Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros













Base de dados
Intervalo de ano de publicação
1.
Dev Cell ; 58(20): 2140-2162.e5, 2023 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-37591247

RESUMO

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.


Assuntos
Epiderme , Pele , Camundongos , Animais , Folículo Piloso/patologia , Cabelo , Epitélio
2.
Nat Cell Biol ; 25(2): 222-234, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36717629

RESUMO

Substantial follicle remodelling during the regression phase of the hair growth cycle is coordinated by the contraction of the dermal sheath smooth muscle, but how dermal-sheath-generated forces are regulated is unclear. Here, we identify spatiotemporally controlled endothelin signalling-a potent vasoconstriction-regulating pathway-as the key activating mechanism of dermal sheath contraction. Pharmacological blocking or genetic ablation of both endothelin receptors, ETA and ETB, impedes dermal sheath contraction and halts follicle regression. Epithelial progenitors at the club hair-epithelial strand bottleneck produce the endothelin ligand ET-1, which is required for follicle regression. ET signalling in dermal sheath cells and downstream contraction is dynamically regulated by cytoplasmic Ca2+ levels through cell membrane and sarcoplasmic reticulum calcium channels. Together, these findings illuminate an epithelial-mesenchymal interaction paradigm in which progenitors-destined to undergo programmed cell death-control the contraction of the surrounding sheath smooth muscle to orchestrate homeostatic tissue regression and reorganization for the next stem cell activation and regeneration cycle.


Assuntos
Endotelinas , Folículo Piloso , Folículo Piloso/metabolismo , Endotelinas/metabolismo , Endotelinas/farmacologia , Receptores de Endotelina/metabolismo , Músculo Liso/metabolismo , Transdução de Sinais , Contração Muscular
3.
Nature ; 594(7864): 500-501, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34108720
4.
Nature ; 592(7854): 428-432, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33790465

RESUMO

Chronic, sustained exposure to stressors can profoundly affect tissue homeostasis, although the mechanisms by which these changes occur are largely unknown. Here we report that the stress hormone corticosterone-which is derived from the adrenal gland and is the rodent equivalent of cortisol in humans-regulates hair follicle stem cell (HFSC) quiescence and hair growth in mice. In the absence of systemic corticosterone, HFSCs enter substantially more rounds of the regeneration cycle throughout life. Conversely, under chronic stress, increased levels of corticosterone prolong HFSC quiescence and maintain hair follicles in an extended resting phase. Mechanistically, corticosterone acts on the dermal papillae to suppress the expression of Gas6, a gene that encodes the secreted factor growth arrest specific 6. Restoring Gas6 expression overcomes the stress-induced inhibition of HFSC activation and hair growth. Our work identifies corticosterone as a systemic inhibitor of HFSC activity through its effect on the niche, and demonstrates that the removal of such inhibition drives HFSCs into frequent regeneration cycles, with no observable defects in the long-term.


Assuntos
Corticosterona/farmacologia , Folículo Piloso/citologia , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Glândulas Suprarrenais/metabolismo , Glândulas Suprarrenais/cirurgia , Adrenalectomia , Animais , Divisão Celular/efeitos dos fármacos , Feminino , Folículo Piloso/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Psicológico/metabolismo , Estresse Psicológico/patologia , Transcriptoma , Regulação para Cima
6.
Exp Dermatol ; 30(4): 512-521, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33006790

RESUMO

Hair follicles cyclically regenerate throughout adult mammalian life, owing to a resident population of epithelial hair follicle stem cells. Stem cell (SC) activity drives bouts of follicle growth, which are periodically interrupted by follicle regression and rest. These phases and the transitions between them are tightly spatiotemporally coordinated by signalling crosstalk between stem/progenitor cells and the various cell types of the microenvironment, or niche. The dermal papilla (DP) is a cluster of specialized mesenchymal cells that have long been recognized for important niche roles in regulating hair follicle SC activation, as well as progenitor proliferation and differentiation during follicle growth. In addition to the DP, the mesenchyme of the murine pelage follicle is also comprised of a follicle-lining smooth muscle known as the dermal sheath (DS), which has been far less studied than the DP yet may be equally specialized and important for hair cycling. In this review, we define the murine pelage DS in comparison with human DS and discuss recent work that highlights the emergent importance of the DS in the hair follicle SC niche. Last, we examine potential therapeutic applications for the DS in hair regeneration and wound healing.


Assuntos
Folículo Piloso/fisiologia , Regeneração/fisiologia , Nicho de Células-Tronco/fisiologia , Células-Tronco/fisiologia , Alopecia , Animais , Humanos , Camundongos , Cicatrização
7.
Cell Stem Cell ; 26(6): 880-895.e6, 2020 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-32302523

RESUMO

Mature adipocytes store fatty acids and are a common component of tissue stroma. Adipocyte function in regulating bone marrow, skin, muscle, and mammary gland biology is emerging, but the role of adipocyte-derived lipids in tissue homeostasis and repair is poorly understood. Here, we identify an essential role for adipocyte lipolysis in regulating inflammation and repair after injury in skin. Genetic mouse studies revealed that dermal adipocytes are necessary to initiate inflammation after injury and promote subsequent repair. We find through histological, ultrastructural, lipidomic, and genetic experiments in mice that adipocytes adjacent to skin injury initiate lipid release necessary for macrophage inflammation. Tamoxifen-inducible genetic lineage tracing of mature adipocytes and single-cell RNA sequencing revealed that dermal adipocytes alter their fate and generate ECM-producing myofibroblasts within wounds. Thus, adipocytes regulate multiple aspects of repair and may be therapeutic for inflammatory diseases and defective wound healing associated with aging and diabetes.


Assuntos
Lipólise , Miofibroblastos , Adipócitos , Animais , Macrófagos , Camundongos , Pele
8.
Science ; 367(6474): 161-166, 2020 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-31857493

RESUMO

Tissue homeostasis requires the balance of growth by cell production and regression through cell loss. In the hair cycle, during follicle regression, the niche traverses the skin through an unknown mechanism to reach the stem cell reservoir and trigger new growth. Here, we identify the dermal sheath that lines the follicle as the key driver of tissue regression and niche relocation through the smooth muscle contractile machinery that generates centripetal constriction force. We reveal that the calcium-calmodulin-myosin light chain kinase pathway controls sheath contraction. When this pathway is blocked, sheath contraction is inhibited, impeding follicle regression and niche relocation. Thus, our study identifies the dermal sheath as smooth muscle that drives follicle regression for reuniting niche and stem cells in order to regenerate tissue structure during homeostasis.


Assuntos
Derme/fisiologia , Folículo Piloso/fisiologia , Músculo Liso/fisiologia , Regeneração , Nicho de Células-Tronco/fisiologia , Agrecanas/genética , Animais , Humanos , Camundongos , Camundongos Mutantes , Contração Muscular
9.
Exp Dermatol ; 28(4): 332-344, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30887615

RESUMO

Hair follicle (HF) formation in developing embryonic skin requires stepwise signalling between the epithelial epidermis and mesenchymal dermis, and their specialized derivatives, the placode/germ/peg and dermal condensate/papilla, respectively. Classically, distinct stages of HF morphogenesis have been defined, in the mouse model, based on (a) changes in cell morphology and aggregation; (b) expression of few known molecular markers; (c) the extent of follicle downgrowth; and (d) the presence of differentiating cell types. Refined genetic strategies and recent emerging technologies, such as live imaging and transcriptome analyses of isolated cell populations or single cells, have enabled a closer dissection of the signalling requirements at different stages of HF formation, particularly early on. They have also led to the discovery of precursor cells for placode, dermal condensate and future bulge stem cells that, combined with molecular insights into their fate specification and subsequent formation, serve as novel landmarks for early HF morphogenetic events and studies of the signalling networks mediating these processes. In this review, we integrate the emergence of HF precursor cell states and novel molecular markers of fate and formation to update the widely used 20-year-old seminal classification guide of HF morphogenetic stages by Paus et al. We then temporally describe the latest insights into the early cellular and molecular events and signalling requirements for HF morphogenesis in relation to one another in a holistic manner.


Assuntos
Folículo Piloso/embriologia , Animais , Humanos , Morfogênese , Transdução de Sinais
10.
Dev Cell ; 48(1): 32-48.e5, 2019 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-30595537

RESUMO

Cell fate transitions are essential for specification of stem cells and their niches, but the precise timing and sequence of molecular events during embryonic development are largely unknown. Here, we identify, with 3D and 4D microscopy, unclustered precursors of dermal condensates (DC), signaling niches for epithelial progenitors in hair placodes. With population-based and single-cell transcriptomics, we define a molecular time-lapse from pre-DC fate specification through DC niche formation and establish the developmental trajectory as the DC lineage emerges from fibroblasts. Co-expression of downregulated fibroblast and upregulated DC genes in niche precursors reveals a transitory molecular state following a proliferation shutdown. Waves of transcription factor and signaling molecule expression then coincide with DC formation. Finally, ablation of epidermal Wnt signaling and placode-derived FGF20 demonstrates their requirement for pre-DC specification. These findings uncover a progenitor-dependent niche precursor fate and the transitory molecular events controlling niche formation and function.


Assuntos
Diferenciação Celular/fisiologia , Derme/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Folículo Piloso/metabolismo , Animais , Fibroblastos/citologia , Folículo Piloso/embriologia , Transdução de Sinais/genética , Pele/metabolismo , Células-Tronco/citologia
11.
Cell Rep ; 25(4): 823-824, 2018 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-30355488

RESUMO

Compared to mouse models, less is known about human epidermal cell states and differentiation. In this issue of Cell Reports, Cheng et al. (2018) dissect the cell states and heterogeneity in human epidermis with large-scale transcriptomics of 92,889 single epidermal cells from normal and inflamed skin.


Assuntos
Células Epidérmicas , Transcriptoma , Animais , Diferenciação Celular , Epiderme , Humanos , Camundongos , Pele
12.
Curr Opin Cell Biol ; 55: 87-95, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30031324

RESUMO

Adult tissue-specific stem cells are essential for homeostatic tissue maintenance and key to regeneration during injury repair or disease. Many critical stem cell functions rely on the presence of well-timed cues from the microenvironment or niche, which includes a diverse range of components, including neuronal, circulating and extracellular matrix inputs as well as an array of neighboring niche cells directly interacting with the stem cells. However, studies of stem cells and their niche have been challenging due to the complexity of adult stem cell functions, their intrinsic controls and the multiple regulatory niche components. Here, we review recent major advances in our understanding of the complex interplay between stem cells and their niche that were enabled by the tremendous technological leaps in single-cell transcriptome analyses, 3D in vitro cultures and 4D in vivo microscopy of stem cell niches.


Assuntos
Nicho de Células-Tronco , Pesquisa com Células-Tronco , Animais , Humanos , Imageamento Tridimensional , Modelos Biológicos , Organoides/metabolismo , Análise de Célula Única
13.
Cell Stem Cell ; 22(5): 726-739.e7, 2018 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-29727681

RESUMO

Polycomb repressive complexes (PRCs) 1 and 2 are essential chromatin regulators of cell identity. PRC1, a dominant executer of Polycomb-mediated control, functions as multiple sub-complexes that possess catalytic-dependent H2AK119 mono-ubiquitination (H2AK119ub) and catalytic-independent activities. Here, we show that, despite its well-established repressor functions, PRC1 binds to both silent and active genes. Through in vivo loss-of-function studies, we show that global PRC1 function is essential for skin development and stem cell (SC) specification, whereas PRC1 catalytic activity is dispensable. Further dissection demonstrated that both canonical and non-canonical PRC1 complexes bind to repressed genes, marked by H2AK119ub and PRC2-mediated H3K27me3. Interestingly, loss of canonical PRC1, PRC1 catalytic activity, or PRC2 leads to expansion of mechanosensitive Merkel cells in neonatal skin. Non-canonical PRC1 complexes, however, also bind to and promote expression of genes critical for skin development and SC formation. Together, our findings highlight PRC1's diverse roles in executing a precise developmental program.


Assuntos
Complexo Repressor Polycomb 1/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Pele/metabolismo , Células-Tronco/metabolismo , Animais , Biocatálise , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Complexo Repressor Polycomb 1/deficiência , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 2/genética , Pele/citologia
14.
Cell Rep ; 14(12): 3001-18, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-27009580

RESUMO

The hair follicle (HF) is a complex miniorgan that serves as an ideal model system to study stem cell (SC) interactions with the niche during growth and regeneration. Dermal papilla (DP) cells are required for SC activation during the adult hair cycle, but signal exchange between niche and SC precursors/transit-amplifying cell (TAC) progenitors that regulates HF morphogenetic growth is largely unknown. Here we use six transgenic reporters to isolate 14 major skin and HF cell populations. With next-generation RNA sequencing, we characterize their transcriptomes and define unique molecular signatures. SC precursors, TACs, and the DP niche express a plethora of ligands and receptors. Signaling interaction network analysis reveals a bird's-eye view of pathways implicated in epithelial-mesenchymal interactions. Using a systematic tissue-wide approach, this work provides a comprehensive platform, linked to an interactive online database, to identify and further explore the SC/TAC/niche crosstalk regulating HF growth.


Assuntos
Folículo Piloso/metabolismo , Pele/metabolismo , Células-Tronco/citologia , Animais , Citometria de Fluxo , Queratina-14/genética , Queratina-14/metabolismo , Camundongos , Microscopia de Fluorescência , Análise de Componente Principal , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Análise de Sequência de RNA , Transdução de Sinais/genética , Pele/citologia , Nicho de Células-Tronco , Células-Tronco/metabolismo , Transcriptoma
15.
Dev Cell ; 34(5): 577-91, 2015 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-26256211

RESUMO

Defining the unique molecular features of progenitors and their niche requires a genome-wide, whole-tissue approach with cellular resolution. Here, we co-isolate embryonic hair follicle (HF) placode and dermal condensate cells, precursors of adult HF stem cells and the dermal papilla/sheath niche, along with lineage-related keratinocytes and fibroblasts, Schwann cells, melanocytes, and a population inclusive of all remaining skin cells. With next-generation RNA sequencing, we define gene expression patterns in the context of the entire embryonic skin, and through transcriptome cross-comparisons, we uncover hundreds of enriched genes in cell-type-specific signatures. Axon guidance signaling and many other pathway genes are enriched in multiple signatures, implicating these factors in driving the large-scale cellular rearrangements necessary for HF formation. Finally, we share all data in an interactive, searchable companion website. Our study provides an overarching view of signaling within the entire embryonic skin and captures a molecular snapshot of HF progenitors and their niche.


Assuntos
Folículo Piloso/citologia , Folículo Piloso/embriologia , Queratinócitos/citologia , Pele/metabolismo , Células-Tronco/citologia , Transcriptoma/fisiologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Camundongos , Organogênese/fisiologia , Transdução de Sinais/fisiologia , Pele/citologia , Pele/embriologia , Nicho de Células-Tronco
17.
Exp Dermatol ; 24(6): 468-70, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25708924

RESUMO

Embryonic hair follicle (HF) induction and formation is dependent on signalling crosstalk between the dermis and specialized dermal condensates on the mesenchymal side and epidermal cells and incipient placodes on the epithelial side, but the precise nature and succession of signals remain unclear. Platelet-derived growth factor (PDGF) signalling is involved in the development of several organs and the maintenance of adult tissues, including HF regeneration in the hair cycle. As both PDGF receptors, PDGFRα and PDGFRß, are expressed in embryonic dermis and dermal condensates, we explored in this study the role of PDGF signalling in HF induction and formation in the developing skin mesenchyme. We conditionally ablated both PDGF receptors with Tbx18(Cre) in early dermal condensates before follicle formation, and with Prx1-Cre broadly in the ventral dermis prior to HF induction. In both PDGFR double mutants, HF induction and formation ensued normally, and the pattern of HF formation and HF numbers were unaffected. These data demonstrate that mesenchymal PDGF signalling, either in the specialized niche or broadly in the dermis, is dispensable for HF induction and formation.


Assuntos
Derme/embriologia , Folículo Piloso/embriologia , Morfogênese/fisiologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Transdução de Sinais/fisiologia , Animais , Derme/citologia , Derme/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Folículo Piloso/citologia , Folículo Piloso/fisiologia , Mesoderma/citologia , Mesoderma/embriologia , Mesoderma/fisiologia , Camundongos , Camundongos Mutantes , Modelos Animais , Morfogênese/genética , Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/fisiologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Transdução de Sinais/genética
19.
Nat Commun ; 5: 4511, 2014 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-25077433

RESUMO

Although the principles that balance stem cell self-renewal and differentiation in normal tissue homeostasis are beginning to emerge, it is still unclear whether cancer cells with tumour initiating potential are similarly governed, or whether they have acquired distinct mechanisms to sustain self-renewal and long-term tumour growth. Here we show that the transcription factor Sox2, which is not expressed in normal skin epithelium and is dispensable for epidermal homeostasis, marks tumour initiating cells (TICs) in cutaneous squamous cell carcinomas (SCCs). We demonstrate that Sox2 is required for SCC growth in mouse and human, where it enhances Nrp1/Vegf signalling to promote the expansion of TICs along the tumour-stroma interface. Our findings suggest that distinct transcriptional programmes govern self-renewal and long-term growth of TICs and normal skin epithelial stem and progenitor cells. These programmes present promising diagnostic markers and targets for cancer-specific therapies.


Assuntos
Carcinoma de Células Escamosas/genética , Células-Tronco Neoplásicas/metabolismo , Neuropilina-1/genética , Fatores de Transcrição SOXB1/genética , Neoplasias Cutâneas/genética , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Células-Tronco Neoplásicas/patologia , Neuropilina-1/antagonistas & inibidores , Neuropilina-1/metabolismo , Especificidade de Órgãos , Cultura Primária de Células , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Fatores de Transcrição SOXB1/antagonistas & inibidores , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais , Pele/metabolismo , Pele/patologia , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Células Estromais/metabolismo , Células Estromais/patologia , Transcrição Gênica , Microambiente Tumoral/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
20.
Exp Dermatol ; 23(10): 748-50, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25066162

RESUMO

Hair follicle (HF) morphogenesis relies on the coordinated exchange of signals between mesenchymal and epithelial compartments of embryonic skin. Chemokine receptor Cxcr4 expression was recently identified in dermal condensates (DCs) of nascent HFs, but its role in promoting HF morphogenesis remains unknown. Our analyses confirmed Cxcr4 expression in condensate cells, and additionally revealed transient Cxcr4 expression in incipient epithelial hair placodes. Placodal Cxcr4 appeared prior to detection in DCs, representing a switch of expression between epithelial and mesenchymal compartments. To explore the functional role of this receptor in both compartments for early HF formation, we conditionally ablated Cxcr4 with condensate-targeting Tbx18(cre) knock-in and epidermis-targeting Krt14-cre transgenic mice. Conditional knockouts for both crosses were viable throughout embryogenesis and into adulthood. Morphological and biochemical marker analyses revealed comparable numbers of HFs forming in knockout embryos compared to wild-type littermate controls in both cases, suggesting that neither dermal nor epithelial Cxcr4 expression is required for early HF morphogenesis. We conclude that Cxcr4 expression and chemokine signaling through this receptor in embryonic mouse skin is dispensable for HF formation.


Assuntos
Folículo Piloso/embriologia , Folículo Piloso/metabolismo , Receptores CXCR4/metabolismo , Animais , Epitélio/embriologia , Epitélio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Morfogênese , Receptores CXCR4/deficiência , Receptores CXCR4/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA