide evidence that dysregulation of inner ring nucleoporin genes that have been identified in patients may contribute to pathogenesis through cilia dysfunction.


Subject(s)
Cilia/physiology , Nuclear Pore Complex Proteins/physiology , Xenopus Proteins/physiology , Animals , Body Patterning , Cilia/ultrastructure , Epidermis/embryology , Epidermis/ultrastructure , Gene Knockdown Techniques , Heart Defects, Congenital/genetics , Humans , Nuclear Pore Complex Proteins/genetics , Pronephros/ultrastructure , Xenopus/embryology , Xenopus Proteins/genetics
9.
Dev Biol ; 470: 121-135, 2021 02.
Article in English | MEDLINE | ID: mdl-33248112

ABSTRACT

Actin filament crosslinking, bundling and molecular motor proteins are necessary for the assembly of epithelial projections such as microvilli, stereocilia, hairs, and bristles. Mutations in such proteins cause defects in the shape, structure, and function of these actin - based protrusions. One protein necessary for stereocilia formation, Myosin VIIA, is an actin - based motor protein conserved throughout phylogeny. In Drosophila melanogaster, severe mutations in the MyoVIIA homolog crinkled (ck) are "semi - lethal" with only a very small percentage of flies surviving to adulthood. Such survivors show morphological defects related to actin bundling in hairs and bristles. To better understand ck/MyoVIIA's function in bundled - actin structures, we used dominant female sterile approaches to analyze the loss of maternal and zygotic (M/Z) ck/MyoVIIA in the morphogenesis of denticles, small actin - based projections on the ventral epidermis of Drosophila embryos. M/Z ck mutants displayed severe defects in denticle morphology - actin filaments initiated in the correct location, but failed to elongate and bundle to form normal projections. Using deletion mutant constructs, we demonstrated that both of the C - terminal MyTH4 and FERM domains are necessary for proper denticle formation. Furthermore, we show that ck/MyoVIIA interacts genetically with dusky - like (dyl), a member of the ZPD family of proteins that links the extracellular matrix to the plasma membrane, and when mutated also disrupts normal denticle formation. Loss of either protein alone does not alter the localization of the other; however, loss of the two proteins together dramatically enhances the defects in denticle shape observed when either protein alone was absent. Our data indicate that ck/MyoVIIA plays a key role in the formation and/or organization of actin filament bundles, which drive proper shape of cellular projections.


Subject(s)
Actin Cytoskeleton/ultrastructure , Cell Surface Extensions/ultrastructure , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Myosin VIIa/metabolism , Actin Cytoskeleton/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Epidermis/embryology , Female , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Morphogenesis , Mutant Proteins/metabolism , Mutation , Myosin VIIa/genetics
10.
Open Biol ; 10(12): 200214, 2020 12.
Article in English | MEDLINE | ID: mdl-33292106

ABSTRACT

Tissue function and shape rely on the organization of the extracellular matrix (ECM) produced by the respective cells. Our understanding of the underlying molecular mechanisms is limited. Here, we show that extracellular Tweedle (Twdl) proteins in the fruit fly Drosophila melanogaster form two adjacent two-dimensional sheets underneath the cuticle surface and above a distinct layer of dityrosinylated and probably elastic proteins enwrapping the whole body. Dominant mutations in twdl genes cause ectopic spherical aggregation of Twdl proteins that recruit dityrosinylated proteins at their periphery within lower cuticle regions. These aggregates perturb parallel ridges at the surface of epidermal cells that have been demonstrated to be crucial for body shaping. In one scenario, hence, this disorientation of epidermal ridges may explain the squatty phenotype of Twdl mutant larvae. In an alternative scenario, this phenotype may be due to the depletion of the dityrosinylated and elastic layer, and the consequent weakening of cuticle resistance against the internal hydrostatic pressure. According to Barlow's formula describing the distribution of internal pressure forces in pipes in dependence of pipe wall material properties, it follows that this reduction in turn causes lateral expansion at the expense of the antero-posterior elongation of the body.


Subject(s)
Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Cell Shape/genetics , Extracellular Matrix/metabolism , Morphogenesis/genetics , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism , Alleles , Animals , Biomarkers , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Embryo, Nonmammalian , Embryonic Development/genetics , Epidermis/embryology , Epidermis/metabolism , Epidermis/ultrastructure , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Larva , Mutation , Phenotype
11.
Development ; 147(22)2020 11 15.
Article in English | MEDLINE | ID: mdl-33191273

ABSTRACT

Cell divisions and cell-fate decisions require stringent regulation for proper tissue development and homeostasis. The mammalian epidermis is a highly organized tissue structure that is sustained by epidermal stem cells (ESCs) that balance self-renewal and cell-fate decisions to establish a protective barrier, while replacing dying cells during homeostasis and in response to injury. Extensive work over past decades has provided insights into the regulatory mechanisms that control ESC specification, self-renewal and maintenance during different stages of the lifetime of an organism. In this Review, we discuss recent findings that have furthered our understanding of key regulatory features that allow ESCs to establish a functional barrier during development and to maintain tissue homeostasis in adults.


Subject(s)
Epidermal Cells/metabolism , Epidermis/embryology , Epidermis/growth & development , Homeostasis/genetics , Stem Cells/metabolism , Animals , Cell Differentiation/physiology , Cell Lineage/physiology , Cell Proliferation/physiology , Cell Self Renewal/physiology , Humans , Transcription, Genetic , Wound Healing/physiology
12.
Nat Commun ; 11(1): 5434, 2020 10 28.
Article in English | MEDLINE | ID: mdl-33116143

ABSTRACT

The interfollicular epidermis (IFE) forms a water-tight barrier that is often disrupted in inflammatory skin diseases. During homeostasis, the IFE is replenished by stem cells in the basal layer that differentiate as they migrate toward the skin surface. Conventionally, IFE differentiation is thought to be stepwise as reflected in sharp boundaries between its basal, spinous, granular and cornified layers. The transcription factor GRHL3 regulates IFE differentiation by transcriptionally activating terminal differentiation genes. Here we use single cell RNA-seq to show that murine IFE differentiation is best described as a single step gradualistic process with a large number of transition cells between the basal and spinous layer. RNA-velocity analysis identifies a commitment point that separates the plastic basal and transition cell state from unidirectionally differentiating cells. We also show that in addition to promoting IFE terminal differentiation, GRHL3 is essential for suppressing epidermal stem cell expansion and the emergence of an abnormal stem cell state by suppressing Wnt signaling in stem cells.


Subject(s)
DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epidermal Cells/cytology , Epidermal Cells/metabolism , Transcription Factors/metabolism , Animals , Animals, Newborn , Cell Differentiation , Cell Lineage , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Epidermis/embryology , Epidermis/metabolism , Female , Gene Expression Profiling , Gestational Age , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Single-Cell Analysis , Transcription Factors/deficiency , Transcription Factors/genetics
13.
Article in English | MEDLINE | ID: mdl-32975294

ABSTRACT

INTRODUCTION: Galectins constitute a phylogenetically conserved family of proteins with high binding affinity for glycoconjugates bearing ß-galactoside residues. Surprisingly, knowledge of the expression pattern of galectins during human epidermal morphogenesis is very limited. METHODS: Fifty-eight biopsy skin specimens obtained from human embryos and 10 biopsy specimens obtained from healthy adult volunteers were processed for immunohistochemistry using a panel of antibodies against galectins 1, 3, 7, and 9. RESULTS: Fetal human epidermis was devoid of any galectin 1 immunoreactivity, whereas clear-cut changes were found in the galectin 3 immunoreactivity of fetal epidermis with advancing gestational age. The expression pattern of galectins 7 and 9 remained constant at all stages of gestation. CONCLUSIONS: The changes in galectin 3 immunoreactivity of human fetal epidermis with advancing gestational age, which are reported here for the first time, suggest that this galectin and its ligands may be implicated in the molecular events underlying human epidermal morphogenesis. It remains to be elucidated in future investigations whether the expression pattern of galectins, particularly that of galectin 3, in the developing human epidermis reveals alterations in fetuses with inherited cutaneous disorders that may be important for prenatal diagnosis of these disorders.


Subject(s)
Epidermis/embryology , Epidermis/metabolism , Fetus/embryology , Fetus/metabolism , Galectins/metabolism , Adult , Age Factors , Female , Gestational Age , Humans , Male , Reference Values , Young Adult
14.
Philos Trans R Soc Lond B Biol Sci ; 375(1809): 20190682, 2020 10 12.
Article in English | MEDLINE | ID: mdl-32829688

ABSTRACT

Correct cell shape is indispensable for tissue architecture, with cell shape being determined by cortical actin and surface adhesion. The role of adhesion in remodelling tissue is to counteract the deformation of cells by force, resulting from actomyosin contractility, and to maintain tissue integrity. The dynamics of this adhesion are critical to the processes of cell shape formation and maintenance. Here, we show that the trafficking molecule Arf6 has a direct impact on cell elongation, by acting to stabilize E-cadherin-based adhesion complexes at the cell surface, in addition to its canonical role in endocytosis. We demonstrate that these functions of Arf6 are dependent on the molecule Flotillin1, which recruits Arf6 to the plasma membrane. Our data suggest that Arf6 and Flotillin1 operate in a pathway distinct from clathrin-mediated endocytosis. Altogether, we demonstrate that Arf6- and Flotillin1-dependent regulation of the dynamics of cell adhesion contribute to moulding tissue in vivo. This article is part of the discussion meeting issue 'Contemporary morphogenesis'.


Subject(s)
ADP-Ribosylation Factors/genetics , Drosophila Proteins/genetics , Drosophila/embryology , Drosophila/genetics , Embryo, Nonmammalian/embryology , Epidermis/embryology , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/metabolism , Animals , Cell Adhesion , Cell Movement , Drosophila Proteins/metabolism , Protein Transport
15.
Curr Osteoporos Rep ; 18(4): 378-387, 2020 08.
Article in English | MEDLINE | ID: mdl-32748325

ABSTRACT

PURPOSE OF REVIEW: Epigenetic regulation is a distinct mechanism of gene regulation that functions by modulating chromatin structure and accessibility. Polycomb Repressive Complex 2 (PRC2) is a conserved chromatin regulator that is required in the developing embryo to control the expression of key developmental genes. An emerging feature of PRC2 is that it not only allows for binary ON/OFF states of gene expression but can also modulate gene expression in feed-forward loops to change the outcome of gene regulatory networks. This striking feature of epigenetic modulation has improved our understanding of musculoskeletal development. RECENT FINDINGS: Recent advances in mouse embryos unravel a range of phenotypes that demonstrate the tissue-specific, temporal, and spatial role of PRC2 during organogenesis and cell fate decisions in vivo. Here, we take a detailed view of how PRC2 functions during the development of calvarial bone and skin. Based on the emerging evidence, we propose that PRC2 serves as a "dimmer switch" to modulate gene expression of target genes by altering the expression of activators and inhibitors. This review highlights the findings from contemporary research that allow us to investigate the unique developmental potential of intramembranous calvarial bones.


Subject(s)
Bone Development/genetics , Epidermis/embryology , Gene Expression Regulation, Developmental/genetics , Polycomb Repressive Complex 2/genetics , Skull/embryology , Animals , Humans , Mice
16.
Int J Dev Biol ; 64(4-5-6): 343-352, 2020.
Article in English | MEDLINE | ID: mdl-32658994

ABSTRACT

The epidermis, being the outermost epithelial layer in metazoans, experiences multiple external and self-generated mechanical stimuli. The tissue-scale response to these mechanical stresses has been actively studied in the adult stratified epidermis. However, the response of the developing bi-layered epidermis to differential tension and its molecular regulation has remained poorly characterised. Here we report an oil injection based method, which in combination with atomic force microscopy (AFM), allows manipulation as well as estimation of tension in the developing epidermis. Our results show that the injection of mineral oil into the brain ventricle of developing zebrafish embryos stretches the overlying epidermis. The epidermal tension increases linearly with the injected volume of oil and the injection of 14-17 nL oil results in a two-fold increase in epidermal tension. This increase in epidermal tension is sufficient to elicit a physiological response characterised by temporal changes in the cell cross-sectional area and an increase in cell proliferation. Our data further indicate that the depletion of E-cadherin in the epidermis is detrimental for tissue integrity under increased mechanical stress. The application of this experimental paradigm in a genetically tractable organism such as zebrafish can be useful in uncovering mechanisms of tension sustenance in the developing epidermis.


Subject(s)
Cadherins/metabolism , Embryo, Nonmammalian/metabolism , Epidermis/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Embryo, Nonmammalian/embryology , Epidermal Cells/metabolism , Epidermis/embryology , Microscopy, Atomic Force/methods , Microscopy, Confocal/methods , Stress, Mechanical , Zebrafish/embryology
17.
Dev Biol ; 464(2): 188-201, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32562757

ABSTRACT

Dorsal closure during Drosophila embryogenesis provides a robust genetic platform to study the basic cellular mechanisms that govern epithelial wound healing and morphogenesis. As dorsal closure proceeds, the lateral epithelial tissue (LE) adjacent to the dorsal opening advance contra-laterally, with a simultaneous retraction of the amnioserosa. The process involves a fair degree of coordinated cell shape changes in the dorsal most epithelial (DME) cells as well as a few penultimate rows of lateral epithelial (LE) cells (collectively referred here as Dorsolateral Epithelial (DLE) cells), lining the periphery of the amnioserosa, which in due course of time extend contra-laterally and ultimately fuse over the dorsal hole, giving rise to a dorsal epithelial continuum. The JNK-Dpp signaling in the dorsolateral epidermis, plays an instrumental role in guiding their fate during this process. A large array of genes have been reported to be involved in the regulation of this core signaling pathway, yet the mechanisms by which they do so is hitherto unclear, which forms the objective of our present study. Here we show a probable mechanism via which lgl, a conserved tumour suppressor gene, regulates the JNK-Dpp pathway during dorsal closure and epithelial morphogenesis. A conditional/targeted knock-down of lgl in the dorsolateral epithelium of embryos results in failure of dorsal closure. Interestingly, we also observed a similar phenotype in a Rab11 knockdown condition. Our experiment suggests Rab11 to be interacting with lgl as they seem to synergize in order to regulate the core JNK-Dpp signaling pathway during dorsal closure and also during adult thorax closure process.


Subject(s)
Drosophila Proteins/metabolism , MAP Kinase Kinase 4/metabolism , Morphogenesis/physiology , Signal Transduction/physiology , Tumor Suppressor Proteins/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Epidermis/embryology , Epithelial Cells/cytology , Epithelial Cells/metabolism , MAP Kinase Kinase 4/genetics , Tumor Suppressor Proteins/genetics , rab GTP-Binding Proteins/genetics
18.
Genes Dev ; 34(13-14): 973-988, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32467224

ABSTRACT

Chromatin modifiers play critical roles in epidermal development, but the functions of histone deacetylases in this context are poorly understood. The class I HDAC, HDAC3, is of particular interest because it plays divergent roles in different tissues by partnering with tissue-specific transcription factors. We found that HDAC3 is expressed broadly in embryonic epidermis and is required for its orderly stepwise stratification. HDAC3 protein stability in vivo relies on NCoR and SMRT, which function redundantly in epidermal development. However, point mutations in the NCoR and SMRT deacetylase-activating domains, which are required for HDAC3's enzymatic function, permit normal stratification, indicating that HDAC3's roles in this context are largely independent of its histone deacetylase activity. HDAC3-bound sites are significantly enriched for predicted binding motifs for critical epidermal transcription factors including AP1, GRHL, and KLF family members. Our results suggest that among these, HDAC3 operates in conjunction with KLF4 to repress inappropriate expression of Tgm1, Krt16, and Aqp3 In parallel, HDAC3 suppresses expression of inflammatory cytokines through a Rela-dependent mechanism. These data identify HDAC3 as a hub coordinating multiple aspects of epidermal barrier acquisition.


Subject(s)
Cell Differentiation/genetics , Epidermal Cells/cytology , Epidermis/embryology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Animals , Embryo, Mammalian , Gene Deletion , Gene Expression Regulation, Developmental , Genes, Lethal/genetics , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Inbred C57BL , Mutation , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Co-Repressor 2/genetics , Nuclear Receptor Co-Repressor 2/metabolism , Protein Interaction Domains and Motifs/genetics , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
19.
Development ; 147(5)2020 03 11.
Article in English | MEDLINE | ID: mdl-32161061

ABSTRACT

The replacement of cells is a common strategy during animal development. In the Drosophila pupal abdomen, larval epidermal cells (LECs) are replaced by adult progenitor cells (histoblasts). Previous work showed that interactions between histoblasts and LECs result in apoptotic extrusion of LECs during early pupal development. Extrusion of cells is closely preceded by caspase activation and is executed by contraction of a cortical actomyosin cable. Here, we identify a population of LECs that extrudes independently of the presence of histoblasts during late pupal development. Extrusion of these LECs is not closely preceded by caspase activation, involves a pulsatile medial actomyosin network, and correlates with a developmental time period when mechanical tension and E-cadherin turnover at adherens junctions is particularly high. Our work reveals a developmental switch in the cell extrusion mechanism that correlates with changes in tissue mechanical properties.


Subject(s)
Abdomen/embryology , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Epidermal Cells/cytology , Epidermis/embryology , Adherens Junctions/metabolism , Animals , Animals, Genetically Modified , Cadherins/metabolism , Caspases/metabolism , Cell Proliferation , Larva/cytology , Pupa/cytology , Stress, Mechanical
20.
Epigenetics ; 15(9): 932-948, 2020 09.
Article in English | MEDLINE | ID: mdl-32175801

ABSTRACT

Post-translational modifications on nucleosomal histones represent a key epigenetic regulatory mechanism to mediate the complex gene expression, DNA replication, and cell cycle changes that occur in embryonic cells undergoing lineage specification, maturation, and differentiation during development. Here, we investigated the dynamics of 13 key histone marks in epidermal cells at three distinct stages of embryonic skin development and identified significant changes that corresponded with the maturation of the proliferative basal epidermal cells and terminally differentiated cells in the stratified layers. In particular, H3K4me3 and H3K27ac were accumulated and became more prominent in the basal cells at later stages of epidermal development, while H3K27me3 was found to be low in the basal cells but highly enriched in the differentiated suprabasal cell types. Constitutive heterochromatin marked by H4K20me3 was also significantly elevated in differentiated epidermal cells at late gestation stages, which exhibited a concomitant loss of H4K16 acetylation. These differential chromatin profiles were established in the embryonic skin by gestation day 15 and further amplified at E18 and in postnatal skin. Our results reveal the dynamic chromatin states that occur as epidermal progenitor cells commit to the lineage and differentiate into the different cells of the stratified epidermis and provide insight to the underlying epigenetic pathways that support normal epidermal development and homoeostasis.


Subject(s)
Cell Differentiation , Cell Proliferation , Chromatin Assembly and Disassembly , Epidermis/embryology , Epithelial Cells/metabolism , Animals , Cell Line , Epithelial Cells/cytology , Epithelial Cells/physiology , Histone Code , Histones/chemistry , Histones/metabolism , Humans , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL