Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros











Intervalo de año de publicación
1.
Science ; 383(6687): eadi7342, 2024 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-38452090

RESUMEN

Lineage plasticity-a state of dual fate expression-is required to release stem cells from their niche constraints and redirect them to tissue compartments where they are most needed. In this work, we found that without resolving lineage plasticity, skin stem cells cannot effectively generate each lineage in vitro nor regrow hair and repair wounded epidermis in vivo. A small-molecule screen unearthed retinoic acid as a critical regulator. Combining high-throughput approaches, cell culture, and in vivo mouse genetics, we dissected its roles in tissue regeneration. We found that retinoic acid is made locally in hair follicle stem cell niches, where its levels determine identity and usage. Our findings have therapeutic implications for hair growth as well as chronic wounds and cancers, where lineage plasticity is unresolved.


Asunto(s)
Células Madre Adultas , Plasticidad de la Célula , Epidermis , Folículo Piloso , Tretinoina , Cicatrización de Heridas , Animales , Ratones , Células Madre Adultas/citología , Células Madre Adultas/fisiología , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/fisiología , Plasticidad de la Célula/efectos de los fármacos , Plasticidad de la Célula/fisiología , Epidermis/efectos de los fármacos , Epidermis/fisiología , Folículo Piloso/citología , Folículo Piloso/efectos de los fármacos , Folículo Piloso/fisiología , Tretinoina/metabolismo , Tretinoina/farmacología , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología , Rejuvenecimiento/fisiología , Técnicas de Cultivo de Célula , Neoplasias/patología , Ratones Endogámicos C57BL
2.
Nature ; 612(7940): 555-563, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36450983

RESUMEN

Squamous cell carcinomas are triggered by marked elevation of RAS-MAPK signalling and progression from benign papilloma to invasive malignancy1-4. At tumour-stromal interfaces, a subset of tumour-initiating progenitors, the cancer stem cells, obtain increased resistance to chemotherapy and immunotherapy along this pathway5,6. The distribution and changes in cancer stem cells during progression from a benign state to invasive squamous cell carcinoma remain unclear. Here we show in mice that, after oncogenic RAS activation, cancer stem cells rewire their gene expression program and trigger self-propelling, aberrant signalling crosstalk with their tissue microenvironment that drives their malignant progression. The non-genetic, dynamic cascade of intercellular exchanges involves downstream pathways that are often mutated in advanced metastatic squamous cell carcinomas with high mutational burden7. Coupling our clonal skin HRASG12V mouse model with single-cell transcriptomics, chromatin landscaping, lentiviral reporters and lineage tracing, we show that aberrant crosstalk between cancer stem cells and their microenvironment triggers angiogenesis and TGFß signalling, creating conditions that are conducive for hijacking leptin and leptin receptor signalling, which in turn launches downstream phosphoinositide 3-kinase (PI3K)-AKT-mTOR signalling during the benign-to-malignant transition. By functionally examining each step in this pathway, we reveal how dynamic temporal crosstalk with the microenvironment orchestrated by the stem cells profoundly fuels this path to malignancy. These insights suggest broad implications for cancer therapeutics.


Asunto(s)
Carcinoma de Células Escamosas , Genes ras , Células Madre Neoplásicas , Transducción de Señal , Microambiente Tumoral , Proteínas ras , Animales , Ratones , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Leptina/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neovascularización Patológica , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
3.
Elife ; 92020 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-32845239

RESUMEN

N6-methyladenosine is the most prominent RNA modification in mammals. Here, we study mouse skin embryogenesis to tackle m6A's functions and physiological importance. We first landscape the m6A modifications on skin epithelial progenitor mRNAs. Contrasting with in vivo ribosomal profiling, we unearth a correlation between m6A modification in coding sequences and enhanced translation, particularly of key morphogenetic signaling pathways. Tapping physiological relevance, we show that m6A loss profoundly alters these cues and perturbs cellular fate choices and tissue architecture in all skin lineages. By single-cell transcriptomics and bioinformatics, both signaling and canonical translation pathways show significant downregulation after m6A loss. Interestingly, however, many highly m6A-modified mRNAs are markedly upregulated upon m6A loss, and they encode RNA-methylation, RNA-processing and RNA-metabolism factors. Together, our findings suggest that m6A functions to enhance translation of key morphogenetic regulators, while also destabilizing sentinel mRNAs that are primed to activate rescue pathways when m6A levels drop.


Asunto(s)
Adenosina/análogos & derivados , Organogénesis/genética , ARN Mensajero , Piel , Adenosina/química , Adenosina/genética , Adenosina/metabolismo , Animales , Biología Computacional , Femenino , Metilación , Metiltransferasas/metabolismo , Ratones , ARN Mensajero/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Piel/química , Piel/metabolismo , Transcriptoma/genética
4.
Proc Natl Acad Sci U S A ; 117(10): 5339-5350, 2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-32094197

RESUMEN

Aging manifests with architectural alteration and functional decline of multiple organs throughout an organism. In mammals, aged skin is accompanied by a marked reduction in hair cycling and appearance of bald patches, leading researchers to propose that hair follicle stem cells (HFSCs) are either lost, differentiate, or change to an epidermal fate during aging. Here, we employed single-cell RNA-sequencing to interrogate aging-related changes in the HFSCs. Surprisingly, although numbers declined, aging HFSCs were present, maintained their identity, and showed no overt signs of shifting to an epidermal fate. However, they did exhibit prevalent transcriptional changes particularly in extracellular matrix genes, and this was accompanied by profound structural perturbations in the aging SC niche. Moreover, marked age-related changes occurred in many nonepithelial cell types, including resident immune cells, sensory neurons, and arrector pili muscles. Each of these SC niche components has been shown to influence HF regeneration. When we performed skin injuries that are known to mobilize young HFSCs to exit their niche and regenerate HFs, we discovered that aged skin is defective at doing so. Interestingly, however, in transplantation assays in vivo, aged HFSCs regenerated HFs when supported with young dermis, while young HFSCs failed to regenerate HFs when combined with aged dermis. Together, our findings highlight the importance of SC:niche interactions and favor a model where youthfulness of the niche microenvironment plays a dominant role in dictating the properties of its SCs and tissue health and fitness.


Asunto(s)
Folículo Piloso/fisiología , Regeneración/fisiología , Envejecimiento de la Piel/fisiología , Nicho de Células Madre/fisiología , Células Madre/fisiología , Animales , Dermis/fisiología , Células Epidérmicas/fisiología , Epidermis/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculos/fisiología , Repitelización , Regeneración/genética , Células Receptoras Sensoriales/fisiología , Envejecimiento de la Piel/genética , Nicho de Células Madre/genética , Trasplante de Células Madre , Transcriptoma , Cicatrización de Heridas/genética , Cicatrización de Heridas/fisiología
5.
Cell ; 177(5): 1172-1186.e14, 2019 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-31031009

RESUMEN

Our bodies are equipped with powerful immune surveillance to clear cancerous cells as they emerge. How tumor-initiating stem cells (tSCs) that form and propagate cancers equip themselves to overcome this barrier remains poorly understood. To tackle this problem, we designed a skin cancer model for squamous cell carcinoma (SCC) that can be effectively challenged by adoptive cytotoxic T cell transfer (ACT)-based immunotherapy. Using single-cell RNA sequencing (RNA-seq) and lineage tracing, we found that transforming growth factor ß (TGF-ß)-responding tSCs are superior at resisting ACT and form the root of tumor relapse. Probing mechanism, we discovered that during malignancy, tSCs selectively acquire CD80, a surface ligand previously identified on immune cells. Moreover, upon engaging cytotoxic T lymphocyte antigen-4 (CTLA4), CD80-expressing tSCs directly dampen cytotoxic T cell activity. Conversely, upon CTLA4- or TGF-ß-blocking immunotherapies or Cd80 ablation, tSCs become vulnerable, diminishing tumor relapse after ACT treatment. Our findings place tSCs at the crux of how immune checkpoint pathways are activated.


Asunto(s)
Traslado Adoptivo , Carcinoma de Células Escamosas/inmunología , Inmunidad Celular , Vigilancia Inmunológica , Células Madre Neoplásicas/inmunología , Neoplasias Cutáneas/inmunología , Linfocitos T/inmunología , Animales , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/terapia , Línea Celular Tumoral , Humanos , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/inmunología , Células Madre Neoplásicas/patología , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia , Linfocitos T/patología
6.
Nature ; 550(7677): 475-480, 2017 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-29045388

RESUMEN

The skin barrier is the body's first line of defence against environmental assaults, and is maintained by epithelial stem cells (EpSCs). Despite the vulnerability of EpSCs to inflammatory pressures, neither the primary response to inflammation nor its enduring consequences are well understood. Here we report a prolonged memory to acute inflammation that enables mouse EpSCs to hasten barrier restoration after subsequent tissue damage. This functional adaptation does not require skin-resident macrophages or T cells. Instead, EpSCs maintain chromosomal accessibility at key stress response genes that are activated by the primary stimulus. Upon a secondary challenge, genes governed by these domains are transcribed rapidly. Fuelling this memory is Aim2, which encodes an activator of the inflammasome. The absence of AIM2 or its downstream effectors, caspase-1 and interleukin-1ß, erases the ability of EpSCs to recollect inflammation. Although EpSCs benefit from inflammatory tuning by heightening their responsiveness to subsequent stressors, this enhanced sensitivity probably increases their susceptibility to autoimmune and hyperproliferative disorders, including cancer.


Asunto(s)
Células Epiteliales/citología , Inflamación/genética , Inflamación/patología , Piel/citología , Piel/patología , Células Madre/citología , Cicatrización de Heridas/fisiología , Aminoquinolinas/farmacología , Animales , Enfermedades Autoinmunes/patología , Caspasa 1/metabolismo , Linaje de la Célula , Cromatina/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Imiquimod , Inflamasomas/metabolismo , Inflamación/inducido químicamente , Inflamación/inmunología , Interleucina-1beta/metabolismo , Macrófagos , Ratones , Neoplasias/patología , Regeneración/efectos de los fármacos , Regeneración/genética , Piel/efectos de los fármacos , Piel/inmunología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Estrés Fisiológico/genética , Linfocitos T , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/genética
7.
Cell ; 169(4): 636-650.e14, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28434617

RESUMEN

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.


Asunto(s)
Carcinoma de Células Escamosas/patología , Linaje de la Célula , Células Epidérmicas , Folículo Piloso/citología , Neoplasias Cutáneas/patología , Piel/citología , Células Madre/metabolismo , Animales , Línea Celular Tumoral , Cromatina/metabolismo , Epidermis/metabolismo , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Cutáneas/metabolismo , Factores de Transcripción/metabolismo , Transcriptoma , Trasplante Heterólogo , Cicatrización de Heridas
8.
Cell ; 167(5): 1323-1338.e14, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27863246

RESUMEN

Aged skin heals wounds poorly, increasing susceptibility to infections. Restoring homeostasis after wounding requires the coordinated actions of epidermal and immune cells. Here we find that both intrinsic defects and communication with immune cells are impaired in aged keratinocytes, diminishing their efficiency in restoring the skin barrier after wounding. At the wound-edge, aged keratinocytes display reduced proliferation and migration. They also exhibit a dampened ability to transcriptionally activate epithelial-immune crosstalk regulators, including a failure to properly activate/maintain dendritic epithelial T cells (DETCs), which promote re-epithelialization following injury. Probing mechanism, we find that aged keratinocytes near the wound edge don't efficiently upregulate Skints or activate STAT3. Notably, when epidermal Stat3, Skints, or DETCs are silenced in young skin, re-epithelialization following wounding is perturbed. These findings underscore epithelial-immune crosstalk perturbations in general, and Skints in particular, as critical mediators in the age-related decline in wound-repair.


Asunto(s)
Envejecimiento/fisiología , Subgrupos Linfocitarios/citología , Transducción de Señal , Cicatrización de Heridas , Animales , Interleucina-6/administración & dosificación , Queratinocitos/metabolismo , Ratones , Piel/citología , Fenómenos Fisiológicos de la Piel , Cicatrización de Heridas/efectos de los fármacos
9.
Nature ; 521(7552): 366-70, 2015 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-25799994

RESUMEN

Adult stem cells occur in niches that balance self-renewal with lineage selection and progression during tissue homeostasis. Following injury, culture or transplantation, stem cells outside their niche often display fate flexibility. Here we show that super-enhancers underlie the identity, lineage commitment and plasticity of adult stem cells in vivo. Using hair follicle as a model, we map the global chromatin domains of hair follicle stem cells and their committed progenitors in their native microenvironments. We show that super-enhancers and their dense clusters ('epicentres') of transcription factor binding sites undergo remodelling upon lineage progression. New fate is acquired by decommissioning old and establishing new super-enhancers and/or epicentres, an auto-regulatory process that abates one master regulator subset while enhancing another. We further show that when outside their niche, either in vitro or in wound-repair, hair follicle stem cells dynamically remodel super-enhancers in response to changes in their microenvironment. Intriguingly, some key super-enhancers shift epicentres, enabling their genes to remain active and maintain a transitional state in an ever-changing transcriptional landscape. Finally, we identify SOX9 as a crucial chromatin rheostat of hair follicle stem cell super-enhancers, and provide functional evidence that super-enhancers are dynamic, dense transcription-factor-binding platforms which are acutely sensitive to pioneer master regulators whose levels define not only spatial and temporal features of lineage-status but also stemness, plasticity in transitional states and differentiation.


Asunto(s)
Adaptación Fisiológica , Células Madre Adultas/citología , Diferenciación Celular/genética , Linaje de la Célula/genética , Elementos de Facilitación Genéticos/genética , Folículo Piloso/citología , Factor de Transcripción SOX9/metabolismo , Células Madre Adultas/metabolismo , Animales , Secuencia de Bases , Cromatina/genética , Cromatina/metabolismo , Femenino , Ratones , Especificidad de Órganos , Nicho de Células Madre , Factores de Tiempo
10.
Cell ; 150(1): 136-50, 2012 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-22770217

RESUMEN

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


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/fisiología , Homeostasis , Glándulas Sudoríparas/citología , Cicatrización de Heridas , Células Madre Adultas/clasificación , Animales , Células Epidérmicas , Epidermis/fisiología , Femenino , Humanos , Glándulas Mamarias Animales/citología , Ratones , Morfogénesis , Células Madre Multipotentes/fisiología , Análisis de Componente Principal , Trasplante de Células Madre , Glándulas Sudoríparas/embriología , Glándulas Sudoríparas/fisiología
11.
Nat Genet ; 41(10): 1068-75, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19718027

RESUMEN

Single-layered embryonic skin either stratifies to form epidermis or responds to Wnt signaling (stabilized beta-catenin) to form hair follicles. Postnatally, stem cells continue to differentially use Wnt signaling in long-term tissue homeostasis. We have discovered that embryonic progenitor cells and postnatal hair follicle stem cells coexpress Tcf3 and Tcf4, which can act as transcriptional activators or repressors. Using loss-of-function studies and transcriptional analyses, we uncovered consequences to the absence of Tcf3 and Tcf4 in skin that only partially overlap with those caused by beta-catenin deficiency. We established roles for Tcf3 and Tcf4 in long-term maintenance and wound repair of both epidermis and hair follicles, suggesting that Tcf proteins have both Wnt-dependent and Wnt-independent roles in lineage determination.


Asunto(s)
Células Epiteliales/metabolismo , Homeostasis , Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal , Piel/metabolismo , Factores de Transcripción TCF/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Diferenciación Celular , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Células Epiteliales/citología , Femenino , Masculino , Ratones , Ratones Noqueados , Ratones Desnudos , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción TCF/deficiencia , Factores de Transcripción TCF/genética , Factor de Transcripción 4 , Proteína 1 Similar al Factor de Transcripción 7 , Proteínas Wnt/metabolismo , Cicatrización de Heridas , beta Catenina/deficiencia , beta Catenina/metabolismo
12.
Cell Stem Cell ; 3(1): 33-43, 2008 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-18593557

RESUMEN

In adult skin, epithelial hair follicle stem cells (SCs) reside in a quiescent niche and are essential for cyclic bouts of hair growth. Niche architecture becomes pronounced postnatally at the start of the first hair cycle. Whether SCs exist or function earlier is unknown. Here we show that slow-cycling cells appear early in skin development, express SC markers, and later give rise to the adult SC population. To test whether these early slow-cycling cells function as SCs, we use Sox9-Cre for genetic marking and K14-Cre to embryonically ablate Sox9, an essential adult SC gene. We find that the progeny of Sox9-expressing cells contribute to all skin epithelial lineages and Sox9 is required for SC specification. In the absence of early SCs, hair follicle and sebaceous gland morphogenesis is blocked, and epidermal wound repair is compromised. These findings establish the existence of early hair follicle SCs and reveal their physiological importance in tissue morphogenesis.


Asunto(s)
Folículo Piloso/fisiología , Piel/crecimiento & desarrollo , Células Madre/citología , Células Madre/fisiología , Adulto , Animales , Biomarcadores , Ciclo Celular , Células Epiteliales/citología , Células Epiteliales/fisiología , Folículo Piloso/citología , Proteínas del Grupo de Alta Movilidad/fisiología , Humanos , Integrasas/metabolismo , Ratones , Morfogénesis , Factor de Transcripción SOX9 , Piel/citología , Fenómenos Fisiológicos de la Piel , Trasplante de Células Madre , Factores de Transcripción/fisiología , Cicatrización de Heridas
13.
Genes Dev ; 22(4): 543-57, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18281466

RESUMEN

Hair follicle (HF) formation is initiated when epithelial stem cells receive cues from specialized mesenchymal dermal papilla (DP) cells. In culture, DP cells lose their HF-inducing properties, but during hair growth in vivo, they reside within the HF bulb and instruct surrounding epithelial progenitors to orchestrate the complex hair differentiation program. To gain insights into the molecular program that maintains DP cell fate, we previously purified DP cells and four neighboring populations and defined their cell-type-specific molecular signatures. Here, we exploit this information to show that the bulb microenvironment is rich in bone morphogenetic proteins (BMPs) that act on DP cells to maintain key signature features in vitro and hair-inducing activity in vivo. By employing a novel in vitro/in vivo hybrid knockout assay, we ablate BMP receptor 1a in purified DP cells. When DPs cannot receive BMP signals, they lose signature characteristics in vitro and fail to generate HFs when engrafted with epithelial stem cells in vivo. These results reveal that BMP signaling, in addition to its key role in epithelial stem cell maintenance and progenitor cell differentiation, is essential for DP cell function, and suggest that it is a critical feature of the complex epithelial-mesenchymal cross-talk necessary to make hair.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/fisiología , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Dermis/citología , Dermis/metabolismo , Células Epiteliales/metabolismo , Folículo Piloso/metabolismo , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Proteínas Morfogenéticas Óseas/genética , Calcificación Fisiológica , Proliferación Celular , Células Cultivadas , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Folículo Piloso/citología , Hibridación in Situ , Integrasas/metabolismo , Ligandos , Masculino , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Ratones Desnudos , Osteoblastos , Osteogénesis , Receptores de Superficie Celular/metabolismo , Células Madre/citología , Células Madre/metabolismo
14.
Cell ; 132(2): 299-310, 2008 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-18243104

RESUMEN

Quiescent adult stem cells reside in specialized niches where they become activated to proliferate and differentiate during tissue homeostasis and injury. How stem cell quiescence is governed is poorly understood. We report here that NFATc1 is preferentially expressed by hair follicle stem cells in their niche, where its expression is activated by BMP signaling upstream and it acts downstream to transcriptionally repress CDK4 and maintain stem cell quiescence. As stem cells become activated during hair growth, NFATc1 is downregulated, relieving CDK4 repression and activating proliferation. When calcineurin/NFATc1 signaling is suppressed, pharmacologically or via complete or conditional NFATc1 gene ablation, stem cells are activated prematurely, resulting in precocious follicular growth. Our findings may explain why patients receiving cyclosporine A for immunosuppressive therapy display excessive hair growth, and unveil a functional role for calcium-NFATc1-CDK4 circuitry in governing stem cell quiescence.


Asunto(s)
Proliferación Celular , Factores de Transcripción NFATC/metabolismo , Piel/citología , Células Madre/citología , Células Madre/fisiología , Animales , Antígenos CD34/metabolismo , Biomarcadores , Núcleo Celular/metabolismo , Células Cultivadas , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Ciclosporina/farmacología , Regulación hacia Abajo , Embrión de Mamíferos , Eliminación de Gen , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Folículo Piloso/citología , Folículo Piloso/fisiología , Inmunohistoquímica , Inmunosupresores/farmacología , Ratones , Ratones Noqueados , Ratones Desnudos , Morfogénesis , Factores de Transcripción NFATC/genética , ARN Mensajero/metabolismo , Retroviridae/genética , Piel/embriología , Trasplante de Piel , Células Madre/efectos de los fármacos , Factores de Transcripción/metabolismo , Transgenes , Trasplante Homólogo
15.
Cancer Cell ; 12(4): 313-27, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17936557

RESUMEN

Although TGFbeta is a potent inhibitor of proliferation, epithelia lacking the essential receptor (TbetaRII) for TGFbeta signaling display normal tissue homeostasis. By studying asymptomatic TbetaRII-deficient stratified epithelia, we show that tissue homeostasis is maintained by balancing hyperproliferation with elevated apoptosis. Moreover, rectal and genital epithelia, which are naturally proliferative, develop spontaneous squamous cell carcinomas with age when TbetaRII is absent. This progression is associated with a reduction in apoptosis and can be accelerated in phenotypically normal epidermis by oncogenic mutations in Ras. We show that TbetaRII deficiency leads to enhanced keratinocyte motility and integrin-FAK-Src signaling. Together, these mechanisms provide a molecular framework to account for many of the characteristics of TbetaRII-deficient invasive SQCCs.


Asunto(s)
Apoptosis , Carcinoma de Células Escamosas/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Células Epiteliales/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Neoplasias del Ano/metabolismo , Neoplasias del Ano/patología , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Movimiento Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Células Cultivadas , Células Epiteliales/patología , Matriz Extracelular/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Homeostasis , Humanos , Integrinas/metabolismo , Queratina-14/genética , Queratinocitos/metabolismo , Queratinocitos/patología , Masculino , Ratones , Ratones Noqueados , Mutación , Invasividad Neoplásica , Papiloma/metabolismo , Papiloma/patología , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/genética , Piel/metabolismo , Piel/patología , Piel/fisiopatología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Factores de Tiempo , Neoplasias Urogenitales/metabolismo , Neoplasias Urogenitales/patología , Cicatrización de Heridas , Proteínas ras/genética , Proteínas ras/metabolismo , Familia-src Quinasas/metabolismo
16.
Proc Natl Acad Sci U S A ; 104(24): 10063-8, 2007 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-17553962

RESUMEN

During the hair cycle, follicle stem cells (SCs) residing in a specialized niche called the "bulge" undergo bouts of quiescence and activation to cyclically regenerate new hairs. Developmental studies have long implicated the canonical bone morphogenetic protein (BMP) pathway in hair follicle (HF) determination and differentiation, but how BMP signaling functions in the hair follicle SC niche remains unknown. Here, we use loss and gain of function studies to manipulate BMP signaling in the SC niche. We show that when the Bmpr1a gene is conditionally ablated, otherwise quiescent SCs are activated to proliferate, causing an expansion of the niche and loss of slow-cycling cells. Surprisingly, follicle SCs are not lost, however, but rather, they generate long-lived, tumor-like branches that express Sox4, Lhx2, and Sonic Hedgehog but fail to terminally differentiate to make hair. A key component of BMPR1A-deficient SCs is their elevated levels of both Lef1 and beta-catenin, which form a bipartite transcription complex required for initiation of the hair cycle. Although beta-catenin can be stabilized by Wnt signaling, we show that BMPR1A deficiency enhances beta-catenin stabilization in the niche through a pathway involving PTEN inhibition and PI3K/AKT activation. Conversely, sustained BMP signaling in the SC niche blocks activation and promotes premature hair follicle differentiation. Together, these studies reveal the importance of balancing BMP signaling in the SC niche.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/deficiencia , Proteínas Morfogenéticas Óseas/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/fisiología , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Proteínas Morfogenéticas Óseas/genética , Proliferación Celular , Regulación de la Expresión Génica , Folículo Piloso/citología , Folículo Piloso/fisiología , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas con Homeodominio LIM , Factor de Unión 1 al Potenciador Linfoide/análisis , Factor de Unión 1 al Potenciador Linfoide/genética , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Ratones , Ratones Transgénicos , Fosfohidrolasa PTEN/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción SOXC , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , beta Catenina/análisis , beta Catenina/genética , beta Catenina/metabolismo
17.
Science ; 312(5782): 1946-9, 2006 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-16809539

RESUMEN

During embryogenesis, stem cells are set aside to fuel the postnatal hair cycle and repair the epidermis after injury. To define how hair follicle stem cells are specified and maintained in an undifferentiated state, we developed a strategy to isolate and transcriptionally profile embryonic hair progenitors in mice. We identified Lhx2 as a transcription factor positioned downstream of signals necessary to specify hair follicle stem cells, but upstream from signals required to drive activated stem cells to terminally differentiate. Using gain- and loss-of-function studies, we uncovered a role for Lhx2 in maintaining the growth and undifferentiated properties of hair follicle progenitors.


Asunto(s)
Folículo Piloso/citología , Proteínas de Homeodominio/fisiología , Células Madre/fisiología , Factores de Transcripción/fisiología , Animales , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Células Epidérmicas , Epidermis/embriología , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Cabello/embriología , Cabello/crecimiento & desarrollo , Folículo Piloso/embriología , Folículo Piloso/fisiología , Proteínas de Homeodominio/genética , Proteínas con Homeodominio LIM , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Morfogénesis , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal , Trasplante de Piel , Factores de Transcripción/genética , Regulación hacia Arriba
18.
J Cell Biol ; 172(3): 409-21, 2006 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-16449191

RESUMEN

AP-2 transcription factors have been implicated in epidermal biology, but their functional significance has remained elusive. Using conditional knockout technology, we show that AP-2alpha is essential for governing the balance between growth and differentiation in epidermis. In vivo, epidermis lacking AP-2alpha exhibits elevated expression of the epidermal growth factor receptor (EGFR) in the differentiating layers, resulting in hyperproliferation when the receptors are activated. Chromatin immunoprecipitation and promoter activity assays identify EGFR as a direct target gene for AP-2alpha repression, and, in the absence of AP-2alpha, this is manifested primarily in excessive EGF-dependent phosphoinositol-3 kinase/Akt activity. Together, our findings unveil a hitherto unrecognized repressive role for AP-2alpha in governing EGFR gene transcription as cells exit the basal layer and withdraw from the cell cycle. These results provide insights into why elevated AP-2alpha levels are often associated with terminal differentiation and why tumor cells often display reduced AP-2alpha and elevated EGFR proteins.


Asunto(s)
Receptores ErbB/fisiología , Transducción de Señal/fisiología , Factor de Transcripción AP-2/fisiología , Animales , Animales Recién Nacidos , Calcio/farmacología , Caspasa 3 , Caspasas/metabolismo , Proliferación Celular/efectos de los fármacos , Inmunoprecipitación de Cromatina , Cromonas/farmacología , ADN/genética , ADN/metabolismo , Dermis/metabolismo , Embrión de Mamíferos/metabolismo , Inhibidores Enzimáticos/farmacología , Células Epidérmicas , Factor de Crecimiento Epidérmico/farmacología , Epidermis/fisiología , Receptores ErbB/genética , Receptores ErbB/metabolismo , Expresión Génica/genética , Regulación de la Expresión Génica , Enfermedades del Cabello/genética , Enfermedades del Cabello/patología , Integrasas/genética , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Queratinas/metabolismo , Antígeno Ki-67/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Transgénicos , Morfolinas/farmacología , Fosforilación , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinazolinas , Transducción de Señal/genética , Piel/efectos de los fármacos , Piel/metabolismo , Piel/patología , Anomalías Cutáneas/genética , Anomalías Cutáneas/patología , Acetato de Tetradecanoilforbol/farmacología , Factor de Transcripción AP-2/genética , Factor de Transcripción AP-2/metabolismo , Factor de Crecimiento Transformador alfa/genética , Tirfostinos/farmacología
19.
Cell ; 118(5): 635-48, 2004 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-15339667

RESUMEN

In adult skin, each hair follicle contains a reservoir of stem cells (the bulge), which can be mobilized to regenerate the new follicle with each hair cycle and to reepithelialize epidermis during wound repair. Here we report new methods that permit their clonal analyses and engraftment and demonstrate the two defining features of stem cells, namely self-renewal and multipotency. We also show that, within the bulge, there are two distinct populations, one of which maintains basal lamina contact and temporally precedes the other, which is suprabasal and arises only after the start of the first postnatal hair cycle. This spatial distinction endows them with discrete transcriptional programs, but surprisingly, both populations are growth inhibited in the niche but can self-renew in vitro and make epidermis and hair when grafted. These findings suggest that the niche microenvironment imposes intrinsic "stemness" features without restricting the establishment of epithelial polarity and changes in gene expression.


Asunto(s)
Diferenciación Celular/fisiología , Epidermis/crecimiento & desarrollo , Células Epiteliales/metabolismo , Folículo Piloso/crecimiento & desarrollo , Células Madre Multipotentes/metabolismo , Regeneración/fisiología , Células 3T3 , Animales , Membrana Basal/fisiología , Polaridad Celular/genética , Células Clonales/citología , Células Clonales/metabolismo , Células Epidérmicas , Epidermis/metabolismo , Células Epiteliales/clasificación , Células Epiteliales/citología , Regulación del Desarrollo de la Expresión Génica/genética , Folículo Piloso/citología , Folículo Piloso/metabolismo , Ratones , Ratones Transgénicos , Células Madre Multipotentes/clasificación , Células Madre Multipotentes/citología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcripción Genética/genética
20.
Development ; 131(2): 263-74, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14668413

RESUMEN

The roles of Lef/Tcf proteins in determining cell fate characteristics have been described in many contexts during vertebrate embryogenesis, organ and tissue homeostasis, and cancer formation. Although much of the accumulated work on these proteins involves their ability to transactivate target genes when stimulated by beta-catenin, Lef/Tcf proteins can repress target genes in the absence of stabilized beta-catenin. By ablating Tcf3 function, we have uncovered an important requirement for a repressor function of Lef/Tcf proteins during early mouse development. Tcf3-/- embryos proceed through gastrulation to form mesoderm, but they develop expanded and often duplicated axial mesoderm structures, including nodes and notochords. These duplications are preceded by ectopic expression of Foxa2, an axial mesoderm gene involved in node specification, with a concomitant reduction in Lefty2, a marker for lateral mesoderm. By contrast, expression of a beta-catenin-dependent, Lef/Tcf reporter (TOPGal), is not ectopically activated but is faithfully maintained in the primitive streak. Taken together, these data reveal a unique requirement for Tcf3 repressor function in restricting induction of the anterior-posterior axis.


Asunto(s)
Tipificación del Cuerpo/fisiología , Proteínas de Unión al ADN/fisiología , Factores de Transcripción/fisiología , Proteínas de Pez Cebra , Animales , Tipificación del Cuerpo/genética , Sistema Nervioso Central/anomalías , Proteínas del Citoesqueleto/fisiología , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Femenino , Gástrula/metabolismo , Regulación del Desarrollo de la Expresión Génica , Marcación de Gen , Mesodermo/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fenotipo , Embarazo , Proteínas Proto-Oncogénicas/fisiología , Transducción de Señal , Transactivadores/fisiología , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Proteínas Wnt , beta Catenina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA