Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.405
Filter
Add more filters

Publication year range
1.
Cell ; 181(3): 604-620.e22, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32259486

ABSTRACT

During embryonic and postnatal development, organs and tissues grow steadily to achieve their final size at the end of puberty. However, little is known about the cellular dynamics that mediate postnatal growth. By combining in vivo clonal lineage tracing, proliferation kinetics, single-cell transcriptomics, and in vitro micro-pattern experiments, we resolved the cellular dynamics taking place during postnatal skin epidermis expansion. Our data revealed that harmonious growth is engineered by a single population of developmental progenitors presenting a fixed fate imbalance of self-renewing divisions with an ever-decreasing proliferation rate. Single-cell RNA sequencing revealed that epidermal developmental progenitors form a more uniform population compared with adult stem and progenitor cells. Finally, we found that the spatial pattern of cell division orientation is dictated locally by the underlying collagen fiber orientation. Our results uncover a simple design principle of organ growth where progenitors and differentiated cells expand in harmony with their surrounding tissues.


Subject(s)
Epidermal Cells/metabolism , Epidermis/growth & development , Skin/growth & development , Animals , Animals, Outbred Strains , Cell Differentiation/physiology , Cell Division/physiology , Cell Lineage/genetics , Cell Proliferation/physiology , Cells, Cultured , Epidermal Cells/pathology , Epidermis/metabolism , Female , Male , Mice , Mice, Transgenic , Stem Cells/cytology
2.
Cell ; 181(3): 492-494, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32234524

ABSTRACT

The 2020 Canada Gairdner International Award has been awarded to Elaine Fuchs for her discovery of the role of adult skin stem cells in homeostasis, wound repair, inflammation, and cancer. These insights have established a foundation for basic knowledge on how adult stem cells form, maintain, and repair tissues and have provided the groundwork for additional exploration and discovery of pathways in other stem cell systems.


Subject(s)
Adult Stem Cells/metabolism , Adult Stem Cells/physiology , Skin/metabolism , Animals , Awards and Prizes , Canada , Epidermal Cells/metabolism , Female , History, 20th Century , History, 21st Century , Homeostasis/physiology , Humans , Neoplasms/metabolism , Wound Healing/physiology
3.
Cell ; 169(4): 636-650.e14, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28434617

ABSTRACT

Tissue stem cells contribute to tissue regeneration and wound repair through cellular programs that can be hijacked by cancer cells. Here, we investigate such a phenomenon in skin, where during homeostasis, stem cells of the epidermis and hair follicle fuel their respective tissues. We find that breakdown of stem cell lineage confinement-granting privileges associated with both fates-is not only hallmark but also functional in cancer development. We show that lineage plasticity is critical in wound repair, where it operates transiently to redirect fates. Investigating mechanism, we discover that irrespective of cellular origin, lineage infidelity occurs in wounding when stress-responsive enhancers become activated and override homeostatic enhancers that govern lineage specificity. In cancer, stress-responsive transcription factor levels rise, causing lineage commanders to reach excess. When lineage and stress factors collaborate, they activate oncogenic enhancers that distinguish cancers from wounds.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cell Lineage , Epidermal Cells , Hair Follicle/cytology , Skin Neoplasms/pathology , Skin/cytology , Stem Cells/metabolism , Animals , Cell Line, Tumor , Chromatin/metabolism , Epidermis/metabolism , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Skin Neoplasms/metabolism , Transcription Factors/metabolism , Transcriptome , Transplantation, Heterologous , Wound Healing
4.
Genes Dev ; 35(5-6): 354-366, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33602871

ABSTRACT

Polycomb repressive complex 1 (PRC1) and PRC2 are critical epigenetic developmental regulators. PRC1 and PRC2 largely overlap in their genomic binding and cooperate to establish repressive chromatin domains demarcated by H2AK119ub and H3K27me3. However, the functional contribution of each complex to gene repression has been a subject of debate, and understanding of its physiological significance requires further studies. Here, using the developing murine epidermis as a paradigm, we uncovered a previously unappreciated functional redundancy between Polycomb complexes. Coablation of PRC1 and PRC2 in embryonic epidermal progenitors resulted in severe defects in epidermal stratification, a phenotype not observed in the single PRC1-null or PRC2-null epidermis. Molecular dissection indicated a loss of epidermal identity that was coupled to a strong derepression of nonlineage transcription factors, otherwise repressed by either PRC1 or PRC2 in the absence of its counterpart. Ectopic expression of subsets of PRC1/2-repressed nonepidermal transcription factors in wild-type epidermal stem cells was sufficient to suppress epidermal identity genes, highlighting the importance of functional redundancy between PRC1 and PRC2. Altogether, our studies show how PRC1 and PRC2 function as two independent counterparts, thereby providing a repressive safety net that protects and preserves lineage identity.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Epidermal Cells/cytology , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 2/metabolism , Polycomb-Group Proteins/metabolism , Animals , Embryonic Stem Cells/metabolism , Epidermal Cells/metabolism , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Mice , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 2/genetics , Polycomb-Group Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Nat Immunol ; 17(4): 441-50, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26855029

ABSTRACT

Epithelial tissues continually undergo apoptosis. Commensal organisms that inhabit the epithelium influence tissue homeostasis, in which regulatory T cells (Treg cells) have a central role. However, the physiological importance of epithelial cell apoptosis and how the number of Treg cells is regulated are both incompletely understood. Here we found that apoptotic epithelial cells negatively regulated the commensal-stimulated proliferation of Treg cells. Gut commensals stimulated CX3CR1(+)CD103(-)CD11b(+) dendritic cells (DCs) to produce interferon-ß (IFN-ß), which augmented the proliferation of Treg cells in the intestine. Conversely, phosphatidylserine exposed on apoptotic epithelial cells suppressed IFN-ß production by the DCs via inhibitory signaling mediated by the cell-surface glycoprotein CD300a and thus suppressed Treg cell proliferation. Our findings reveal a regulatory role for apoptotic epithelial cells in maintaining the number of Treg cell and tissue homeostasis.


Subject(s)
Apoptosis/immunology , Epidermis/immunology , Epithelial Cells/immunology , Gastrointestinal Microbiome/immunology , Interferon-beta/immunology , Intestinal Mucosa/immunology , Respiratory Mucosa/immunology , T-Lymphocytes, Regulatory/immunology , Allergens/toxicity , Animals , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , Colon/cytology , Colon/immunology , Dendritic Cells/immunology , Dermatitis, Allergic Contact/etiology , Dermatitis, Allergic Contact/immunology , Dermatitis, Allergic Contact/pathology , Dextran Sulfate/toxicity , Epidermal Cells , Flow Cytometry , Immunohistochemistry , Intestinal Mucosa/cytology , Langerhans Cells/immunology , Lung/cytology , Lung/immunology , Mice , Mice, Knockout , Ovalbumin/toxicity , Real-Time Polymerase Chain Reaction , Receptors, Immunologic/genetics , Respiratory Mucosa/cytology , Salmonella Infections/immunology , Salmonella typhimurium
6.
Nat Immunol ; 17(4): 414-21, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26901152

ABSTRACT

Cells of the immune system that reside in barrier epithelia provide a first line of defense against pathogens. Langerhans cells (LCs) and CD8(+) tissue-resident memory T cells (TRM cells) require active transforming growth factor-ß1 (TGF-ß) for epidermal residence. Here we found that integrins αvß6 and αvß8 were expressed in non-overlapping patterns by keratinocytes (KCs) and maintained the epidermal residence of LCs and TRM cells by activating latent TGF-ß. Similarly, the residence of dendritic cells and TRM cells in the small intestine epithelium also required αvß6. Treatment of the skin with ultraviolet irradiation decreased integrin expression on KCs and reduced the availability of active TGF-ß, which resulted in LC migration. Our data demonstrated that regulated activation of TGF-ß by stromal cells was able to directly control epithelial residence of cells of the immune system through a novel mechanism of intercellular communication.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epidermis/immunology , Intestinal Mucosa/immunology , Keratinocytes/immunology , Langerhans Cells/immunology , Transforming Growth Factor beta/immunology , Animals , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/cytology , Cell Movement , Epidermal Cells , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunity, Mucosal , Integrins/immunology , Intestinal Mucosa/cytology , Intestine, Small/cytology , Intestine, Small/immunology , Langerhans Cells/cytology , Mice , Mice, Knockout , Mink , Polymerase Chain Reaction , Stromal Cells , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transforming Growth Factor beta1/immunology
7.
Immunity ; 50(3): 655-667.e4, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30893588

ABSTRACT

Restoration of barrier-tissue integrity after injury is dependent on the function of immune cells and stem cells (SCs) residing in the tissue. In response to skin injury, hair-follicle stem cells (HFSCs), normally poised for hair generation, are recruited to the site of injury and differentiate into cells that repair damaged epithelium. We used a SC fate-mapping approach to examine the contribution of regulatory T (Treg) cells to epidermal-barrier repair after injury. Depletion of Treg cells impaired skin-barrier regeneration and was associated with a Th17 inflammatory response and failed HFSC differentiation. In this setting, damaged epithelial cells preferentially expressed the neutrophil chemoattractant CXCL5, and blockade of CXCL5 or neutrophil depletion restored barrier function and SC differentiation after epidermal injury. Thus, Treg-cell regulation of localized inflammation enables HFSC differentiation and, thereby, skin-barrier regeneration, with implications for the maintenance and repair of other barrier tissues.


Subject(s)
Cell Differentiation/physiology , Chemokine CXCL5/metabolism , Epidermis/metabolism , Hair Follicle/metabolism , Interleukin-17/metabolism , Regeneration/physiology , T-Lymphocytes, Regulatory/metabolism , Animals , Epidermal Cells/metabolism , Epithelial Cells/metabolism , Hair/metabolism , Mice , Mice, Inbred C57BL , Stem Cells/metabolism
8.
Nat Rev Mol Cell Biol ; 17(10): 643-58, 2016 10.
Article in English | MEDLINE | ID: mdl-27405257

ABSTRACT

Mammalian embryonic development is a tightly regulated process that, from a single zygote, produces a large number of cell types with hugely divergent functions. Distinct cellular differentiation programmes are facilitated by tight transcriptional and epigenetic regulation. However, the contribution of epigenetic regulation to tissue homeostasis after the completion of development is less well understood. In this Review, we explore the effects of epigenetic dysregulation on adult stem cell function. We conclude that, depending on the tissue type and the epigenetic regulator affected, the consequences range from negligible to stem cell malfunction and disruption of tissue homeostasis, which may predispose to diseases such as cancer.


Subject(s)
Adult Stem Cells/physiology , Epigenesis, Genetic , Animals , Cell Differentiation , DNA Methylation , Epidermal Cells , Epidermis/physiology , Gene Expression Regulation, Developmental , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/physiology , Regeneration
9.
Cell ; 154(2): 337-50, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23870123

ABSTRACT

Synaptic contacts are largely established during embryogenesis and are then maintained during growth. To identify molecules involved in this process, we conducted a forward genetic screen in C. elegans and identified cima-1. In cima-1 mutants, synaptic contacts are correctly established during embryogenesis, but ectopic synapses emerge during postdevelopmental growth. cima-1 encodes a solute carrier in the SLC17 family of transporters that includes sialin, a protein that when mutated in humans results in neurological disorders. cima-1 does not function in neurons but rather functions in the nearby epidermal cells to correctly position glia during postlarval growth. Our findings indicate that CIMA-1 antagonizes the FGF receptor (FGFR), and does so most likely by inhibiting FGFR's role in epidermal-glia adhesion rather than signaling. Our data suggest that epidermal-glia crosstalk, in this case mediated by a transporter and the FGF receptor, is vital to preserve embryonically derived circuit architecture during postdevelopmental growth.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Neuroglia/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Sodium-Phosphate Cotransporter Proteins, Type I/metabolism , Synapses , Animals , Body Size , Caenorhabditis elegans/embryology , Caenorhabditis elegans Proteins/genetics , Embryonic Development , Epidermal Cells , Epidermis/metabolism , Mutation , Neurites/metabolism , Sodium-Phosphate Cotransporter Proteins, Type I/genetics
10.
Nature ; 606(7912): 188-196, 2022 06.
Article in English | MEDLINE | ID: mdl-35585237

ABSTRACT

Proper ectodermal patterning during human development requires previously identified transcription factors such as GATA3 and p63, as well as positional signalling from regional mesoderm1-6. However, the mechanism by which ectoderm and mesoderm factors act to stably pattern gene expression and lineage commitment remains unclear. Here we identify the protein Gibbin, encoded by the Xia-Gibbs AT-hook DNA-binding-motif-containing 1 (AHDC1) disease gene7-9, as a key regulator of early epithelial morphogenesis. We find that enhancer- or promoter-bound Gibbin interacts with dozens of sequence-specific zinc-finger transcription factors and methyl-CpG-binding proteins to regulate the expression of mesoderm genes. The loss of Gibbin causes an increase in DNA methylation at GATA3-dependent mesodermal genes, resulting in a loss of signalling between developing dermal and epidermal cell types. Notably, Gibbin-mutant human embryonic stem-cell-derived skin organoids lack dermal maturation, resulting in p63-expressing basal cells that possess defective keratinocyte stratification. In vivo chimeric CRISPR mouse mutants reveal a spectrum of Gibbin-dependent developmental patterning defects affecting craniofacial structure, abdominal wall closure and epidermal stratification that mirror patient phenotypes. Our results indicate that the patterning phenotypes seen in Xia-Gibbs and related syndromes derive from abnormal mesoderm maturation as a result of gene-specific DNA methylation decisions.


Subject(s)
DNA-Binding Proteins , Epithelium , Gene Expression Regulation, Developmental , Mesoderm , Morphogenesis , Animals , Humans , Mice , Dermis/cytology , Dermis/embryology , Dermis/metabolism , DNA Methylation , DNA-Binding Proteins/metabolism , Ectoderm/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epidermal Cells/cytology , Epidermal Cells/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelium/embryology , GATA3 Transcription Factor , Mesoderm/metabolism , Mutation , Organoids , Trans-Activators , Transcription Factors/metabolism
11.
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
12.
Development ; 151(2)2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38265193

ABSTRACT

Basal stem cells of the epidermis continuously differentiate into keratinocytes and replenish themselves via self-renewal to maintain skin homeostasis. Numerous studies have attempted to reveal how basal cells undergo differentiation or self-renewal; however, this has been hampered by a lack of robust basal cell markers and analytical platforms that allow single-cell tracking. Here, we report that zebrafish integrin beta 4 is a useful marker for basal cell labelling, irrespective of the body region, stage and regenerative status. We employed Cre-loxP recombination in combination with live cell tracking of single basal clones in the caudal fin and investigated the embryonic origin and behaviour of basal cells during fish growth and homeostasis. Although most basal cells, including those in fins, became quiescent in the adult stage, genetic cell ablation showed that basal cells were reactivated to either self-renew or differentiate, depending on the injured cell type. Our study provides a simple and easy-to-use platform for quantitative in vivo imaging of basal stem cells at wider stages and under various conditions.


Subject(s)
Epidermis , Zebrafish , Animals , Epidermal Cells , Keratinocytes , Homeostasis
13.
Development ; 151(12)2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38934416

ABSTRACT

Transit-amplifying (TA) cells are progenitors that undergo an amplification phase followed by transition into an extinction phase. A long postulated epidermal TA progenitor with biphasic behavior has not yet been experimentally observed in vivo. Here, we identify such a TA population using clonal analysis of Aspm-CreER genetic cell-marking in mice, which uncovers contribution to both homeostasis and injury repair of adult skin. This TA population is more frequently dividing than a Dlx1-CreER-marked long-term self-renewing (e.g. stem cell) population. Newly developed generalized birth-death modeling of long-term lineage tracing data shows that both TA progenitors and stem cells display neutral competition, but only the stem cells display neutral drift. The quantitative evolution of a nascent TA cell and its direct descendants shows that TA progenitors indeed amplify the basal layer before transition and that the homeostatic TA population is mostly in extinction phase. This model will be broadly useful for analyzing progenitors whose behavior changes with their clone age. This work identifies a long-missing class of non-self-renewing biphasic epidermal TA progenitors and has broad implications for understanding tissue renewal mechanisms.


Subject(s)
Epidermal Cells , Epidermis , Stem Cells , Animals , Mice , Stem Cells/cytology , Stem Cells/metabolism , Epidermal Cells/cytology , Epidermal Cells/metabolism , Epidermis/metabolism , Cell Proliferation , Cell Lineage , Homeostasis , Cell Differentiation , Cell Self Renewal/physiology
14.
Plant Cell ; 36(3): 764-789, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38057163

ABSTRACT

Precise control over how and where actin filaments are created leads to the construction of unique cytoskeletal arrays within a common cytoplasm. Actin filament nucleators are key players in this activity and include the conserved actin-related protein 2/3 (Arp2/3) complex as well as a large family of formins. In some eukaryotic cells, these nucleators compete for a common pool of actin monomers and loss of one favors the activity of the other. To test whether this mechanism is conserved, we combined the ability to image single filament dynamics in the homeostatic cortical actin array of living Arabidopsis (Arabidopsis thaliana) epidermal cells with genetic and/or small molecule inhibitor approaches to stably or acutely disrupt nucleator activity. We found that Arp2/3 mutants or acute CK-666 treatment markedly reduced the frequency of side-branched nucleation events as well as overall actin filament abundance. We also confirmed that plant formins contribute to side-branched filament nucleation in vivo. Surprisingly, simultaneous inhibition of both classes of nucleator increased overall actin filament abundance and enhanced the frequency of de novo nucleation events by an unknown mechanism. Collectively, our findings suggest that multiple actin nucleation mechanisms cooperate to generate and maintain the homeostatic cortical array of plant epidermal cells.


Subject(s)
Actins , Arabidopsis , Actins/metabolism , Arabidopsis/genetics , Arabidopsis/metabolism , Formins/metabolism , Actin-Related Protein 2-3 Complex/genetics , Actin-Related Protein 2-3 Complex/metabolism , Actin Cytoskeleton/metabolism , Epidermal Cells/metabolism
15.
PLoS Biol ; 22(6): e3002662, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38870210

ABSTRACT

The polygonal shape of cells in proliferating epithelia is a result of the tensile forces of the cytoskeletal cortex and packing geometry set by the cell cycle. In the larval Drosophila epidermis, two cell populations, histoblasts and larval epithelial cells, compete for space as they grow on a limited body surface. They do so in the absence of cell divisions. We report a striking morphological transition of histoblasts during larval development, where they change from a tensed network configuration with straight cell outlines at the level of adherens junctions to a highly folded morphology. The apical surface of histoblasts shrinks while their growing adherens junctions fold, forming deep lobules. Volume increase of growing histoblasts is accommodated basally, compensating for the shrinking apical area. The folded geometry of apical junctions resembles elastic buckling, and we show that the imbalance between the shrinkage of the apical domain of histoblasts and the continuous growth of junctions triggers buckling. Our model is supported by laser dissections and optical tweezer experiments together with computer simulations. Our analysis pinpoints the ability of histoblasts to store mechanical energy to a much greater extent than most other epithelial cell types investigated so far, while retaining the ability to dissipate stress on the hours time scale. Finally, we propose a possible mechanism for size regulation of histoblast apical size through the lateral pressure of the epidermis, driven by the growth of cells on a limited surface. Buckling effectively compacts histoblasts at their apical plane and may serve to avoid physical harm to these adult epidermis precursors during larval life. Our work indicates that in growing nondividing cells, compressive forces, instead of tension, may drive cell morphology.


Subject(s)
Epidermis , Larva , Morphogenesis , Animals , Epidermis/metabolism , Larva/growth & development , Drosophila melanogaster/growth & development , Epidermal Cells , Epithelial Cells/cytology , Epithelial Cells/physiology , Epithelial Cells/metabolism , Biomechanical Phenomena , Adherens Junctions/metabolism , Cell Shape , Computer Simulation , Drosophila/growth & development , Models, Biological
16.
Nat Rev Mol Cell Biol ; 21(7): 359, 2020 07.
Article in English | MEDLINE | ID: mdl-32488219
17.
Cell ; 150(1): 136-50, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22770217

ABSTRACT

Sweat glands are abundant in the body and essential for thermoregulation. Like mammary glands, they originate from epidermal progenitors. However, they display few signs of cellular turnover, and whether they have stem cells and tissue-regenerative capacity remains largely unexplored. Using lineage tracing, we here identify in sweat ducts multipotent progenitors that transition to unipotency after developing the sweat gland. In characterizing four adult stem cell populations of glandular skin, we show that they display distinct regenerative capabilities and remain unipotent when healing epidermal, myoepithelial-specific, and lumenal-specific injuries. We devise purification schemes and isolate and transcriptionally profile progenitors. Exploiting molecular differences between sweat and mammary glands, we show that only some progenitors regain multipotency to produce de novo ductal and glandular structures, but that these can retain their identity even within certain foreign microenvironments. Our findings provide insight into glandular stem cells and a framework for the further study of sweat gland biology.


Subject(s)
Adult Stem Cells/cytology , Adult Stem Cells/physiology , Homeostasis , Sweat Glands/cytology , Wound Healing , Adult Stem Cells/classification , Animals , Epidermal Cells , Epidermis/physiology , Female , Humans , Mammary Glands, Animal/cytology , Mice , Morphogenesis , Multipotent Stem Cells/physiology , Principal Component Analysis , Stem Cell Transplantation , Sweat Glands/embryology , Sweat Glands/physiology
18.
PLoS Genet ; 20(4): e1011237, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38662763

ABSTRACT

An animal's skin provides a first point of contact with the sensory environment, including noxious cues that elicit protective behavioral responses. Nociceptive somatosensory neurons densely innervate and intimately interact with epidermal cells to receive these cues, however the mechanisms by which epidermal interactions shape processing of noxious inputs is still poorly understood. Here, we identify a role for dendrite intercalation between epidermal cells in tuning sensitivity of Drosophila larvae to noxious mechanical stimuli. In wild-type larvae, dendrites of nociceptive class IV da neurons intercalate between epidermal cells at apodemes, which function as body wall muscle attachment sites, but not at other sites in the epidermis. From a genetic screen we identified miR-14 as a regulator of dendrite positioning in the epidermis: miR-14 is expressed broadly in the epidermis but not in apodemes, and miR-14 inactivation leads to excessive apical dendrite intercalation between epidermal cells. We found that miR-14 regulates expression and distribution of the epidermal Innexins ogre and Inx2 and that these epidermal gap junction proteins restrict epidermal dendrite intercalation. Finally, we found that altering the extent of epidermal dendrite intercalation had corresponding effects on nociception: increasing epidermal intercalation sensitized larvae to noxious mechanical inputs and increased mechanically evoked calcium responses in nociceptive neurons, whereas reducing epidermal dendrite intercalation had the opposite effects. Altogether, these studies identify epidermal dendrite intercalation as a mechanism for mechanical coupling of nociceptive neurons to the epidermis, with nociceptive sensitivity tuned by the extent of intercalation.


Subject(s)
Connexins , Dendrites , Drosophila Proteins , Epidermis , Larva , MicroRNAs , Nociceptors , Animals , Larva/genetics , Dendrites/metabolism , Dendrites/physiology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Nociceptors/metabolism , Epidermis/metabolism , Drosophila melanogaster/genetics , Epidermal Cells/metabolism , Nociception/physiology , Drosophila/genetics
19.
Genes Dev ; 33(1-2): 55-60, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30567998

ABSTRACT

Polycomb-repressive complex 1 (PRC1) and PRC2 are critical chromatin regulators of gene expression and tissue development. Here, we show that despite extensive genomic cobinding, PRC1 is essential for epidermal integrity, whereas PRC2 is dispensable. Loss of PRC1 resulted in blistering skin, reminiscent of human skin fragility syndromes. Conversely, PRC1 does not restrict epidermal stratification during skin morphogenesis, whereas PRC2 does. Molecular dissection demonstrated that PRC1 functions with PRC2 to silence/dampen expression of adhesion genes. In contrast, PRC1 promotes expression of critical epidermal adhesion genes independently of PRC2-mediated H3K27me3. Together, we demonstrate a functional link between epigenetic regulation and skin diseases.


Subject(s)
Epidermal Cells/physiology , Epidermis/physiology , Epigenesis, Genetic , Gene Expression Regulation, Developmental/genetics , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 2/metabolism , Skin Diseases/genetics , Animals , Cell Adhesion/genetics , Epidermis/growth & development , Histones/metabolism , Mice , Polycomb Repressive Complex 1/genetics , Skin Diseases/physiopathology
20.
EMBO J ; 41(18): e110488, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35949182

ABSTRACT

Adulte interfollicular epidermis (IFE) renewal is likely orchestrated by physiological demands of its complex tissue architecture comprising spatial and cellular heterogeneity. Mouse tail and back skin display two kinds of basal IFE spatial domains that regenerate at different rates. Here, we elucidate the molecular and cellular states of basal IFE domains by marker expression and single-cell transcriptomics in mouse and human skin. We uncover two paths of basal cell differentiation that in part reflect the IFE spatial domain organization. We unravel previously unrecognized similarities between mouse tail IFE basal domains defined as scales and interscales versus human rete ridges and inter-ridges, respectively. Furthermore, our basal IFE transcriptomics and gene targeting in mice provide evidence supporting a physiological role of IFE domains in adaptation to differential UV exposure. We identify Sox6 as a novel UV-induced and interscale/inter-ridge preferred basal IFE-domain transcription factor, important for IFE proliferation and survival. The spatial, cellular, and molecular organization of IFE basal domains underscores skin adaptation to environmental exposure and its unusual robustness in adult homeostasis.


Subject(s)
Epidermal Cells , Epidermis , Adult , Animals , Cell Differentiation/genetics , Environmental Exposure , Humans , Mice , Skin
SELECTION OF CITATIONS
SEARCH DETAIL