Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Cell ; 186(2): 305-326.e27, 2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36638792

RESUMEN

All living things experience an increase in entropy, manifested as a loss of genetic and epigenetic information. In yeast, epigenetic information is lost over time due to the relocalization of chromatin-modifying proteins to DNA breaks, causing cells to lose their identity, a hallmark of yeast aging. Using a system called "ICE" (inducible changes to the epigenome), we find that the act of faithful DNA repair advances aging at physiological, cognitive, and molecular levels, including erosion of the epigenetic landscape, cellular exdifferentiation, senescence, and advancement of the DNA methylation clock, which can be reversed by OSK-mediated rejuvenation. These data are consistent with the information theory of aging, which states that a loss of epigenetic information is a reversible cause of aging.


Asunto(s)
Envejecimiento , Epigénesis Genética , Animales , Envejecimiento/genética , Metilación de ADN , Epigenoma , Mamíferos/genética , Nucleoproteínas , Saccharomyces cerevisiae/genética
3.
Biochem Biophys Res Commun ; 596: 29-35, 2022 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-35108651

RESUMEN

Thymic dendritic cells (DCs) promote immune tolerance by regulating negative selection of autoreactive T cells in the thymus. How DC homing to the thymus is transcriptionally regulated is still unclear. Microphthalmia-associated transcription factor (Mitf) is broadly expressed and plays essential roles in the hematopoietic system. Here, we used Mitf-mutated mice (Mitfvit/vit) and found enlargement of the thymus and expansion of CD4/CD8 double-positive T cells. Mitf was highly expressed in a subset of thymic DCs among the hematopoietic system. Genetic mutation or pharmacological inhibition of Mitf in DCs decreased the expression levels of Itga4, which are critical molecules for the homing of DCs to the thymus. Further, inhibition of Mitf decreased thymic DC number. These results suggest a pivotal role of Mitf in the maintenance of T cell differentiation by regulating the homing of DC subsets within the thymus.


Asunto(s)
Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Factor de Transcripción Asociado a Microftalmía/inmunología , Linfocitos T/inmunología , Timo/inmunología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/genética , Células Cultivadas , Células Dendríticas/metabolismo , Citometría de Flujo , Regulación de la Expresión Génica/inmunología , Hiperplasia , Integrina alfa4/genética , Integrina alfa4/inmunología , Integrina alfa4/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/metabolismo , Timo/metabolismo , Timo/patología
4.
Nat Aging ; 2(2): 105-114, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-37117756

RESUMEN

Canonically, hormones are produced in the endocrine organs and delivered to target tissues. However, for steroids, the concept of tissue intracrinology, whereby hormones are produced in the tissues where they exert their effect without release into circulation, has been proposed, but its role in physiology/disease remains unclear. The meibomian glands in the eyelids produce oil to prevent tear evaporation, which reduces with aging. Here, we demonstrate that (re)activation of local intracrine activity through nicotinamide adenine dinucleotide (NAD+)-dependent circadian 3ß-hydroxyl-steroid dehydrogenase (3ß-HSD) activity ameliorates age-associated meibomian gland dysfunction and accompanying evaporative dry eye disease. Genetic ablation of 3ß-HSD nullified local steroidogenesis and led to atrophy of the meibomian gland. Conversely, reactivation of 3ß-HSD activity by boosting its coenzyme NAD+ availability improved glandular cell proliferation and alleviated the dry eye disease phenotype. Both women and men express 3ß-HSD in the meibomian gland. Enhancing local steroidogenesis may help combat age-associated meibomian gland dysfunction.


Asunto(s)
Síndromes de Ojo Seco , Disfunción de la Glándula de Meibomio , Femenino , Humanos , NAD , Glándulas Tarsales , Lágrimas/fisiología , Esteroides , Hormonas
5.
Dev Cell ; 56(24): 3309-3320.e5, 2021 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-34932948

RESUMEN

Maintaining genomic integrity and stability is crucial for life; yet, no tissue-driven mechanism that robustly safeguards the epithelial genome has been discovered. Epidermal stem cells (EpiSCs) continuously replenish the stratified layers of keratinocytes that protect organisms against various environmental stresses. To study the dynamics of DNA-damaged cells in tissues, we devised an in vivo fate tracing system for EpiSCs with DNA double-strand breaks (DSBs) and demonstrated that those cells exit from their niches. The clearance of EpiSCs with DSBs is caused by selective differentiation and delamination through the DNA damage response (DDR)-p53-Notch/p21 axis, with the downregulation of ITGB1. Moreover, concomitant enhancement of symmetric cell divisions of surrounding stem cells indicates that the selective elimination of cells with DSBs is coupled with the augmented clonal expansion of intact stem cells. These data collectively demonstrate that tissue autonomy through the dynamic coupling of cell-autonomous and non-cell-autonomous mechanisms coordinately maintains the genomic quality of the epidermis.


Asunto(s)
Epidermis/metabolismo , Genoma , Células Madre/citología , Animales , Apoptosis/genética , Diferenciación Celular/genética , División Celular/genética , Proliferación Celular/genética , Células Clonales , Roturas del ADN de Doble Cadena , Daño del ADN/genética , Reparación del ADN/genética , Humanos , Integrina beta1/metabolismo , Ratones Endogámicos C57BL , Modelos Biológicos , Receptores Notch/metabolismo , Transducción de Señal/genética , Nicho de Células Madre , Células Madre/metabolismo
6.
J Cell Biol ; 220(11)2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34550317

RESUMEN

Skin regenerative capacity declines with age, but the underlying mechanisms are largely unknown. Here we demonstrate a functional link between epidermal growth factor receptor (EGFR) signaling and type XVII collagen (COL17A1) proteolysis on age-associated alteration of keratinocyte stem cell dynamics in skin regeneration. Live-imaging and computer simulation experiments predicted that human keratinocyte stem cell motility is coupled with self-renewal and epidermal regeneration. Receptor tyrosine kinase array identified the age-associated decline of EGFR signaling in mouse skin wound healing. Culture experiments proved that EGFR activation drives human keratinocyte stem cell motility with increase of COL17A1 by inhibiting its proteolysis through the secretion of tissue inhibitor of metalloproteinases 1 (TIMP1). Intriguingly, COL17A1 directly regulated keratinocyte stem cell motility and collective cell migration by coordinating actin and keratin filament networks. We conclude that EGFR-COL17A1 axis-mediated keratinocyte stem cell motility drives epidermal regeneration, which provides a novel therapeutic approach for age-associated impaired skin regeneration.


Asunto(s)
Autoantígenos/metabolismo , Movimiento Celular/fisiología , Colágenos no Fibrilares/metabolismo , Regeneración/fisiología , Piel/metabolismo , Células 3T3 , Animales , Línea Celular , Células Epidérmicas/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Folículo Piloso/metabolismo , Humanos , Queratinocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteolisis , Transducción de Señal/fisiología , Células Madre/metabolismo , Cicatrización de Heridas/fisiología , Colágeno Tipo XVII
7.
Cell Rep ; 36(5): 109492, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34348144

RESUMEN

Early differential diagnosis between malignant and benign tumors and their underlying intrinsic differences are the most critical issues for life-threatening cancers. To study whether human acral melanomas, deadly cancers that occur on non-hair-bearing skin, have distinct origins that underlie their invasive capability, we develop fate-tracing technologies of melanocyte stem cells in sweat glands (glandular McSCs) and in melanoma models in mice and compare the cellular dynamics with human melanoma. Herein, we report that glandular McSCs self-renew to expand their migratory progeny in response to genotoxic stress and trauma to generate invasive melanomas in mice that mimic human acral melanomas. The analysis of melanocytic lesions in human volar skin reveals that genetically unstable McSCs expand in sweat glands and in the surrounding epidermis in melanomas but not in nevi. The detection of such cell spreading dynamics provides an innovative method for an early differential diagnosis of acral melanomas from nevi.


Asunto(s)
Movimiento Celular , Melanoma/patología , Nevo/patología , Células Madre/patología , Animales , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Ciclina D1/metabolismo , Modelos Animales de Enfermedad , Epidermis/patología , Epidermis/efectos de la radiación , Amplificación de Genes , Inestabilidad Genómica/efectos de la radiación , Melanocitos/patología , Melanocitos/efectos de la radiación , Melanoma/diagnóstico , Ratones Endogámicos C57BL , Factores de Riesgo , Piel/patología , Piel/efectos de la radiación , Pigmentación de la Piel/efectos de la radiación , Glándulas Sudoríparas/efectos de la radiación , Rayos Ultravioleta
8.
Nature ; 595(7866): 266-271, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34163066

RESUMEN

Obesity is a worldwide epidemic that predisposes individuals to many age-associated diseases, but its exact effects on organ dysfunction are largely unknown1. Hair follicles-mini-epithelial organs that grow hair-are miniaturized by ageing to cause hair loss through the depletion of hair follicle stem cells (HFSCs)2. Here we report that obesity-induced stress, such as that induced by a high-fat diet (HFD), targets HFSCs to accelerate hair thinning. Chronological gene expression analysis revealed that HFD feeding for four consecutive days in young mice directed activated HFSCs towards epidermal keratinization by generating excess reactive oxygen species, but did not reduce the pool of HFSCs. Integrative analysis using stem cell fate tracing, epigenetics and reverse genetics showed that further feeding with an HFD subsequently induced lipid droplets and NF-κB activation within HFSCs via autocrine and/or paracrine IL-1R signalling. These integrated factors converge on the marked inhibition of Sonic hedgehog (SHH) signal transduction in HFSCs, thereby further depleting lipid-laden HFSCs through their aberrant differentiation and inducing hair follicle miniaturization and eventual hair loss. Conversely, transgenic or pharmacological activation of SHH rescued HFD-induced hair loss. These data collectively demonstrate that stem cell inflammatory signals induced by obesity robustly represses organ regeneration signals to accelerate the miniaturization of mini-organs, and suggests the importance of daily prevention of organ dysfunction.


Asunto(s)
Alopecia/patología , Alopecia/fisiopatología , Folículo Piloso/patología , Obesidad/fisiopatología , Células Madre/patología , Animales , Comunicación Autocrina , Recuento de Células , Diferenciación Celular , Linaje de la Célula , Senescencia Celular , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Proteínas Hedgehog/metabolismo , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/patología , Estrés Oxidativo , Comunicación Paracrina , Receptores de Interleucina-1/metabolismo
9.
Stem Cells ; 39(8): 1091-1100, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33783921

RESUMEN

Stem cell-based products have clinical and industrial applications. Thus, there is a need to develop quality control methods to standardize stem cell manufacturing. Here, we report a deep learning-based automated cell tracking (DeepACT) technology for noninvasive quality control and identification of cultured human stem cells. The combination of deep learning-based cascading cell detection and Kalman filter algorithm-based tracking successfully tracked the individual cells within the densely packed human epidermal keratinocyte colonies in the phase-contrast images of the culture. DeepACT rapidly analyzed the motion of individual keratinocytes, which enabled the quantitative evaluation of keratinocyte dynamics in response to changes in culture conditions. Furthermore, DeepACT can distinguish keratinocyte stem cell colonies from non-stem cell-derived colonies by analyzing the spatial and velocity information of cells. This system can be widely applied to stem cell cultures used in regenerative medicine and provides a platform for developing reliable and noninvasive quality control technology.


Asunto(s)
Aprendizaje Profundo , Células Epidérmicas , Diferenciación Celular , Rastreo Celular , Células Cultivadas , Humanos , Queratinocitos , Control de Calidad , Células Madre
10.
Nat Commun ; 12(1): 1826, 2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33758188

RESUMEN

Somatic mutations of ASXL1 are frequently detected in age-related clonal hematopoiesis (CH). However, how ASXL1 mutations drive CH remains elusive. Using knockin (KI) mice expressing a C-terminally truncated form of ASXL1-mutant (ASXL1-MT), we examined the influence of ASXL1-MT on physiological aging in hematopoietic stem cells (HSCs). HSCs expressing ASXL1-MT display competitive disadvantage after transplantation. Nevertheless, in genetic mosaic mouse model, they acquire clonal advantage during aging, recapitulating CH in humans. Mechanistically, ASXL1-MT cooperates with BAP1 to deubiquitinate and activate AKT. Overactive Akt/mTOR signaling induced by ASXL1-MT results in aberrant proliferation and dysfunction of HSCs associated with age-related accumulation of DNA damage. Treatment with an mTOR inhibitor rapamycin ameliorates aberrant expansion of the HSC compartment as well as dysregulated hematopoiesis in aged ASXL1-MT KI mice. Our findings suggest that ASXL1-MT provokes dysfunction of HSCs, whereas it confers clonal advantage on HSCs over time, leading to the development of CH.


Asunto(s)
Envejecimiento/genética , Hematopoyesis Clonal/genética , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Proteínas Represoras/genética , Serina-Treonina Quinasas TOR/metabolismo , Anciano , Envejecimiento/metabolismo , Envejecimiento/fisiología , Animales , Apoptosis/genética , Ciclo Celular/genética , Proliferación Celular/genética , Células Cultivadas , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Técnicas de Sustitución del Gen , Hematopoyesis/fisiología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/fisiología , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/genética , Ratones , Ratones Transgénicos , Mutación , Proteínas Proto-Oncogénicas c-akt/metabolismo , RNA-Seq , Especies Reactivas de Oxígeno/farmacología , Proteínas Represoras/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Sirolimus/farmacología , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinación/efectos de los fármacos , Ubiquitinación/genética
11.
Nat Aging ; 1(2): 190-204, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-37118636

RESUMEN

Hair follicles, mammalian mini-organs that grow hair, miniaturize during aging, leading to hair thinning and loss. Here we report that hair follicle stem cells (HFSCs) lose their regenerative capabilities during aging owing to the adoption of an atypical cell division program. Cell fate tracing and cell division axis analyses revealed that while HFSCs in young mice undergo typical symmetric and asymmetric cell divisions to regenerate hair follicles, upon aging or stress, they adopt an atypical 'stress-responsive' type of asymmetric cell division. This type of division is accompanied by the destabilization of hemidesmosomal protein COL17A1 and cell polarity protein aPKCλ and generates terminally differentiating epidermal cells instead of regenerating the hair follicle niche. With the repetition of these atypical divisions, HFSCs detach from the basal membrane causing their exhaustion, elimination and organ aging. The experimentally induced stabilization of COL17A1 rescued organ homeostasis through aPKCλ stabilization. These results demonstrate that distinct stem cell division programs may govern tissue and organ aging.


Asunto(s)
Folículo Piloso , Células Madre , Animales , Ratones , División Celular , Cabello , Mamíferos , Regeneración , Envejecimiento
12.
Regen Ther ; 14: 222-226, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32435674

RESUMEN

Impaired wound healing in critical limb ischemia (CLI) results from multiple factors that affect many cell types and their behavior. Epidermal keratinocytes and dermal fibroblasts play crucial roles in wound healing. However, it remains unclear whether these cell types irreversibly convert into a non-proliferative phenotype and are involved in impaired wound healing in CLI. Here, we demonstrate that skin keratinocytes and fibroblasts isolated from CLI patients maintain their proliferative potentials. Epidermal keratinocytes and dermal fibroblasts were isolated from the surrounding skin of foot wounds in CLI patients with diabetic nephropathy on hemodialysis, and their growth potentials were evaluated. It was found that keratinocytes from lower limbs and trunk of patients can give rise to proliferative growing colonies and can be serially passaged. Fibroblasts can also form colonies with a proliferative phenotype. These results indicate that skin keratinocytes and fibroblasts maintain their proliferative capacity even in diabetic and ischemic microenvironments and can be reactivated under appropriate conditions. This study provides strong evidence that the improvement of the cellular microenvironments is a promising therapeutic approach for CLI and these cells can also be used for potential sources of skin reconstruction.

13.
J Dermatol Sci ; 97(2): 143-151, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32001115

RESUMEN

BACKGROUND: NUAK2 is a critical gene that participates in the carcinogenesis of various types of cancers including melanomas. However, the expression patterns of NUAK2 in normal skin and in various types of skin tumors have not been fully elucidated to date. OBJECTIVES: To elucidate the distribution and localization of NUAK2 expression in normal skin, and characterize the expression patterns of NUAK2 and YAP in various types of skin tumors. METHODS: In this study, we characterized the expression of NUAK2 in tissues by developing a novel NUAK2-specific monoclonal antibody and using that to determine NUAK2 expression patterns in normal skin and in 155 cases of various types of skin tumors, including extramammary Paget's disease (EMPD), squamous cell carcinoma (SCC), Bowen's disease (BD), actinic keratosis (AK), basal cell carcinoma (BCC) and angiosarcoma (AS). Further, we analyzed the expression patterns of YAP and p-Akt in those tumors. RESULTS: Our analyses revealed that NUAK2 is expressed at high frequencies in EMPD, SCC, BD, AK, BCC and AS. The expression of p-Akt was positively correlated with tumor size in EMPD (P = 0.001). Importantly, the expression of NUAK2 was significantly correlated with YAP in SCC (P = 0.012) and in BD (P = 0.009). CONCLUSIONS: Our results suggest that the YAP-NUAK2 axis has critical importance in the tumorigenesis of SCC and BD, and that therapeutic modalities targeting the YAP-NUAK2 axis may be an effective approach against skin tumors including SCC and BD.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinogénesis/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Neoplasias Cutáneas/patología , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/análisis , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad de Bowen/patología , Carcinogénesis/genética , Carcinoma de Células Escamosas/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Proteínas Serina-Treonina Quinasas/análisis , Estudios Retrospectivos , Transducción de Señal/genética , Piel/metabolismo , Piel/patología , Factores de Transcripción/análisis , Proteínas Señalizadoras YAP
14.
Sci Rep ; 9(1): 18725, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31822757

RESUMEN

Identification and quality assurance of stem cells cultured in heterogeneous cell populations are indispensable for successful stem cell therapy. Here we present an image-processing pipeline for automated identification and quality assessment of human keratinocyte stem cells. When cultivated under appropriate conditions, human epidermal keratinocyte stem cells give rise to colonies and exhibit higher locomotive capacity as well as significant proliferative potential. Image processing and kernel density estimation were used to automatically extract the area of keratinocyte colonies from phase-contrast images of cultures containing feeder cells. The DeepFlow algorithm was then used to calculate locomotion speed of the colony area by analyzing serial images. This image-processing pipeline successfully identified keratinocyte stem cell colonies by measuring cell locomotion speed, and also assessed the effect of oligotrophic culture conditions and chemical inhibitors on keratinocyte behavior. Therefore, this study provides automated procedures for image-based quality control of stem cell cultures and high-throughput screening of small molecules targeting stem cells.


Asunto(s)
Movimiento Celular/fisiología , Procesamiento de Imagen Asistido por Computador/métodos , Queratinocitos/citología , Algoritmos , Automatización de Laboratorios/métodos , Técnicas de Cultivo de Célula , Diferenciación Celular , Proliferación Celular , Células Epidérmicas , Células Nutrientes , Humanos , Queratinocitos/fisiología , Microscopía de Contraste de Fase/métodos , Movimiento (Física) , Células Madre/citología
15.
Nat Commun ; 10(1): 5023, 2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31685822

RESUMEN

Melanoma, the deadliest skin cancer, remains largely incurable at advanced stages. Currently, there is a lack of animal models that resemble human melanoma initiation and progression. Recent studies using a Tyr-CreER driven mouse model have drawn contradictory conclusions about the potential of melanocyte stem cells (McSCs) to form melanoma. Here, we employ a c-Kit-CreER-driven model that specifically targets McSCs to show that oncogenic McSCs are a bona fide source of melanoma that expand in the niche, and then establish epidermal melanomas that invade into the underlying dermis. Further, normal Wnt and Endothelin niche signals during hair anagen onset are hijacked to promote McSC malignant transformation during melanoma induction. Finally, molecular profiling reveals strong resemblance of murine McSC-derived melanoma to human melanoma in heterogeneity and gene signatures. These findings provide experimental validation of the human melanoma progression model and key insights into the transformation and heterogeneity of McSC-derived melanoma.


Asunto(s)
Carcinogénesis/patología , Melanocitos/patología , Melanoma/patología , Células Madre Neoplásicas/patología , Animales , Carcinogénesis/metabolismo , Transformación Celular Neoplásica/patología , Dermis/patología , Modelos Animales de Enfermedad , Epidermis/patología , Homeostasis , Humanos , Melanocitos/metabolismo , Ratones , Mutación/genética , Células Madre Neoplásicas/metabolismo , Fenotipo , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Microambiente Tumoral , Vía de Señalización Wnt
16.
J Dermatol Sci ; 94(2): 298-305, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-31122679

RESUMEN

BACKGROUND: Epidermal stem cells (ESCs) are keratinocytes that reside in the basal layer of the epidermis and mediate epidermal homeostasis. Insulin-like growth factor 1 (IGF-1) signaling through its receptor (IGF-1R) has been identified as an important regulator in rodent skin development and differentiation. However, the role of IGF-1/IGF-1R signaling in human keratinocytes is not yet well understood. OBJECTIVE: This study aimed to clarify the role of IGF-1/IGF-1R signaling in human epidermal homeostasis. METHODS: IGF-1R specific knockout (KO) HaCaT keratinocytes were generated by CRISPR-Caspase-9-mediated non-homologous end joining frame-shift mutations. Further, the behavior of these keratinocytes in epidermal homeostasis was investigated using reconstructed epidermis and human skin equivalents. RESULTS: IGF-1R KO HaCaT keratinocytes were successfully established and produced thin epidermis in three-dimensional culture models. Keratin10-positive cells were frequently found in the basal layer of the reconstructed epidermis. CONCLUSIONS: IGF-1/IGF-1R signaling was demonstrated to play a key role in maintaining human epidermal homeostasis. This method provides a new framework to investigate gene function in human epidermal homeostasis.


Asunto(s)
Epidermis/fisiología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Queratinocitos/metabolismo , Receptor IGF Tipo 1/metabolismo , Células Madre/fisiología , Diferenciación Celular , Línea Celular , Técnicas de Inactivación de Genes , Humanos , Receptor IGF Tipo 1/genética , Transducción de Señal
17.
Nature ; 568(7752): 344-350, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30944469

RESUMEN

Stem cells underlie tissue homeostasis, but their dynamics during ageing-and the relevance of these dynamics to organ ageing-remain unknown. Here we report that the expression of the hemidesmosome component collagen XVII (COL17A1) by epidermal stem cells fluctuates physiologically through genomic/oxidative stress-induced proteolysis, and that the resulting differential expression of COL17A1 in individual stem cells generates a driving force for cell competition. In vivo clonal analysis in mice and in vitro 3D modelling show that clones that express high levels of COL17A1, which divide symmetrically, outcompete and eliminate adjacent stressed clones that express low levels of COL17A1, which divide asymmetrically. Stem cells with higher potential or quality are thus selected for homeostasis, but their eventual loss of COL17A1 limits their competition, thereby causing ageing. The resultant hemidesmosome fragility and stem cell delamination deplete adjacent melanocytes and fibroblasts to promote skin ageing. Conversely, the forced maintenance of COL17A1 rescues skin organ ageing, thereby indicating potential angles for anti-ageing therapeutic intervention.


Asunto(s)
Homeostasis , Envejecimiento de la Piel/patología , Envejecimiento de la Piel/fisiología , Piel/citología , Piel/patología , Células Madre/citología , Células Madre/patología , Animales , Atrofia , Autoantígenos/química , Autoantígenos/metabolismo , División Celular , Proliferación Celular , Células Clonales/citología , Células Epidérmicas/citología , Células Epidérmicas/patología , Femenino , Genoma , Hemidesmosomas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Colágenos no Fibrilares/química , Colágenos no Fibrilares/metabolismo , Estrés Oxidativo , Proteolisis , Colágeno Tipo XVII
18.
Cell Stem Cell ; 19(2): 192-204, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27345838

RESUMEN

Hematopoietic stem cells (HSCs) maintain quiescence by activating specific metabolic pathways, including glycolysis. We do not yet have a clear understanding of how this metabolic activity changes during stress hematopoiesis, such as bone marrow transplantation. Here, we report a critical role for the p38MAPK family isoform p38α in initiating hematopoietic stem and progenitor cell (HSPC) proliferation during stress hematopoiesis in mice. We found that p38MAPK is immediately phosphorylated in HSPCs after a hematological stress, preceding increased HSPC cycling. Conditional deletion of p38α led to defective recovery from hematological stress and a delay in initiation of HSPC proliferation. Mechanistically, p38α signaling increases expression of inosine-5'-monophosphate dehydrogenase 2 in HSPCs, leading to altered levels of amino acids and purine-related metabolites and changes in cell-cycle progression in vitro and in vivo. Our studies have therefore uncovered a p38α-mediated pathway that alters HSPC metabolism to respond to stress and promote recovery.


Asunto(s)
Ciclo Celular , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Purinas/metabolismo , Estrés Fisiológico , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Apoptosis , Proliferación Celular , Activación Enzimática , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/enzimología , IMP Deshidrogenasa/antagonistas & inhibidores , IMP Deshidrogenasa/metabolismo , Ratones Endogámicos C57BL , Factor de Transcripción Asociado a Microftalmía/metabolismo , Mutación/genética , Fenotipo , Especies Reactivas de Oxígeno/metabolismo
19.
Science ; 351(6273): aad4395, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26912707

RESUMEN

Hair thinning and loss are prominent aging phenotypes but have an unknown mechanism. We show that hair follicle stem cell (HFSC) aging causes the stepwise miniaturization of hair follicles and eventual hair loss in wild-type mice and in humans. In vivo fate analysis of HFSCs revealed that the DNA damage response in HFSCs causes proteolysis of type XVII collagen (COL17A1/BP180), a critical molecule for HFSC maintenance, to trigger HFSC aging, characterized by the loss of stemness signatures and by epidermal commitment. Aged HFSCs are cyclically eliminated from the skin through terminal epidermal differentiation, thereby causing hair follicle miniaturization. The aging process can be recapitulated by Col17a1 deficiency and prevented by the forced maintenance of COL17A1 in HFSCs, demonstrating that COL17A1 in HFSCs orchestrates the stem cell-centric aging program of the epithelial mini-organ.


Asunto(s)
Alopecia/metabolismo , Senescencia Celular/fisiología , Folículo Piloso/patología , Colágenos no Fibrilares/deficiencia , Proteolisis , Células Madre/patología , Anciano , Envejecimiento/metabolismo , Envejecimiento/patología , Alopecia/genética , Alopecia/patología , Animales , Autoantígenos/genética , Diferenciación Celular , Senescencia Celular/genética , Daño del ADN , Desmosomas/metabolismo , Desmosomas/patología , Femenino , Inestabilidad Genómica , Folículo Piloso/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Colágenos no Fibrilares/genética , Células Madre/metabolismo , Colágeno Tipo XVII
20.
J Dermatol Sci ; 82(2): 84-94, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26867959

RESUMEN

BACKGROUND: Skin fibroblast heterogeneity is of growing interest due to its relevance in not only skin development but also cutaneous wound healing. However, the characterization of human dermal fibroblasts at a clonal level has not been accomplished and their functional heterogeneity remains poorly understood. OBJECTIVE: The aim of this study was to define the clonal heterogeneity of human dermal fibroblasts. METHODS: Isolated human dermal fibroblasts were clonally expanded and categorized by comprehensive phenotypic and gene expression profiling. RESULTS: Single fibroblasts were significantly multiplied and efficiently cloned without chromosomal abnormalities under hypoxic conditions. Individual clones were heterogeneous in their proliferative capacity, and gene expression profiling revealed differences in the expression of genes involved in extracellular matrix synthesis and degradation. Each cloned fibroblast also had different abilities in terms of collagen remodeling. All phenotypic and gene expression data were analyzed with Spearman's rank correlation, and fibroblasts were categorized into at least two functional clonal types. One was highly proliferative, while the other was less proliferative but had the ability to remodel the tissue architecture. The proliferative clones were predominant in infants, but decreased with physiological aging. CONCLUSION: This study provides strong evidence for the functional heterogeneity of human dermal fibroblasts at a clonal level, which has implications regarding skin repair and aging.


Asunto(s)
Proliferación Celular/fisiología , Células Clonales/fisiología , Fibroblastos/fisiología , Piel/citología , Anciano , Anciano de 80 o más Años , Células Cultivadas , Niño , Preescolar , Colágeno/metabolismo , Colágeno/fisiología , Femenino , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Humanos , Lactante , Masculino , Persona de Mediana Edad , Envejecimiento de la Piel/fisiología , Cicatrización de Heridas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...