Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.637
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 ; 175(6): 1575-1590.e22, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30415840

ABSTRACT

During aging, stromal functions are thought to be impaired, but little is known whether this stems from changes of fibroblasts. Using population- and single-cell transcriptomics, as well as long-term lineage tracing, we studied whether murine dermal fibroblasts are altered during physiological aging under different dietary regimes that affect longevity. We show that the identity of old fibroblasts becomes undefined, with the fibroblast states present in young skin no longer clearly demarcated. In addition, old fibroblasts not only reduce the expression of genes involved in the formation of the extracellular matrix, but also gain adipogenic traits, paradoxically becoming more similar to neonatal pro-adipogenic fibroblasts. These alterations are sensitive to systemic metabolic changes: long-term caloric restriction reversibly prevents them, whereas a high-fat diet potentiates them. Our results therefore highlight loss of cell identity and the acquisition of adipogenic traits as a mechanism underlying cellular aging, which is influenced by systemic metabolism.


Subject(s)
Adipogenesis , Cellular Senescence , Fibroblasts/metabolism , Skin Aging , Animals , Caloric Restriction , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Mice , Mice, Transgenic
3.
Genes Dev ; 2022 Aug 25.
Article in English | MEDLINE | ID: mdl-36008138

ABSTRACT

Stem cells are fundamental units of tissue remodeling whose functions are dictated by lineage-specific transcription factors. Home to epidermal stem cells and their upward-stratifying progenies, skin relies on its secretory functions to form the outermost protective barrier, of which a transcriptional orchestrator has been elusive. KLF5 is a Krüppel-like transcription factor broadly involved in development and regeneration whose lineage specificity, if any, remains unclear. Here we report KLF5 specifically marks the epidermis, and its deletion leads to skin barrier dysfunction in vivo. Lipid envelopes and secretory lamellar bodies are defective in KLF5-deficient skin, accompanied by preferential loss of complex sphingolipids. KLF5 binds to and transcriptionally regulates genes encoding rate-limiting sphingolipid metabolism enzymes. Remarkably, skin barrier defects elicited by KLF5 ablation can be rescued by dietary interventions. Finally, we found that KLF5 is widely suppressed in human diseases with disrupted epidermal secretion, and its regulation of sphingolipid metabolism is conserved in human skin. Altogether, we established KLF5 as a disease-relevant transcription factor governing sphingolipid metabolism and barrier function in the skin, likely representing a long-sought secretory lineage-defining factor across tissue types.

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.
Genes Dev ; 35(5-6): 301-303, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33649160

ABSTRACT

The Polycomb repressive system functions through chromatin to regulate gene expression and development. In this issue of Genes & Development, Cohen and colleagues (pp. 354-366) use the developing mouse epidermis as a model system to show that the two central Polycomb repressive complexes, PRC1 and PRC2, have autonomous yet overlapping functions in repressing Polycomb target genes. They show that this cooperation enables the stable repression of nonepidermal transcription factors that would otherwise compromise epidermal cell identity and disrupt normal skin development.


Subject(s)
Epidermis/growth & development , Gene Expression Regulation, Developmental/genetics , Polycomb-Group Proteins/metabolism , Transcription Factors/genetics , Animals , Mice
6.
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
7.
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
8.
Proc Natl Acad Sci U S A ; 121(2): e2316396121, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38165937

ABSTRACT

Plant epidermal cell walls maintain the mechanical integrity of plants and restrict organ growth. Mechanical analyses can give insights into wall structure and are inputs for mechanobiology models of plant growth. To better understand the intrinsic mechanics of epidermal cell walls and how they may accommodate large deformations during growth, we analyzed a geometrically simple material, onion epidermal strips consisting of only the outer (periclinal) cell wall, ~7 µm thick. With uniaxial stretching by >40%, the wall showed complex three-phase stress-strain responses while cyclic stretching revealed reversible and irreversible deformations and elastic hysteresis. Stretching at varying strain rates and temperatures indicated the wall behaved more like a network of flexible cellulose fibers capable of sliding than a viscoelastic composite with pectin viscosity. We developed an analytic framework to quantify nonlinear wall mechanics in terms of stiffness, deformation, and energy dissipation, finding that the wall stretches by combined elastic and plastic deformation without compromising its stiffness. We also analyzed mechanical changes in slightly dehydrated walls. Their extension became stiffer and more irreversible, highlighting the influence of water on cellulose stiffness and sliding. This study offers insights into the structure and deformation modes of primary cell walls and presents a framework that is also applicable to tissues and whole organs.


Subject(s)
Cell Wall , Cellulose , Cellulose/chemistry , Cell Wall/chemistry , Cell Membrane , Pectins , Plant Epidermis
9.
Genes Dev ; 33(1-2): 4-5, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30602437

ABSTRACT

All of the cells in our body share largely identical DNA, yet functionally distinct cells are generated to give rise to different tissues and organs. A fundamental question in biology is how different cell fates are specified and maintained. Epigenetic mechanisms hold a key answer to the question. Without changing the sequence of DNA but through modifying DNA, histones, or RNA, epigenetic mechanisms can decide which genes to express and which to suppress. Polycomb group (PcG) proteins are a group of evolutionarily conserved proteins that can regulate gene expression through histone modification. Although PcG proteins have been traditionally described as epigenetic repressors, emerging evidence suggests a more complex scenario in which PcG proteins can have a dynamic effect on gene expression. In this issue of Genes & Development, Cohen and colleagues (pp. 55-60) studied the function of Polycomb-repressive complex 1 (PRC1) in mouse skin development and identified PRC1's unique function independent of PRC2. Notably, the total loss of PRC1 but not canonical PRC1 in the skin leads to widespread down-regulation of genes involved in cell adhesion and cytoskeleton organization, resulting in skin fragility. This new study lays a foundation to examine the role of PRC1 in activating gene expression.


Subject(s)
Histones , Polycomb Repressive Complex 1 , Animals , Cell Nucleus , Epigenesis, Genetic , Mice , Polycomb-Group Proteins
10.
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
11.
Development ; 150(17)2023 09 01.
Article in English | MEDLINE | ID: mdl-37622728

ABSTRACT

The planar cell polarity (PCP) pathway collectively orients cells with respect to a body axis. Hair follicles of the murine epidermis provide a striking readout of PCP activity in their uniform alignment across the skin. Here, we characterize, from the molecular to tissue-scale, PCP establishment in the rosette fancy mouse, a natural variant with posterior-specific whorls in its fur, to understand how epidermal polarity is coordinated across the tissue. We find that rosette hair follicles emerge with reversed orientations specifically in the posterior region, creating a mirror image of epidermal polarity. The rosette trait is associated with a missense mutation in the core PCP gene Fzd6, which alters a consensus site for N-linked glycosylation, inhibiting its membrane localization. Unexpectedly, the Fzd6 trafficking defect does not block asymmetric localization of the other PCP proteins. Rather, the normally uniform axis of PCP asymmetry rotates where the PCP-directed cell movements that orient follicles are reversed, suggesting the PCP axis rotates 180°. Collectively, our multiscale analysis of epidermal polarity reveals PCP patterning can be regionally decoupled to produce posterior whorls in the rosette fancy mouse.


Subject(s)
Epidermis , Hair Follicle , Animals , Mice , Skin , Epidermal Cells , Cell Movement
12.
J Biol Chem ; 300(7): 107449, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38844132

ABSTRACT

Hyaluronan (HA) is a high-molecular-weight (HMW) glycosaminoglycan, which is a fundamental component of the extracellular matrix that is involved in a variety of biological processes. We previously showed that the HYBID/KIAA1199/CEMIP axis plays a key role in the depolymerization of HMW-HA in normal human dermal fibroblasts (NHDFs). However, its roles in normal human epidermal keratinocytes (NHEKs) remained unclear. HYBID mRNA expression in NHEKs was lower than that in NHDFs, and NHEKs showed no depolymerization of extracellular HMW-HA in culture, indicating that HYBID does not contribute to extracellular HA degradation. In this study, we found that the cell-free conditioned medium of NHEKs degraded HMW-HA under weakly acidic conditions (pH 4.8). This degrading activity was abolished by hyaluronidase 1 (HYAL1) knockdown but not by HYAL2 knockdown. Newly synthesized HYAL1 was mainly secreted extracellularly, and the secretion of HYAL1 was increased during differentiation, suggesting that epidermal interspace HA is physiologically degraded by HYAL1 according to pH decrease during stratum corneum formation. In HA synthesis, hyaluronan synthase 3 (HAS3) knockdown reduced HA production by NHEKs, and interferon-γ-dependent HA synthesis was correlated with increased HAS3 expression. Furthermore, HA production was increased by TMEM2 knockdown through enhanced HAS3 expression. These results indicate that NHEKs regulate HA metabolism via HYAL1 and HAS3, and TMEM2 is a regulator of HAS3-dependent HA production.

13.
EMBO J ; 40(12): e106787, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33998017

ABSTRACT

Eukaryotic development relies on dynamic cell shape changes and segregation of fate determinants to achieve coordinated compartmentalization at larger scale. Studies in invertebrates have identified polarity programmes essential for morphogenesis; however, less is known about their contribution to adult tissue maintenance. While polarity-dependent fate decisions in mammals utilize molecular machineries similar to invertebrates, the hierarchies and effectors can differ widely. Recent studies in epithelial systems disclosed an intriguing interplay of polarity proteins, adhesion molecules and mechanochemical pathways in tissue organization. Based on major advances in biophysics, genome editing, high-resolution imaging and mathematical modelling, the cell polarity field has evolved to a remarkably multidisciplinary ground. Here, we review emerging concepts how polarity and cell fate are coupled, with emphasis on tissue-scale mechanisms, mechanobiology and mammalian models. Recent findings on the role of polarity signalling for tissue mechanics, micro-environmental functions and fate choices in health and disease will be summarized.


Subject(s)
Cell Polarity , Animals , Biomechanical Phenomena , Homeostasis , Humans , Neoplasms , Regeneration
14.
J Cell Sci ; 136(12)2023 06 15.
Article in English | MEDLINE | ID: mdl-37259855

ABSTRACT

The mammalian epidermis undergoes constant renewal, replenished by a pool of stem cells and terminal differentiation of their progeny. This is accompanied by changes in gene expression and morphology that are orchestrated, in part, by epigenetic modifiers. Here, we define the role of the histone acetyltransferase KAT2A in epidermal homeostasis and provide a comparative analysis that reveals key functional divergence with its paralog KAT2B. In contrast to the reported function of KAT2B in epidermal differentiation, KAT2A supports the undifferentiated state in keratinocytes. RNA-seq analysis of KAT2A- and KAT2B- depleted keratinocytes revealed dysregulated epidermal differentiation. Depletion of KAT2A led to premature expression of epidermal differentiation genes in the absence of inductive signals, whereas loss of KAT2B delayed differentiation. KAT2A acetyltransferase activity was indispensable in regulating epidermal differentiation gene expression. The metazoan-specific N terminus of KAT2A was also required to support its function in keratinocytes. We further showed that the interplay between KAT2A- and KAT2B-mediated regulation was important for normal cutaneous wound healing in vivo. Overall, these findings reveal a distinct mechanism in which keratinocytes use a pair of highly homologous histone acetyltransferases to support divergent functions in self-renewal and differentiation processes.


Subject(s)
Histone Acetyltransferases , Keratinocytes , Animals , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Keratinocytes/metabolism , Cell Differentiation/genetics , Skin/metabolism , Epidermis/metabolism , Mammals/metabolism
15.
Development ; 149(14)2022 07 15.
Article in English | MEDLINE | ID: mdl-35815643

ABSTRACT

The barrier-forming, self-renewing mammalian epidermis comprises keratinocytes, pigment-producing melanocytes and resident immune cells as first-line host defense. In murine tail skin, interfollicular epidermis patterns into pigmented 'scale' and hypopigmented 'interscale' epidermis. Why and how mature melanocytes accumulate in scale epidermis is unresolved. Here, we delineate a cellular hierarchy among epidermal cell types that determines skin patterning. Already during postnatal development, melanocytes co-segregate with newly forming scale compartments. Intriguingly, this process coincides with partitioning of both Langerhans cells and dendritic epidermal T cells to interscale epidermis, suggesting functional segregation of pigmentation and immune surveillance. Analysis of non-pigmented mice and of mice lacking melanocytes or resident immune cells revealed that immunocyte patterning is melanocyte and melanin independent and, vice versa, immune cells do not control melanocyte localization. Instead, genetically enforced progressive scale fusion upon Lrig1 deletion showed that melanocytes and immune cells dynamically follow epithelial scale:interscale patterns. Importantly, disrupting Wnt-Lef1 function in keratinocytes caused melanocyte mislocalization to interscale epidermis, implicating canonical Wnt signaling in organizing the pigmentation pattern. Together, this work uncovers cellular and molecular principles underlying the compartmentalization of tissue functions in skin.


Subject(s)
Epidermis , Tail , Animals , Epidermal Cells/metabolism , Epidermis/metabolism , Keratinocytes/metabolism , Mammals/metabolism , Melanins/metabolism , Melanocytes/metabolism , Membrane Glycoproteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , Skin Pigmentation , Tail/metabolism
16.
Development ; 149(6)2022 03 15.
Article in English | MEDLINE | ID: mdl-35299238

ABSTRACT

The maintenance of epithelial architecture necessitates tight regulation of cell size and shape. However, mechanisms underlying epithelial cell size regulation remain poorly understood. We show that the interaction of Myosin Vb with Rab11 prevents the accumulation of apically derived endosomes to maintain cell-size, whereas that with Rab10 regulates vesicular transport from the trans-Golgi. These interactions are required for the fine-tuning of the epithelial cell morphology during zebrafish development. Furthermore, the compensatory cell growth upon cell-proliferation inhibition involves a preferential expansion of the apical domain, leading to flatter epithelial cells, an efficient strategy to cover the surface with fewer cells. This apical domain growth requires post-trans-Golgi transport mediated by the Rab10-interacting Myosin Vb isoform, downstream of the mTOR-Fatty Acid Synthase (FASN) axis. Changes in trans-Golgi morphology indicate that the Golgi synchronizes mTOR-FASN-regulated biosynthetic input and Myosin Vb-Rab10 dependent output. Our study unravels the mechanism of polarized growth in epithelial cells and delineates functions of Myosin Vb isoforms in cell size regulation during development.


Subject(s)
Myosin Type V , Animals , Epithelial Cells/metabolism , Myosin Type V/metabolism , Protein Isoforms/metabolism , TOR Serine-Threonine Kinases/metabolism , Zebrafish/metabolism , rab GTP-Binding Proteins/metabolism
17.
Stem Cells ; 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38301639

ABSTRACT

Loss of sensory innervation delays wound healing and administration of the neuropeptide substance P improves re-epithelialization. Keratinocyte hyperproliferation post-wounding may result from symmetric stem cell (SC) self-renewal, asymmetric SC self-renewal, committed progenitor divisions, or a combination of these. However, the effects of sensory denervation and of neuropeptides on SC proliferation are not known. Here we show that early after wounding both asymmetric and symmetric SC self-renewal increase, without significant committed progenitor (CP) activation. Decreased sensory innervation is associated with a decrease in both SC and CP proliferation. Based on previous work showing that substance P is decreased in capsaicin-treated mice and improves wound healing in normal skin, we examined the effects of substance P on SC and CP proliferation during wound healing. Substance P restored asymmetric SC proliferation in skin with decreased sensory innervation, both at baseline and following wounding. Epidermis with decreased sensory innervation was severely thinned. Consistent with this, substance P-induced asymmetric SC proliferation resulted in increased stratification in skin with both normal and decreased innervation. Lapatinib prevented the substance P-induced increase in asymmetric SC divisions in murine epidermis, as well as the increase in epidermal stratification, suggesting that asymmetric SC divisions are required for epidermal stratification.

18.
FASEB J ; 38(4): e23476, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38334392

ABSTRACT

The prevalence of alopecia has increased recently. Hair loss is often accompanied by the resting phase of hair follicles (HFs). Dermal papilla (DP) plays a crucial role in HF development, growth, and regeneration. Activating DP can revive resting HFs. Augmenting WNT/ß-catenin signaling stimulates HF growth. However, the factors responsible for activating resting HFs effectively are unclear. In this study, we investigated epidermal cytokines that can activate resting HFs effectively. We overexpressed ß-catenin in both in vivo and in vitro models to observe its effects on resting HFs. Then, we screened potential epidermal cytokines from GEO DATASETs and assessed their functions using mice models and skin-derived precursors (SKPs). Finally, we explored the molecular mechanism underlying the action of the identified cytokine. The results showed that activation of WNT/ß-catenin in the epidermis prompted telogen-anagen transition. Keratinocytes infected with Ctnnb1-overexpressing lentivirus enhanced SKP expansion. Subsequently, we identified endothelin 1 (ET-1) expressed higher in hair-growing epidermis and induced the proliferation of DP cells and activates telogen-phase HFs in vivo. Moreover, ET-1 promotes the proliferation and stemness of SKPs. Western blot analysis and in vivo experiments revealed that ET-1 induces the transition from telogen-to-anagen phase by upregulating the PI3K/AKT pathway. These findings highlight the potential of ET-1 as a promising cytokine for HF activation and the treatment of hair loss.


Subject(s)
Hair Follicle , Proto-Oncogene Proteins c-akt , Animals , Mice , Hair Follicle/metabolism , Proto-Oncogene Proteins c-akt/metabolism , beta Catenin/genetics , beta Catenin/metabolism , Endothelin-1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Cells, Cultured , Cell Proliferation , Epidermis/metabolism , Alopecia/metabolism , Wnt Signaling Pathway , Dermis/metabolism , Cytokines/metabolism
19.
EMBO Rep ; 24(7): e56214, 2023 Jul 05.
Article in English | MEDLINE | ID: mdl-37249012

ABSTRACT

Skin epidermis constitutes the outer permeability barrier that protects the body from dehydration, heat loss, and myriad external assaults. Mechanisms that maintain barrier integrity in constantly challenged adult skin and how epidermal dysregulation shapes the local immune microenvironment and whole-body metabolism remain poorly understood. Here, we demonstrate that inducible and simultaneous ablation of transcription factor-encoding Ovol1 and Ovol2 in adult epidermis results in barrier dysregulation through impacting epithelial-mesenchymal plasticity and inflammatory gene expression. We find that aberrant skin immune activation then ensues, featuring Langerhans cell mobilization and T cell responses, and leading to elevated levels of secreted inflammatory factors in circulation. Finally, we identify failure to gain body weight and accumulate body fat as long-term consequences of epidermal-specific Ovol1/2 loss and show that these global metabolic changes along with the skin barrier/immune defects are partially rescued by immunosuppressant dexamethasone. Collectively, our study reveals key regulators of adult barrier maintenance and suggests a causal connection between epidermal dysregulation and whole-body metabolism that is in part mediated through aberrant immune activation.


Subject(s)
DNA-Binding Proteins , Epidermis , DNA-Binding Proteins/genetics , Epidermis/metabolism , Skin/metabolism , Transcription Factors/metabolism , Epidermal Cells/metabolism
20.
Proc Natl Acad Sci U S A ; 119(25): e2116027119, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35704759

ABSTRACT

The epidermis is the outermost layer of the skin and the body's primary barrier to external pathogens; however, the early epidermal immune response remains to be mechanistically understood. We show that the chemokine CXCL14, produced by epidermal keratinocytes, exhibits robust circadian fluctuations and initiates innate immunity. Clearance of the skin pathogen Staphylococcus aureus in nocturnal mice was associated with CXCL14 expression, which was high during subjective daytime and low at night. In contrast, in marmosets, a diurnal primate, circadian CXCL14 expression was reversed. Rhythmically expressed CXCL14 binds to S. aureus DNA and induces inflammatory cytokine production by activating Toll-like receptor (TLR)9-dependent innate pathways in dendritic cells and macrophages underneath the epidermis. CXCL14 also promoted phagocytosis by macrophages in a TLR9-independent manner. These data indicate that circadian production of the epidermal chemokine CXCL14 rhythmically suppresses skin bacterial proliferation in mammals by activating the innate immune system.


Subject(s)
Epidermis , Immunity, Innate , Skin Diseases, Bacterial , Animals , Chemokines, CXC/genetics , Chemokines, CXC/immunology , Circadian Clocks/immunology , Epidermis/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Keratinocytes/immunology , Mammals , Mice , Skin Diseases, Bacterial/immunology , Skin Diseases, Bacterial/metabolism , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL