Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.755
Filtrar
1.
Cell ; 182(3): 578-593.e19, 2020 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-32679029

RESUMO

Piloerection (goosebumps) requires concerted actions of the hair follicle, the arrector pili muscle (APM), and the sympathetic nerve, providing a model to study interactions across epithelium, mesenchyme, and nerves. Here, we show that APMs and sympathetic nerves form a dual-component niche to modulate hair follicle stem cell (HFSC) activity. Sympathetic nerves form synapse-like structures with HFSCs and regulate HFSCs through norepinephrine, whereas APMs maintain sympathetic innervation to HFSCs. Without norepinephrine signaling, HFSCs enter deep quiescence by down-regulating the cell cycle and metabolism while up-regulating quiescence regulators Foxp1 and Fgf18. During development, HFSC progeny secretes Sonic Hedgehog (SHH) to direct the formation of this APM-sympathetic nerve niche, which in turn controls hair follicle regeneration in adults. Our results reveal a reciprocal interdependence between a regenerative tissue and its niche at different stages and demonstrate sympathetic nerves can modulate stem cells through synapse-like connections and neurotransmitters to couple tissue production with demands.


Assuntos
Nervo Acessório/fisiologia , Folículo Piloso/citologia , Cabelo/crescimento & desenvolvimento , Proteínas Hedgehog/metabolismo , Norepinefrina/metabolismo , Transdução de Sinais/genética , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Nervo Acessório/citologia , Animais , Ciclo Celular/genética , Temperatura Baixa , Feminino , Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Perfilação da Expressão Gênica , Cabelo/citologia , Cabelo/fisiologia , Folículo Piloso/crescimento & desenvolvimento , Folículo Piloso/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Piloereção , RNA-Seq , Receptores Adrenérgicos beta 2/deficiência , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor Smoothened/genética , Receptor Smoothened/metabolismo , Nicho de Células-Tronco , Células-Tronco/citologia , Sistema Nervoso Simpático/citologia , Sistema Nervoso Simpático/fisiologia , Sinapses/fisiologia
2.
Cell ; 169(3): 483-496.e13, 2017 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-28413068

RESUMO

Adult tissue stem cells (SCs) reside in niches, which, through intercellular contacts and signaling, influence SC behavior. Once activated, SCs typically give rise to short-lived transit-amplifying cells (TACs), which then progress to differentiate into their lineages. Here, using single-cell RNA-seq, we unearth unexpected heterogeneity among SCs and TACs of hair follicles. We trace the roots of this heterogeneity to micro-niches along epithelial-mesenchymal interfaces, where progenitors display molecular signatures reflective of spatially distinct local signals and intercellular interactions. Using lineage tracing, temporal single-cell analyses, and chromatin landscaping, we show that SC plasticity becomes restricted in a sequentially and spatially choreographed program, culminating in seven spatially arranged unilineage progenitors within TACs of mature follicles. By compartmentalizing SCs into micro-niches, tissues gain precise control over morphogenesis and regeneration: some progenitors specify lineages immediately, whereas others retain potency, preserving self-renewing features established early while progressively restricting lineages as they experience dynamic changes in microenvironment.


Assuntos
Células-Tronco Adultas/citologia , Linhagem da Célula , Folículo Piloso/citologia , Nicho de Células-Tronco , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência de RNA , Análise de Célula Única , Via de Sinalização Wnt
3.
Cell ; 169(6): 1119-1129.e11, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28552347

RESUMO

The maintenance of tissue homeostasis is critically dependent on the function of tissue-resident immune cells and the differentiation capacity of tissue-resident stem cells (SCs). How immune cells influence the function of SCs is largely unknown. Regulatory T cells (Tregs) in skin preferentially localize to hair follicles (HFs), which house a major subset of skin SCs (HFSCs). Here, we mechanistically dissect the role of Tregs in HF and HFSC biology. Lineage-specific cell depletion revealed that Tregs promote HF regeneration by augmenting HFSC proliferation and differentiation. Transcriptional and phenotypic profiling of Tregs and HFSCs revealed that skin-resident Tregs preferentially express high levels of the Notch ligand family member, Jagged 1 (Jag1). Expression of Jag1 on Tregs facilitated HFSC function and efficient HF regeneration. Taken together, our work demonstrates that Tregs in skin play a major role in HF biology by promoting the function of HFSCs.


Assuntos
Folículo Piloso/citologia , Células-Tronco/metabolismo , Linfócitos T Reguladores/metabolismo , Animais , Células Epiteliais/metabolismo , Folículo Piloso/metabolismo , Humanos , Inflamação/metabolismo , Proteína Jagged-1/metabolismo , Camundongos
4.
Cell ; 169(4): 636-650.e14, 2017 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-28434617

RESUMO

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.


Assuntos
Carcinoma de Células Escamosas/patologia , Linhagem da Célula , Células Epidérmicas , Folículo Piloso/citologia , Neoplasias Cutâneas/patologia , Pele/citologia , Células-Tronco/metabolismo , Animais , Linhagem Celular Tumoral , Cromatina/metabolismo , Epiderme/metabolismo , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Cutâneas/metabolismo , Fatores de Transcrição/metabolismo , Transcriptoma , Transplante Heterólogo , Cicatrização
5.
Cell ; 164(1-2): 156-169, 2016 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-26771489

RESUMO

Adult stem cell (SC) maintenance and differentiation are known to depend on signals received from the niche. Here, however, we demonstrate a mechanism for SC specification and regulation that is niche independent. Using immunofluorescence, live imaging, genetics, cell-cycle analyses, in utero lentiviral transduction, and lineage-tracing, we show that in developing hair buds, SCs are born from asymmetric divisions that differentially display WNT and SHH signaling. Displaced WNT(lo) suprabasal daughters become SCs that respond to paracrine SHH and symmetrically expand. By contrast, basal daughters remain WNT(hi). They express but do not respond to SHH and hence maintain slow-cycling, asymmetric divisions. Over time, they become short-lived progenitors, generating differentiating daughters rather than SCs. Thus, in contrast to an established niche that harbors a fixed SC pool whose expelled progeny differentiate, asymmetric divisions first specify and displace early SCs into an environment conducive to expansion and later restrict their numbers by switching asymmetric fates.


Assuntos
Folículo Piloso/citologia , Proteínas Hedgehog/metabolismo , Camundongos/embriologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Via de Sinalização Wnt , Animais , Divisão Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Folículo Piloso/metabolismo , Microscopia de Fluorescência , Fatores de Transcrição SOX9/metabolismo
6.
Cell ; 161(2): 195-6, 2015 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-25860601

RESUMO

How do cells collectively control an organ's behavior? By plucking various numbers of hairs from the mouse skin, Chen et al. show that hairs regenerate only when a sufficiently high density of them are plucked. Remarkably, a hair follicle can only regenerate in concert with other follicles, but not autonomously.


Assuntos
Folículo Piloso/citologia , Células-Tronco/citologia , Animais
7.
Cell ; 161(2): 277-90, 2015 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-25860610

RESUMO

Coordinated organ behavior is crucial for an effective response to environmental stimuli. By studying regeneration of hair follicles in response to patterned hair plucking, we demonstrate that organ-level quorum sensing allows coordinated responses to skin injury. Plucking hair at different densities leads to a regeneration of up to five times more neighboring, unplucked resting hairs, indicating activation of a collective decision-making process. Through data modeling, the range of the quorum signal was estimated to be on the order of 1 mm, greater than expected for a diffusible molecular cue. Molecular and genetic analysis uncovered a two-step mechanism, where release of CCL2 from injured hairs leads to recruitment of TNF-α-secreting macrophages, which accumulate and signal to both plucked and unplucked follicles. By coupling immune response with regeneration, this mechanism allows skin to respond predictively to distress, disregarding mild injury, while meeting stronger injury with full-scale cooperative activation of stem cells.


Assuntos
Folículo Piloso/citologia , Células-Tronco/citologia , Animais , Comunicação Celular , Quimiocina CCL2/metabolismo , Folículo Piloso/fisiologia , Queratinócitos/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Regeneração , Pele/citologia , Pele/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
8.
Cell ; 157(4): 769-70, 2014 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-24813602

RESUMO

Although stem cells are subject to niche control, evidence is emerging that they also contribute to generating the niche through their offspring. Using the hair follicle as a model, Hsu at al. demonstrate that the transient-amplifying cells, downstream of stem cells and well-known cell producers, signal back to stem cells to maintain long-term regenerative capacity.


Assuntos
Folículo Piloso/citologia , Cabelo/citologia , Cabelo/fisiologia , Nicho de Células-Tronco , Células-Tronco/citologia , Animais
9.
Cell ; 157(4): 935-49, 2014 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-24813615

RESUMO

Transit-amplifying cells (TACs) are an early intermediate in tissue regeneration. Here, using hair follicles (HFs) as a paradigm, we show that emerging TACs constitute a signaling center that orchestrates tissue growth. Whereas primed stem cells (SCs) generate TACs, quiescent SCs only proliferate after TACs form and begin expressing Sonic Hedgehog (SHH). TAC generation is independent of autocrine SHH, but the TAC pool wanes if they can't produce SHH. We trace this paradox to two direct actions of SHH: promoting quiescent-SC proliferation and regulating dermal factors that stoke TAC expansion. Ingrained within quiescent SCs' special sensitivity to SHH signaling is their high expression of GAS1. Without sufficient input from quiescent SCs, replenishment of primed SCs for the next hair cycle is compromised, delaying regeneration and eventually leading to regeneration failure. Our findings unveil TACs as transient but indispensable integrators of SC niche components and reveal an intriguing interdependency of primed and quiescent SC populations on tissue regeneration.


Assuntos
Folículo Piloso/citologia , Cabelo/citologia , Cabelo/fisiologia , Nicho de Células-Tronco , Células-Tronco/citologia , Animais , Proliferação de Células , Folículo Piloso/metabolismo , Proteínas Hedgehog/metabolismo , Camundongos , Regeneração , Transdução de Sinais , Células-Tronco/metabolismo
10.
Cell ; 159(7): 1640-51, 2014 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-25525881

RESUMO

The perception of touch, including the direction of stimulus movement across the skin, begins with activation of low-threshold mechanosensory neurons (LTMRs) that innervate the skin. Here, we show that murine Aδ-LTMRs are preferentially tuned to deflection of body hairs in the caudal-to-rostral direction. This tuning property is explained by the finding that Aδ-LTMR lanceolate endings around hair follicles are polarized; they are concentrated on the caudal (downward) side of each hair follicle. The neurotrophic factor BDNF is synthesized in epithelial cells on the caudal, but not rostral, side of hair follicles, in close proximity to Aδ-LTMR lanceolate endings, which express TrkB. Moreover, ablation of BDNF in hair follicle epithelial cells disrupts polarization of Aδ-LTMR lanceolate endings and results in randomization of Aδ-LTMR responses to hair deflection. Thus, BDNF-TrkB signaling directs polarization of Aδ-LTMR lanceolate endings, which underlies direction-selective responsiveness of Aδ-LTMRs to hair deflection.


Assuntos
Gânglios Espinais/fisiologia , Folículo Piloso/fisiologia , Mecanorreceptores/fisiologia , Tato , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Embrião de Mamíferos , Células Epiteliais/fisiologia , Folículo Piloso/citologia , Técnicas In Vitro , Mecanorreceptores/classificação , Camundongos , Receptor trkB/metabolismo
11.
Nature ; 616(7958): 774-782, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37076619

RESUMO

For unknow reasons, the melanocyte stem cell (McSC) system fails earlier than other adult stem cell populations1, which leads to hair greying in most humans and mice2,3. Current dogma states that McSCs are reserved in an undifferentiated state in the hair follicle niche, physically segregated from differentiated progeny that migrate away following cues of regenerative stimuli4-8. Here we show that most McSCs toggle between transit-amplifying and stem cell states for both self-renewal and generation of mature progeny, a mechanism fundamentally distinct from those of other self-renewing systems. Live imaging and single-cell RNA sequencing revealed that McSCs are mobile, translocating between hair follicle stem cell and transit-amplifying compartments where they reversibly enter distinct differentiation states governed by local microenvironmental cues (for example, WNT). Long-term lineage tracing demonstrated that the McSC system is maintained by reverted McSCs rather than by reserved stem cells inherently exempt from reversible changes. During ageing, there is accumulation of stranded McSCs that do not contribute to the regeneration of melanocyte progeny. These results identify a new model whereby dedifferentiation is integral to homeostatic stem cell maintenance and suggest that modulating McSC mobility may represent a new approach for the prevention of hair greying.


Assuntos
Desdiferenciação Celular , Folículo Piloso , Melanócitos , Nicho de Células-Tronco , Células-Tronco , Animais , Humanos , Camundongos , Folículo Piloso/citologia , Melanócitos/citologia , Células-Tronco/citologia , Microambiente Celular , Linhagem da Célula , Envelhecimento , Homeostase , Cor de Cabelo/fisiologia
12.
Nature ; 618(7966): 808-817, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37344645

RESUMO

Niche signals maintain stem cells in a prolonged quiescence or transiently activate them for proper regeneration1. Altering balanced niche signalling can lead to regenerative disorders. Melanocytic skin nevi in human often display excessive hair growth, suggesting hair stem cell hyperactivity. Here, using genetic mouse models of nevi2,3, we show that dermal clusters of senescent melanocytes drive epithelial hair stem cells to exit quiescence and change their transcriptome and composition, potently enhancing hair renewal. Nevus melanocytes activate a distinct secretome, enriched for signalling factors. Osteopontin, the leading nevus signalling factor, is both necessary and sufficient to induce hair growth. Injection of osteopontin or its genetic overexpression is sufficient to induce robust hair growth in mice, whereas germline and conditional deletions of either osteopontin or CD44, its cognate receptor on epithelial hair cells, rescue enhanced hair growth induced by dermal nevus melanocytes. Osteopontin is overexpressed in human hairy nevi, and it stimulates new growth of human hair follicles. Although broad accumulation of senescent cells, such as upon ageing or genotoxic stress, is detrimental for the regenerative capacity of tissue4, we show that signalling by senescent cell clusters can potently enhance the activity of adjacent intact stem cells and stimulate tissue renewal. This finding identifies senescent cells and their secretome as an attractive therapeutic target in regenerative disorders.


Assuntos
Cabelo , Melanócitos , Transdução de Sinais , Animais , Camundongos , Cabelo/citologia , Cabelo/crescimento & desenvolvimento , Folículo Piloso/citologia , Folículo Piloso/fisiologia , Receptores de Hialuronatos/metabolismo , Melanócitos/citologia , Melanócitos/metabolismo , Nevo/metabolismo , Nevo/patologia , Osteopontina/metabolismo , Células-Tronco/citologia
13.
Immunity ; 48(2): 271-285.e5, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29466757

RESUMO

Stem cells are critical for the maintenance of many tissues, but whether their integrity is maintained in the face of immunity is unclear. Here we found that cycling epithelial stem cells, including Lgr5+ intestinal stem cells, as well as ovary and mammary stem cells, were eliminated by activated T cells, but quiescent stem cells in the hair follicle and muscle were resistant to T cell killing. Immune evasion was an intrinsic property of the quiescent stem cells resulting from systemic downregulation of the antigen presentation machinery, including MHC class I and TAP proteins, and is mediated by the transactivator NLRC5. This process was reversed upon stem cell entry into the cell cycle. These studies identify a link between stem cell quiescence, antigen presentation, and immune evasion. As cancer-initiating cells can derive from stem cells, these findings may help explain how the earliest cancer cells evade immune surveillance.


Assuntos
Folículo Piloso/citologia , Evasão da Resposta Imune , Vigilância Imunológica , Células-Tronco/imunologia , Animais , Apresentação de Antígeno , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Músculos/citologia , Receptores Acoplados a Proteínas G/fisiologia , Evasão Tumoral
14.
Nature ; 594(7864): 547-552, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34108685

RESUMO

Tissue stem cells are generated from a population of embryonic progenitors through organ-specific morphogenetic events1,2. Although tissue stem cells are central to organ homeostasis and regeneration, it remains unclear how they are induced during development, mainly because of the lack of markers that exclusively label prospective stem cells. Here we combine marker-independent long-term 3D live imaging and single-cell transcriptomics to capture a dynamic lineage progression and transcriptome changes in the entire epithelium of the mouse hair follicle as it develops. We found that the precursors of different epithelial lineages were aligned in a 2D concentric manner in the basal layer of the hair placode. Each concentric ring acquired unique transcriptomes and extended to form longitudinally aligned, 3D cylindrical compartments. Prospective bulge stem cells were derived from the peripheral ring of the placode basal layer, but not from suprabasal cells (as was previously suggested3). The fate of placode cells is determined by the cell position, rather than by the orientation of cell division. We also identified 13 gene clusters: the ensemble expression dynamics of these clusters drew the entire transcriptional landscape of epithelial lineage diversification, consistent with cell lineage data. Combining these findings with previous work on the development of appendages in insects4,5, we describe the 'telescope model', a generalized model for the development of ectodermal organs in which 2D concentric zones in the placode telescope out to form 3D longitudinally aligned cylindrical compartments.


Assuntos
Linhagem da Célula , Folículo Piloso/citologia , Células-Tronco/citologia , Animais , Rastreamento de Células , Ectoderma , Embrião de Mamíferos , Células Epiteliais/citologia , Feminino , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Camundongos Transgênicos , Família Multigênica , RNA-Seq , Análise de Célula Única , Pele , Técnicas de Cultura de Tecidos , Transcriptoma , Vibrissas
15.
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
16.
Cell ; 146(5): 678-81, 2011 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-21884930

RESUMO

A recent series of papers, including Festa et al. (2011) in this issue, has revealed unexpected interdependent relationships among cell populations residing in and around the hair follicle. These interactions between different lineages of stem cells are crucial for hair follicle growth and cycling and point to a complex crosstalk in stem cell niches.


Assuntos
Adipócitos/citologia , Folículo Piloso/citologia , Pele/citologia , Células-Tronco/citologia , Animais , Feminino , Humanos , Masculino
17.
Cell ; 146(5): 761-71, 2011 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-21884937

RESUMO

In mammalian skin, multiple types of resident cells are required to create a functional tissue and support tissue homeostasis and regeneration. The cells that compose the epithelial stem cell niche for skin homeostasis and regeneration are not well defined. Here, we identify adipose precursor cells within the skin and demonstrate that their dynamic regeneration parallels the activation of skin stem cells. Functional analysis of adipocyte lineage cells in mice with defects in adipogenesis and in transplantation experiments revealed that intradermal adipocyte lineage cells are necessary and sufficient to drive follicular stem cell activation. Furthermore, we implicate PDGF expression by immature adipocyte cells in the regulation of follicular stem cell activity. These data highlight adipogenic cells as skin niche cells that positively regulate skin stem cell activity, and suggest that adipocyte lineage cells may alter epithelial stem cell function clinically.


Assuntos
Adipócitos/citologia , Folículo Piloso/citologia , Pele/citologia , Células-Tronco/citologia , Adipogenia , Animais , Feminino , Humanos , Masculino , Camundongos , Modelos Animais , Fator de Crescimento Derivado de Plaquetas/metabolismo
18.
Cell ; 144(1): 92-105, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21215372

RESUMO

Here, we exploit the hair follicle to define the point at which stem cells (SCs) become irreversibly committed along a differentiation lineage. Employing histone and nucleotide double-pulse-chase and lineage tracing, we show that the early SC descendents en route to becoming transit-amplifying cells retain stemness and slow-cycling properties and home back to the bulge niche when hair growth stops. These become the primary SCs for the next hair cycle, whereas initial bulge SCs become reserves for injury. Proliferating descendents further en route irreversibly lose their stemness, although they retain many SC markers and survive, unlike their transit-amplifying progeny. Remarkably, these progeny also home back to the bulge. Combining purification and gene expression analysis with differential ablation and functional experiments, we define critical functions for these non-SC niche residents and unveil the intriguing concept that an irreversibly committed cell in an SC lineage can become an essential contributor to the niche microenvironment.


Assuntos
Folículo Piloso/citologia , Folículo Piloso/crescimento & desenvolvimento , Nicho de Células-Tronco/metabolismo , Células-Tronco/metabolismo , Animais , Antígenos CD34/metabolismo , Diferenciação Celular , Folículo Piloso/metabolismo , Humanos , Camundongos , Pele/citologia
19.
Cell ; 145(7): 1129-41, 2011 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-21703454

RESUMO

Ciliogenesis precedes lineage-determining signaling in skin development. To understand why, we performed shRNA-mediated knockdown of seven intraflagellar transport proteins (IFTs) and conditional ablation of Ift-88 and Kif3a during embryogenesis. In both cultured keratinocytes and embryonic epidermis, all of these eliminated cilia, and many (not Kif3a) caused hyperproliferation. Surprisingly and independent of proliferation, ciliary mutants displayed defects in Notch signaling and commitment of progenitors to differentiate. Notch receptors and Notch-processing enzymes colocalized with cilia in wild-type epidermal cells. Moreover, differentiation defects in ciliary mutants were cell autonomous and rescued by activated Notch (NICD). By contrast, Shh signaling was neither operative nor required for epidermal ciliogenesis, Notch signaling, or differentiation. Rather, Shh signaling defects in ciliary mutants occurred later, arresting hair follicle morphogenesis in the skin. These findings unveil temporally and spatially distinct functions for primary cilia at the nexus of signaling, proliferation, and differentiation.


Assuntos
Diferenciação Celular , Cílios/metabolismo , Epiderme/embriologia , Epiderme/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Proteínas de Transporte/genética , Polaridade Celular , Proliferação de Células , Proteínas do Citoesqueleto/metabolismo , Células Epidérmicas , Técnicas de Silenciamento de Genes , Folículo Piloso/citologia , Proteínas Hedgehog/metabolismo , Cinese , Camundongos , Proteínas Supressoras de Tumor/metabolismo
20.
Cell ; 144(4): 577-89, 2011 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-21335239

RESUMO

The hair follicle bulge in the epidermis associates with the arrector pili muscle (APM) that is responsible for piloerection ("goosebumps"). We show that stem cells in the bulge deposit nephronectin into the underlying basement membrane, thus regulating the adhesion of mesenchymal cells expressing the nephronectin receptor, α8ß1 integrin, to the bulge. Nephronectin induces α8 integrin-positive mesenchymal cells to upregulate smooth muscle markers. In nephronectin knockout mice, fewer arrector pili muscles form in the skin, and they attach to the follicle above the bulge, where there is compensatory upregulation of the nephronectin family member EGFL6. Deletion of α8 integrin also abolishes selective APM anchorage to the bulge. Nephronectin is a Wnt target; epidermal ß-catenin activation upregulates epidermal nephronectin and dermal α8 integrin expression. Thus, bulge stem cells, via nephronectin expression, create a smooth muscle cell niche and act as tendon cells for the APM. Our results reveal a functional role for basement membrane heterogeneity in tissue patterning. PAPERCLIP:


Assuntos
Membrana Basal/citologia , Folículo Piloso/citologia , Células-Tronco/metabolismo , Animais , Membrana Basal/metabolismo , Células Epidérmicas , Epiderme/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Regulação da Expressão Gênica , Cadeias alfa de Integrinas/metabolismo , Camundongos , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA