Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 24
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Front Physiol ; 15: 1367425, 2024.
Article de Anglais | MEDLINE | ID: mdl-38434140

RÉSUMÉ

Epithelial tissues, including the skin, are highly proliferative tissues with the capability to constant renewal and regeneration, a feature that is essential for survival as the skin forms a protective barrier against external insults and water loss. In adult mammalian skin, every injury will lead to a scar. The scar tissue that is produced to seal the wound efficiently is usually rigid and lacks elasticity and the skin's original resilience to external impacts, but also secondary appendages such as hair follicles and sebaceous glands. While it was long thought that hair follicles develop solely during embryogenesis, it is becoming increasingly clear that hair follicles can also regenerate within a wound. The ability of the skin to induce hair neogenesis following injury however declines with age. As fetal and neonatal skin have the remarkable capacity to heal without scarring, the recapitulation of a neonatal state has been a primary target of recent regenerative research. In this review we highlight how modulating dermal signaling or the abundance of specific fibroblast subsets could be utilized to induce de novo hair follicles within the wound bed, and thus to shift wound repair with a scar to scarless regeneration.

2.
J Invest Dermatol ; 143(9): 1667-1677, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37612031

RÉSUMÉ

Single-cell technologies have become essential to driving discovery in both basic and translational investigative dermatology. Despite the multitude of available datasets, a central reference atlas of normal human skin, which can serve as a reference resource for skin cell types, cell states, and their molecular signatures, is still lacking. For any such atlas to receive broad acceptance, participation by many investigators during atlas construction is an essential prerequisite. As part of the Human Cell Atlas project, we have assembled a Skin Biological Network to build a consensus Human Skin Cell Atlas and outline a roadmap toward that goal. We define the drivers of skin diversity to be considered when selecting sequencing datasets for the atlas and list practical hurdles during skin sampling that can result in data gaps and impede comprehensive representation and technical considerations for tissue processing and computational analysis, the accounting for which should minimize biases in cell type enrichments and exclusions and decrease batch effects. By outlining our goals for Atlas 1.0, we discuss how it will uncover new aspects of skin biology.


Sujet(s)
Personnel de recherche , Peau , Humains , Consensus
3.
Dev Cell ; 58(20): 2140-2162.e5, 2023 10 23.
Article de Anglais | MEDLINE | ID: mdl-37591247

RÉSUMÉ

A wealth of specialized cell populations within the skin facilitates its hair-producing, protective, sensory, and thermoregulatory functions. How the vast cell-type diversity and tissue architecture develops is largely unexplored. Here, with single-cell transcriptomics, spatial cell-type assignment, and cell-lineage tracing, we deconstruct early embryonic mouse skin during the key transitions from seemingly uniform developmental precursor states to a multilayered, multilineage epithelium, and complex dermal identity. We identify the spatiotemporal emergence of hair-follicle-inducing, muscle-supportive, and fascia-forming fibroblasts. We also demonstrate the formation of the panniculus carnosus muscle (PCM), sprouting blood vessels without pericyte coverage, and the earliest residence of mast and dendritic immune cells in skin. Finally, we identify an unexpected epithelial heterogeneity within the early single-layered epidermis and a signaling-rich periderm layer. Overall, this cellular and molecular blueprint of early skin development-which can be explored at https://kasperlab.org/tools-establishes histological landmarks and highlights unprecedented dynamic interactions among skin cells.


Sujet(s)
Épiderme , Peau , Souris , Animaux , Follicule pileux/anatomopathologie , Poils , Épithélium
4.
J Invest Dermatol ; 142(6): 1737-1748.e5, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-34922948

RÉSUMÉ

Patients suffering from large scars such as burn victims not only encounter aesthetic challenges but also ongoing itching or pain that substantially deteriorates their quality of life. Skin appendages such as hair follicles rarely regenerate within the healing wound. Because they are crucial for skin homeostasis and the lack thereof constitutes one of the main limitations to scarless wound healing, their regeneration represents a major objective for regenerative medicine. Fibroblasts, the main resident cell type of the skin dermis, mediate embryonic hair follicle morphogenesis and are particularly involved in wound healing because they orchestrate extracellular matrix remodeling and collagen deposition in the wound bed. Importantly, dermal fibroblasts originate from two distinct developmental lineages with unique functions that differently mediate the response to epidermal signals such as Hedgehog signaling. In this study, we show that Hedgehog signaling in the reticular fibroblast lineage promotes the initial phase of wound repair, possibly by modulating angiogenesis and fibroblast proliferation, whereas Hedgehog signaling in papillary fibroblasts is essential to induce de novo hair follicle formation within the healing wound.


Sujet(s)
Follicule pileux , Protéines Hedgehog , Régénération , Transduction du signal , Cicatrisation de plaie , Derme/métabolisme , Fibroblastes/métabolisme , Follicule pileux/croissance et développement , Protéines Hedgehog/physiologie , Humains , Qualité de vie , Régénération/physiologie , Cicatrisation de plaie/physiologie
5.
Am J Transplant ; 21(2): 525-539, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33063442

RÉSUMÉ

Epidemiological and experimental data implicate cutaneous human papillomavirus infection as co-factor in the development of cutaneous squamous cell carcinomas (cSCCs), particularly in immunocompromised organ transplant recipients (OTRs). Herein, we established and characterized a skin cancer model, in which Mus musculus papillomavirus 1 (MmuPV1) infection caused cSCCs in cyclosporine A (CsA)-treated mice, even in the absence of UV light. Development of cSCCs and their precursors were observed in 70% of MmuPV1-infected, CsA-treated mice on back as well as on tail skin. Immunosuppression by systemic CsA, but not UV-B irradiation, was a prerequisite, as immunocompetent or UV-B-irradiated mice did not develop skin malignancies after infection. In the virus-driven cSCCs the MmuPV1-E6/E7 oncogenes were abundantly expressed, and transcriptional activity and productive infection demonstrated. MmuPV1 infection induced the expression of phosphorylated H2AX, but not degradation of proapoptotic BAK in the cSCCs. Transfer of primary cells, established from a MmuPV1-induced cSCC from back skin, into athymic nude mice gave rise to secondary cSCCs, which lacked viral DNA, demonstrating that maintenance of the malignant phenotype was virus independent. This papillomavirus-induced skin cancer model opens future investigations into viral involvement, pathogenesis, and cancer surveillance, aiming at understanding and controlling the high incidence of skin cancer in OTRs.


Sujet(s)
Infections à papillomavirus , Tumeurs cutanées , Animaux , Immunosuppression thérapeutique , Souris , Souris nude , Papillomaviridae , Tumeurs cutanées/induit chimiquement
7.
J Invest Dermatol ; 140(4): 878-890.e5, 2020 04.
Article de Anglais | MEDLINE | ID: mdl-31622599

RÉSUMÉ

Melanoma cells can switch between distinct gene expression profiles, resulting in proliferative or invasive phenotypes. Signaling pathways involved in this switch were analyzed by gene expression profiling of a cohort of 22 patient-derived melanoma cell lines. CDH1 negativity was identified as a surrogate marker for the invasive phenotype. CDH1 expression could be turned on and off by modulating activity of p38 or its downstream target MK2, suggesting that this pathway controls melanoma progression. Mechanistically, MK2 inhibition prevented melanoma-induced vascular barrier disruption, reduced the expression of PODXL and DEL-1, and prevented vascular dissemination in vivo. PODXL and DEL-1 expression in patients with melanoma were associated with poor survival and thus can be used as prognostic markers. Downstream targets of MK2 may thus serve as candidate therapeutics.


Sujet(s)
Régulation de l'expression des gènes tumoraux , Mélanome/génétique , Tumeurs cutanées/génétique , Tumeurs vasculaires/prévention et contrôle , p38 Mitogen-Activated Protein Kinases/antagonistes et inhibiteurs , Humains , Mélanome/métabolisme , Mélanome/anatomopathologie , Invasion tumorale , Pronostic , Transduction du signal , Tumeurs cutanées/métabolisme , Tumeurs cutanées/anatomopathologie , Cellules cancéreuses en culture , Tumeurs vasculaires/métabolisme , Tumeurs vasculaires/anatomopathologie , p38 Mitogen-Activated Protein Kinases/biosynthèse , p38 Mitogen-Activated Protein Kinases/génétique
8.
iScience ; 15: 243-256, 2019 May 31.
Article de Anglais | MEDLINE | ID: mdl-31082735

RÉSUMÉ

Epidermal growth factor receptor (EGFR) signaling controls skin development and homeostasis in mice and humans, and its deficiency causes severe skin inflammation, which might affect epidermal stem cell behavior. Here, we describe the inflammation-independent effects of EGFR deficiency during skin morphogenesis and in adult hair follicle stem cells. Expression and alternative splicing analysis of RNA sequencing data from interfollicular epidermis and outer root sheath indicate that EGFR controls genes involved in epidermal differentiation and also in centrosome function, DNA damage, cell cycle, and apoptosis. Genetic experiments employing p53 deletion in EGFR-deficient epidermis reveal that EGFR signaling exhibits p53-dependent functions in proliferative epidermal compartments, as well as p53-independent functions in differentiated hair shaft keratinocytes. Loss of EGFR leads to absence of LEF1 protein specifically in the innermost epithelial hair layers, resulting in disorganization of medulla cells. Thus, our results uncover important spatial and temporal features of cell-autonomous EGFR functions in the epidermis.

10.
J Vis Exp ; (147)2019 05 07.
Article de Anglais | MEDLINE | ID: mdl-31132050

RÉSUMÉ

Fibroblasts are a highly heterogeneous cell population implicated in the pathogenesis of many human diseases. In human skin dermis, fibroblasts have traditionally been attributed to the superficial papillary or lower reticular dermis according to their histological localization. In mouse dermis, papillary and reticular fibroblasts originate from two different lineages with diverging functions regarding physiological and pathological processes and a distinct cell surface marker expression profile by which they can be distinguished. Importantly, evidence from explant cultures from superficial and lower dermal layers suggest that at least two functionally distinct dermal fibroblasts lineages exist in human skin dermis as well. However, unlike for mouse skin, cell surface markers enabling the discrimination of different fibroblast subsets have not yet been established for human skin. We developed a novel protocol for the isolation of human papillary and reticular fibroblast populations via fluorescence-activated cell sorting (FACS) using the two cell surface markers Fibroblast Activation Protein (FAP) and Thymocyte antigen 1 (Thy1)/CD90. This method enables the isolation of pure fibroblast subsets without in vitro manipulation, which was shown to affect gene expression, thus permitting accurate functional analysis of human dermal fibroblast subsets in regard to tissue homeostasis or disease pathology.


Sujet(s)
Fibroblastes/métabolisme , Cytométrie en flux/méthodes , Peau/métabolisme , Cellules cultivées , Humains , Peau/cytologie
11.
J Invest Dermatol ; 139(2): 342-351, 2019 02.
Article de Anglais | MEDLINE | ID: mdl-30179601

RÉSUMÉ

Human skin dermis is composed of the superficial papillary dermis and the reticular dermis in the lower layers, which can easily be distinguished histologically. In vitro analyses of fibroblasts from explant cultures from superficial and lower dermal layers suggest that human skin comprises at least two fibroblast lineages with distinct morphology, expression profiles, and functions. However, while for mouse skin cell surface markers have been identified, allowing the isolation of pure populations of one lineage or the other via FACS, this has not been achieved for human skin fibroblasts. We have now discovered two cell surface markers that discriminate between papillary and reticular fibroblasts. While FAP+CD90- cells display increased proliferative potential, express PDPN and NTN1, and cannot be differentiated into adipocytes, FAP-CD90+ fibroblasts express high levels of ACTA2, MGP, PPARγ, and CD36 and readily undergo adipogenic differentiation, a hallmark of reticular fibroblasts. Flow cytometric analysis of fibroblasts isolated from superficial and lower layers of human dermis showed that FAP+CD90- cells are enriched in the papillary dermis. Altogether, functional analysis and expression profiling confirms that FAP+CD90- cells represent papillary fibroblasts, whereas FAP-CD90+ fibroblasts derive from the reticular lineage. Although papillary and reticular fibroblasts are enriched in the upper or lower dermis, respectively, they are not spatially restricted, and the microenvironment seems to affect their function.


Sujet(s)
Différenciation cellulaire , Derme/cytologie , Fibroblastes/physiologie , Adipocytes/physiologie , Adulte , Marqueurs biologiques/métabolisme , Séparation cellulaire , Cellules cultivées , Endopeptidases , Femelle , Cytométrie en flux , Gelatinases/métabolisme , Humains , Mâle , Protéines membranaires/métabolisme , Adulte d'âge moyen , Culture de cellules primaires , Serine endopeptidases/métabolisme , Antigènes Thy-1/métabolisme
12.
J Invest Dermatol ; 139(1): 186-194, 2019 01.
Article de Anglais | MEDLINE | ID: mdl-30009831

RÉSUMÉ

Chemokines mold the tumor microenvironment by recruiting distinct immune cell populations, thereby strongly influencing disease progression. Previously, we showed that CXCL5 expression is upregulated in advanced stages of primary melanomas, which correlates with the presence of neutrophils in the tumor. The analysis of neutrophil populations in various tissues revealed a distinct phenotype of tumor-associated neutrophils. Tumor-associated neutrophils expressed PD-L1, CXCR4, CCR5, Adam17, and Nos2 and were immunosuppressive in a T-cell proliferation assay. To investigate the impact of CXCL5 and neutrophils in vivo, we established a syngeneic mouse tumor transplantation model using CXCL5-overexpressing and control melanoma cell lines. Growth behavior or vascularization of primary tumors was not affected by CXCL5 expression and neutrophils alone. However, in combination with Poly(I:C), tumor-associated neutrophils were able to attenuate induced antitumoral T-cell responses. CXCL5-overexpressing tumors had reduced lung metastasis compared with control tumors. Neutrophil depletion reversed this effect. In vitro, unstimulated lung-derived neutrophils had higher levels of reactive oxygen species compared with tumor-associated neutrophils, and CXCL5 stimulation further increased reactive oxygen species levels. In summary, in melanoma, neutrophils play a context-dependent role that is influenced by local or systemic factors, and interfere with therapies activating the acquired immune system. Actively switching neutrophils into antitumorigenic mode might be a new therapeutic strategy.


Sujet(s)
Chimiokine CXCL5/génétique , ADN tumoral/génétique , Régulation de l'expression des gènes tumoraux , Mélanome/génétique , Activation des neutrophiles/génétique , Granulocytes neutrophiles/métabolisme , Tumeurs cutanées/génétique , Peau/anatomopathologie , Animaux , Lignée cellulaire tumorale , Prolifération cellulaire , Chimiokine CXCL5/biosynthèse , Humains , Mélanome/métabolisme , Mélanome/anatomopathologie , Souris , Granulocytes neutrophiles/anatomopathologie , Réaction de polymérisation en chaîne , Peau/métabolisme , Tumeurs cutanées/métabolisme , Tumeurs cutanées/anatomopathologie ,
13.
Nature ; 556(7701): 376-380, 2018 04.
Article de Anglais | MEDLINE | ID: mdl-29643507

RÉSUMÉ

Ribosome-associated mRNA quality control mechanisms ensure the fidelity of protein translation1,2. Although these mechanisms have been extensively studied in yeast, little is known about their role in mammalian tissues, despite emerging evidence that stem cell fate is controlled by translational mechanisms3,4. One evolutionarily conserved component of the quality control machinery, Dom34 (in higher eukaryotes known as Pelota (Pelo)), rescues stalled ribosomes 5 . Here we show that Pelo is required for mammalian epidermal homeostasis. Conditional deletion of Pelo in mouse epidermal stem cells that express Lrig1 results in hyperproliferation and abnormal differentiation of these cells. By contrast, deletion of Pelo in Lgr5-expressing stem cells has no effect and deletion in Lgr6-expressing stem cells induces only a mild phenotype. Loss of Pelo results in accumulation of short ribosome footprints and global upregulation of translation, rather than affecting the expression of specific genes. Translational inhibition by rapamycin-mediated downregulation of mTOR (mechanistic target of rapamycin kinase) rescues the epidermal phenotype. Our study reveals that the ribosome-rescue machinery is important for mammalian tissue homeostasis and that it has specific effects on different stem cell populations.


Sujet(s)
Évolution biologique , Épiderme/métabolisme , Homéostasie , Ribosomes/métabolisme , Cellules souches/métabolisme , Animaux , Protéines du cycle cellulaire/déficit , Protéines du cycle cellulaire/génétique , Différenciation cellulaire , Prolifération cellulaire , Évolution de la maladie , Endonucleases , Cellules épidermiques , Épiderme/anatomopathologie , Femelle , Homéostasie/génétique , Mâle , Glycoprotéines membranaires/métabolisme , Souris , Protéines des microfilaments/déficit , Protéines des microfilaments/génétique , Mutation , Protéines de tissu nerveux/métabolisme , Phénotype , Biosynthèse des protéines , ARN messager/métabolisme , Récepteurs couplés aux protéines G/métabolisme , Cellules souches/cytologie , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Sérine-thréonine kinases TOR/métabolisme
14.
J Invest Dermatol ; 137(11): 2270-2281, 2017 11.
Article de Anglais | MEDLINE | ID: mdl-28668474

RÉSUMÉ

B-lymphocyte-induced maturation protein 1 (Blimp1) is a transcriptional repressor that regulates cell growth and differentiation in multiple tissues, including skin. Although in the epidermis Blimp1 is important for keratinocyte and sebocyte differentiation, its role in dermal fibroblasts is unclear. Here we show that Blimp1 is dynamically regulated in dermal papilla cells during hair follicle (HF) morphogenesis and the postnatal hair cycle, preceding dermal Wnt/ß-catenin activation. Blimp1 ablation in E12.5 mouse dermal fibroblasts delayed HF morphogenesis and growth and prevented new HF formation after wounding. By combining targeted quantitative PCR screens with bioinformatic analysis and experimental validation we demonstrated that Blimp1 is both a target and a mediator of key dermal papilla inductive signaling pathways including transforming growth factor-ß and Wnt/ß-catenin. Epidermal overexpression of stabilized ß-catenin was able to override the HF defects in Blimp1 mutant mice, underlining the close reciprocal relationship between the dermal papilla and adjacent HF epithelial cells. Overall, our study reveals the functional role of Blimp1 in promoting the dermal papilla inductive signaling cascade that initiates HF growth.


Sujet(s)
Régulation de l'expression des gènes , Follicule pileux/croissance et développement , Facteurs de transcription/génétique , Facteur de croissance transformant bêta/génétique , Voie de signalisation Wnt/génétique , Animaux , Ponction-biopsie à l'aiguille , Communication cellulaire/génétique , Différenciation cellulaire , Prolifération cellulaire , Cellules cultivées , Modèles animaux de maladie humaine , Régulation négative , Cellules épidermiques , Épiderme/métabolisme , Femelle , Follicule pileux/physiologie , Immunohistochimie , Mâle , Souris , Souris de lignée C57BL , Souris de lignée CBA , Facteur-1 liant le domaine de régulation positive I , ARN messager/analyse , Répartition aléatoire , Réaction de polymérisation en chaine en temps réel , Régénération/génétique , bêta-Caténine/métabolisme
15.
Nat Commun ; 8: 14744, 2017 03 23.
Article de Anglais | MEDLINE | ID: mdl-28332498

RÉSUMÉ

Individual human epidermal cells differ in their self-renewal ability. To uncover the molecular basis for this heterogeneity, we performed genome-wide pooled RNA interference screens and identified genes conferring a clonal growth advantage on normal and neoplastic (cutaneous squamous cell carcinoma, cSCC) human epidermal cells. The Hippo effector YAP was amongst the top positive growth regulators in both screens. By integrating the Hippo network interactome with our data sets, we identify WW-binding protein 2 (WBP2) as an important co-factor of YAP that enhances YAP/TEAD-mediated gene transcription. YAP and WPB2 are upregulated in actively proliferating cells of mouse and human epidermis and cSCC, and downregulated during terminal differentiation. WBP2 deletion in mouse skin results in reduced proliferation in neonatal and wounded adult epidermis. In reconstituted epidermis YAP/WBP2 activity is controlled by intercellular adhesion rather than canonical Hippo signalling. We propose that defective intercellular adhesion contributes to uncontrolled cSCC growth by preventing inhibition of YAP/WBP2.


Sujet(s)
Protéines adaptatrices de la transduction du signal/génétique , Prolifération cellulaire/génétique , Protéines nucléaires/génétique , Cellules souches/métabolisme , Facteurs de transcription/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Animaux , Protéines du cycle cellulaire , Lignée cellulaire tumorale , Cellules cultivées , Cellules épidermiques , Femelle , Régulation de l'expression des gènes , Humains , Souris de lignée C57BL , Souris de lignée CBA , Souris knockout , Protéines nucléaires/métabolisme , Cellules souches/cytologie , Transactivateurs , Facteurs de transcription/métabolisme
17.
Nat Commun ; 7: 10537, 2016 Feb 03.
Article de Anglais | MEDLINE | ID: mdl-26837596

RÉSUMÉ

Sustained epidermal Wnt/ß-catenin signalling expands the stem cell compartment and induces ectopic hair follicles (EFs). This is accompanied by extensive fibroblast proliferation and extracellular matrix (ECM) remodelling in the underlying dermis. Here we show that epidermal Hedgehog (Hh) and Transforming growth factor-beta (TGF-ß) signalling mediate the dermal changes. Pharmacological inhibition or genetic deletion of these pathways prevents ß-catenin-induced dermal reprogramming and EF formation. Epidermal Shh stimulates proliferation of the papillary fibroblast lineage, whereas TGF-ß2 controls proliferation, differentiation and ECM production by reticular fibroblasts. Hh inhibitors do not affect TGF-ß target gene expression in reticular fibroblasts, and TGF-ß inhibition does not prevent Hh target gene induction in papillary fibroblasts. However, when Hh signalling is inhibited the reticular dermis does not respond to epidermal ß-catenin activation. We conclude that the dermal response to epidermal Wnt/ß-catenin signalling depends on distinct fibroblast lineages responding to different paracrine signals.


Sujet(s)
Prolifération cellulaire , Derme/métabolisme , Épiderme/métabolisme , Matrice extracellulaire/métabolisme , Fibroblastes/métabolisme , Follicule pileux/métabolisme , Protéines Hedgehog/métabolisme , Communication paracrine , Facteur de croissance transformant bêta-2/métabolisme , Voie de signalisation Wnt , Animaux , Différenciation cellulaire , Lignage cellulaire , Derme/anatomopathologie , Épiderme/anatomopathologie , Matrice extracellulaire/anatomopathologie , Fibroblastes/anatomopathologie , Cytométrie en flux , Follicule pileux/cytologie , Souris , RT-PCR , Cellules souches , Facteur de croissance transformant bêta/métabolisme , bêta-Caténine/métabolisme
18.
J Invest Dermatol ; 136(6): 1130-1142, 2016 06.
Article de Anglais | MEDLINE | ID: mdl-26902921

RÉSUMÉ

The Wnt/ß-catenin pathway plays a central role in epidermal homeostasis and regeneration, but how it affects fibroblast fate decisions is unknown. We investigated the effect of targeted ß-catenin stabilization in dermal fibroblasts. Comparative gene expression profiling of stem cell antigen 1(-) (Sca1(-)) and Sca1(+) neonatal fibroblasts from upper and lower dermis, respectively, confirmed that Sca1(+) cells had a preadipocyte signature and showed differential expression of Wnt/ß-catenin-associated genes. By targeting all fibroblasts or selectively targeting Dlk1(+) lower dermal fibroblasts, we found that ß-catenin stabilization between developmental stages E16.5 and P2 resulted in a reduction in the dermal adipocyte layer with a corresponding increase in dermal fibrosis and an altered hair cycle. The fibrotic phenotype correlated with a reduction in the potential of Sca1(+) fibroblasts to undergo adipogenic differentiation ex vivo. Our findings indicate that Wnt/ß-catenin signaling controls adipogenic cell fate within the lower dermis, which potentially contributes to the pathogenesis of fibrotic skin diseases.


Sujet(s)
Adipocytes/métabolisme , Fibroblastes/cytologie , Maladies de la peau/anatomopathologie , Voie de signalisation Wnt/génétique , bêta-Caténine/génétique , Animaux , Animaux nouveau-nés , Cellules cultivées , Derme/cytologie , Derme/métabolisme , Modèles animaux de maladie humaine , Femelle , Fibroblastes/métabolisme , Fibrose/anatomopathologie , Cytométrie en flux , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes au cours du développement , Immunohistochimie , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Maladies de la peau/physiopathologie
19.
J Invest Dermatol ; 135(12): 2926-2934, 2015 Dec.
Article de Anglais | MEDLINE | ID: mdl-26288357

RÉSUMÉ

Prominin-1/CD133 (Prom1) is expressed by fibroblasts in the dermal papilla (DP) of the hair follicle (HF). By examining endogenous Prom1 expression and expression of LacZ in the skin of Prom1CreERLacZ (Prom1C-L) mice, in which a CreERT2-IRES-nuclear LacZ cassette is knocked into the first ATG codon of Prom1, we confirmed that Prom1 is expressed in the DP of all developing HFs and also by postnatal anagen follicles. To analyze the fate of Prom1+ DP cells, we crossed Prom1C-L mice with Rosa26-CAG flox/stop/flox tdTomato reporter mice and applied 4-hydroxytamoxifen (4OHT) to back skin at postnatal day (P) 1 and P2. We detected tdTomato+ cells in ~50% of DPs. The proportion of labeled cells per DP increased between P5 and P63, while the total number of cells per DP declined. Following full thickness wounding, there was no migration of tdTomato-labeled cells out of the DP. When ß-catenin was activated in Prom1+ DP cells there was an increase in the size of anagen and telogen DP, but the proportion of tdTomato-labeled cells did not increase. We conclude that Prom1+ DP cells do not contribute to dermal repair but are nevertheless capable of regulating DP size via ß-catenin-mediated intercellular communication.


Sujet(s)
Antigènes CD/physiologie , Derme/physiologie , Glycoprotéines/physiologie , Homéostasie/physiologie , Peptides/physiologie , Protéines de type Wingless/physiologie , Cicatrisation de plaie/physiologie , Antigène AC133 , Animaux , Antigènes CD/analyse , Communication cellulaire , Mouvement cellulaire , Glycoprotéines/analyse , Souris , Souris transgéniques , Peptides/analyse , bêta-Caténine/physiologie
20.
Nature ; 504(7479): 277-281, 2013 Dec 12.
Article de Anglais | MEDLINE | ID: mdl-24336287

RÉSUMÉ

Fibroblasts are the major mesenchymal cell type in connective tissue and deposit the collagen and elastic fibres of the extracellular matrix (ECM). Even within a single tissue, fibroblasts exhibit considerable functional diversity, but it is not known whether this reflects the existence of a differentiation hierarchy or is a response to different environmental factors. Here we show, using transplantation assays and lineage tracing in mice, that the fibroblasts of skin connective tissue arise from two distinct lineages. One forms the upper dermis, including the dermal papilla that regulates hair growth and the arrector pili muscle, which controls piloerection. The other forms the lower dermis, including the reticular fibroblasts that synthesize the bulk of the fibrillar ECM, and the preadipocytes and adipocytes of the hypodermis. The upper lineage is required for hair follicle formation. In wounded adult skin, the initial wave of dermal repair is mediated by the lower lineage and upper dermal fibroblasts are recruited only during re-epithelialization. Epidermal ß-catenin activation stimulates the expansion of the upper dermal lineage, rendering wounds permissive for hair follicle formation. Our findings explain why wounding is linked to formation of ECM-rich scar tissue that lacks hair follicles. They also form a platform for discovering fibroblast lineages in other tissues and for examining fibroblast changes in ageing and disease.


Sujet(s)
Lignage cellulaire , Fibroblastes/cytologie , Peau/cytologie , Peau/croissance et développement , Cicatrisation de plaie/physiologie , Adipocytes/cytologie , Adipocytes/métabolisme , Animaux , Derme/anatomie et histologie , Derme/cytologie , Derme/embryologie , Derme/croissance et développement , Femelle , Fibroblastes/transplantation , Follicule pileux/cytologie , Follicule pileux/métabolisme , Techniques in vitro , Mâle , Souris , Souris de lignée C57BL , Souris de lignée CBA , Souris transgéniques , Muscles lisses/cytologie , Muscles lisses/métabolisme , Peau/anatomie et histologie , Peau/embryologie , bêta-Caténine/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...