Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Cell ; 175(6): 1461-1463, 2018 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-30500532

RESUMEN

Mechanisms underlying aging of the skin dermis are poorly understood. Now, two studies (Marsh et al., 2018; Salzer et al., 2018) describe complementary approaches to this question: Salzer et al. show that aging dermal fibroblasts lose defined identity in a diet-influenced fashion, and Marsh et al. reveal that fibroblast loss over time is compensated by membrane expansion rather than proliferation, resulting in decreased cellular density.


Asunto(s)
Dermis , Envejecimiento de la Piel , Dieta , Fibroblastos , Homeostasis , Piel
2.
Nature ; 616(7958): 774-782, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37076619

RESUMEN

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.


Asunto(s)
Desdiferenciación Celular , Folículo Piloso , Melanocitos , Nicho de Células Madre , Células Madre , Animales , Humanos , Ratones , Folículo Piloso/citología , Melanocitos/citología , Células Madre/citología , Microambiente Celular , Linaje de la Célula , Envejecimiento , Homeostasis , Color del Cabello/fisiología
3.
Genes Dev ; 34(13-14): 973-988, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32467224

RESUMEN

Chromatin modifiers play critical roles in epidermal development, but the functions of histone deacetylases in this context are poorly understood. The class I HDAC, HDAC3, is of particular interest because it plays divergent roles in different tissues by partnering with tissue-specific transcription factors. We found that HDAC3 is expressed broadly in embryonic epidermis and is required for its orderly stepwise stratification. HDAC3 protein stability in vivo relies on NCoR and SMRT, which function redundantly in epidermal development. However, point mutations in the NCoR and SMRT deacetylase-activating domains, which are required for HDAC3's enzymatic function, permit normal stratification, indicating that HDAC3's roles in this context are largely independent of its histone deacetylase activity. HDAC3-bound sites are significantly enriched for predicted binding motifs for critical epidermal transcription factors including AP1, GRHL, and KLF family members. Our results suggest that among these, HDAC3 operates in conjunction with KLF4 to repress inappropriate expression of Tgm1, Krt16, and Aqp3 In parallel, HDAC3 suppresses expression of inflammatory cytokines through a Rela-dependent mechanism. These data identify HDAC3 as a hub coordinating multiple aspects of epidermal barrier acquisition.


Asunto(s)
Diferenciación Celular/genética , Células Epidérmicas/citología , Epidermis/embriología , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Animales , Embrión de Mamíferos , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Genes Letales/genética , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Ratones Endogámicos C57BL , Mutación , Co-Represor 1 de Receptor Nuclear/genética , Co-Represor 1 de Receptor Nuclear/metabolismo , Co-Represor 2 de Receptor Nuclear/genética , Co-Represor 2 de Receptor Nuclear/metabolismo , Dominios y Motivos de Interacción de Proteínas/genética , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo
4.
PLoS Biol ; 21(2): e3001989, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36745682

RESUMEN

Angiotensin-converting enzyme 2 (ACE2) is the cell-surface receptor for Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). While its central role in Coronavirus Disease 2019 (COVID-19) pathogenesis is indisputable, there remains significant debate regarding the role of this transmembrane carboxypeptidase in the disease course. These include the role of soluble versus membrane-bound ACE2, as well as ACE2-independent mechanisms that may contribute to viral spread. Testing these roles requires in vivo models. Here, we report humanized ACE2-floxed mice in which hACE2 is expressed from the mouse Ace2 locus in a manner that confers lethal disease and permits cell-specific, Cre-mediated loss of function, and LSL-hACE2 mice in which hACE2 is expressed from the Rosa26 locus enabling cell-specific, Cre-mediated gain of function. Following exposure to SARS-CoV-2, hACE2-floxed mice experienced lethal cachexia, pulmonary infiltrates, intravascular thrombosis and hypoxemia-hallmarks of severe COVID-19. Cre-mediated loss and gain of hACE2 demonstrate that neuronal infection confers lethal cachexia, hypoxemia, and respiratory failure in the absence of lung epithelial infection. In this series of genetic experiments, we demonstrate that ACE2 is absolutely and cell-autonomously required for SARS-CoV-2 infection in the olfactory epithelium, brain, and lung across diverse cell types. Therapies inhibiting or blocking ACE2 at these different sites are likely to be an effective strategy towards preventing severe COVID-19.


Asunto(s)
COVID-19 , Ratones , Animales , Enzima Convertidora de Angiotensina 2/genética , SARS-CoV-2/metabolismo , Caquexia , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Hipoxia
5.
Nature ; 550(7676): 402-406, 2017 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-28976970

RESUMEN

Chromatin is traditionally viewed as a nuclear entity that regulates gene expression and silencing. However, we recently discovered the presence of cytoplasmic chromatin fragments that pinch off from intact nuclei of primary cells during senescence, a form of terminal cell-cycle arrest associated with pro-inflammatory responses. The functional significance of chromatin in the cytoplasm is unclear. Here we show that cytoplasmic chromatin activates the innate immunity cytosolic DNA-sensing cGAS-STING (cyclic GMP-AMP synthase linked to stimulator of interferon genes) pathway, leading both to short-term inflammation to restrain activated oncogenes and to chronic inflammation that associates with tissue destruction and cancer. The cytoplasmic chromatin-cGAS-STING pathway promotes the senescence-associated secretory phenotype in primary human cells and in mice. Mice deficient in STING show impaired immuno-surveillance of oncogenic RAS and reduced tissue inflammation upon ionizing radiation. Furthermore, this pathway is activated in cancer cells, and correlates with pro-inflammatory gene expression in human cancers. Overall, our findings indicate that genomic DNA serves as a reservoir to initiate a pro-inflammatory pathway in the cytoplasm in senescence and cancer. Targeting the cytoplasmic chromatin-mediated pathway may hold promise in treating inflammation-related disorders.


Asunto(s)
Senescencia Celular/genética , Cromatina/metabolismo , Citoplasma/genética , Inmunidad Innata , Inflamación/genética , Inflamación/patología , Neoplasias/genética , Neoplasias/inmunología , Animales , Línea Celular Tumoral , Cromatina/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Citoplasma/inmunología , Femenino , Humanos , Inflamación/inmunología , Hígado/metabolismo , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Neoplasias/patología , Nucleotidiltransferasas/metabolismo , Proteína Oncogénica p21(ras)/genética , Proteína Oncogénica p21(ras)/inmunología , Radiación Ionizante
6.
BMC Biol ; 18(1): 87, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32664967

RESUMEN

BACKGROUND: The formation of supernumerary teeth is an excellent model for studying the molecular mechanisms that control stem/progenitor cell homeostasis needed to generate a renewable source of replacement cells and tissues. Although multiple growth factors and transcriptional factors have been associated with supernumerary tooth formation, the regulatory inputs of extracellular matrix in this regenerative process remains poorly understood. RESULTS: In this study, we present evidence that disrupting glycosaminoglycans (GAGs) in the dental epithelium of mice by inactivating FAM20B, a xylose kinase essential for GAG assembly, leads to supernumerary tooth formation in a pattern reminiscent of replacement teeth. The dental epithelial GAGs confine murine tooth number by restricting the homeostasis of Sox2(+) dental epithelial stem/progenitor cells in a non-autonomous manner. FAM20B-catalyzed GAGs regulate the cell fate of dental lamina by restricting FGFR2b signaling at the initial stage of tooth development to maintain a subtle balance between the renewal and differentiation of Sox2(+) cells. At the later cap stage, WNT signaling functions as a relay cue to facilitate the supernumerary tooth formation. CONCLUSIONS: The novel mechanism we have characterized through which GAGs control the tooth number in mice may also be more broadly relevant for potentiating signaling interactions in other tissues during development and tissue homeostasis.


Asunto(s)
Glicosaminoglicanos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Transducción de Señal , Diente Supernumerario/genética , Animales , Diferenciación Celular , Ratones , Odontogénesis , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Células Madre/metabolismo
7.
Chem Senses ; 45(7): 493-502, 2020 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-32556127

RESUMEN

The chemical senses of taste and smell play a vital role in conveying information about ourselves and our environment. Tastes and smells can warn against danger and also contribute to the daily enjoyment of food, friends and family, and our surroundings. Over 12% of the US population is estimated to experience taste and smell (chemosensory) dysfunction. Yet, despite this high prevalence, long-term, effective treatments for these disorders have been largely elusive. Clinical successes in other sensory systems, including hearing and vision, have led to new hope for developments in the treatment of chemosensory disorders. To accelerate cures, we convened the "Identifying Treatments for Taste and Smell Disorders" conference, bringing together basic and translational sensory scientists, health care professionals, and patients to identify gaps in our current understanding of chemosensory dysfunction and next steps in a broad-based research strategy. Their suggestions for high-yield next steps were focused in 3 areas: increasing awareness and research capacity (e.g., patient advocacy), developing and enhancing clinical measures of taste and smell, and supporting new avenues of research into cellular and therapeutic approaches (e.g., developing human chemosensory cell lines, stem cells, and gene therapy approaches). These long-term strategies led to specific suggestions for immediate research priorities that focus on expanding our understanding of specific responses of chemosensory cells and developing valuable assays to identify and document cell development, regeneration, and function. Addressing these high-priority areas should accelerate the development of novel and effective treatments for taste and smell disorders.


Asunto(s)
Trastornos del Olfato/terapia , Trastornos del Gusto/terapia , Congresos como Asunto , Terapia Genética , Humanos , Trastornos del Olfato/patología , Medicina Regenerativa , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Trasplante de Células Madre , Células Madre/citología , Células Madre/metabolismo , Trastornos del Gusto/patología
8.
PLoS Genet ; 13(8): e1006990, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28846687

RESUMEN

Taste stimuli are transduced by taste buds and transmitted to the brain via afferent gustatory fibers. Renewal of taste receptor cells from actively dividing progenitors is finely tuned to maintain taste sensitivity throughout life. We show that conditional ß-catenin deletion in mouse taste progenitors leads to rapid depletion of progenitors and Shh+ precursors, which in turn causes taste bud loss, followed by loss of gustatory nerve fibers. In addition, our data suggest LEF1, TCF7 and Wnt3 are involved in a Wnt pathway regulatory feedback loop that controls taste cell renewal in the circumvallate papilla epithelium. Unexpectedly, taste bud decline is greater in the anterior tongue and palate than in the posterior tongue. Mutant mice with this regional pattern of taste bud loss were unable to discern sweet at any concentration, but could distinguish bitter stimuli, albeit with reduced sensitivity. Our findings are consistent with published reports wherein anterior taste buds have higher sweet sensitivity while posterior taste buds are better tuned to bitter, and suggest ß-catenin plays a greater role in renewal of anterior versus posterior taste buds.


Asunto(s)
Papilas Gustativas/crecimiento & desarrollo , Percepción del Gusto/genética , beta Catenina/genética , Animales , Autorrenovación de las Células/genética , Factor Nuclear 1-alfa del Hepatocito/genética , Factor de Unión 1 al Potenciador Linfoide/genética , Ratones , Hueso Paladar/metabolismo , Hueso Paladar/fisiología , Papilas Gustativas/metabolismo , Lengua/metabolismo , Lengua/fisiología , Vía de Señalización Wnt , Proteína Wnt3/genética
9.
Development ; 143(9): 1512-22, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26952977

RESUMEN

In the epidermis of mice lacking transcription factor nuclear factor-kappa B (NF-κB) activity, primary hair follicle (HF) pre-placode formation is initiated without progression to proper placodes. NF-κB modulates WNT and SHH signaling at early stages of HF development, but this does not fully account for the phenotypes observed upon NF-κB inhibition. To identify additional NF-κB target genes, we developed a novel method to isolate and transcriptionally profile primary HF placodes with active NF-κB signaling. In parallel, we compared gene expression at the same developmental stage in NF-κB-deficient embryos and controls. This uncovered novel NF-κB target genes with potential roles in priming HF placodes for down-growth. Importantly, we identify Lhx2 (encoding a LIM/homeobox transcription factor) as a direct NF-κB target gene, loss of which replicates a subset of phenotypes seen in NF-κB-deficient embryos. Lhx2 and Tgfb2 knockout embryos exhibit very similar abnormalities in HF development, including failure of the E-cadherin suppression required for follicle down-growth. We show that TGFß2 signaling is impaired in NF-κB-deficient and Lhx2 knockout embryos and that exogenous TGFß2 rescues the HF phenotypes in Lhx2 knockout skin explants, indicating that it operates downstream of LHX2. These findings identify a novel NF-κB/LHX2/TGFß2 signaling axis that is crucial for primary HF morphogenesis, which may also function more broadly in development and disease.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Folículo Piloso/embriología , Proteínas con Homeodominio LIM/genética , Organogénesis/genética , Factor de Transcripción ReIA/genética , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta2/genética , Animales , Cadherinas/metabolismo , Diferenciación Celular/genética , Movimiento Celular/genética , Embrión de Mamíferos/metabolismo , Ratones , Ratones Noqueados , Técnicas de Cultivo de Órganos , Receptores de Factores de Crecimiento Transformadores beta/metabolismo
10.
PLoS Genet ; 11(5): e1005208, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-26020789

RESUMEN

Continuous taste bud cell renewal is essential to maintain taste function in adults; however, the molecular mechanisms that regulate taste cell turnover are unknown. Using inducible Cre-lox technology, we show that activation of ß-catenin signaling in multipotent lingual epithelial progenitors outside of taste buds diverts daughter cells from a general epithelial to a taste bud fate. Moreover, while taste buds comprise 3 morphological types, ß-catenin activation drives overproduction of primarily glial-like Type I taste cells in both anterior fungiform (FF) and posterior circumvallate (CV) taste buds, with a small increase in Type II receptor cells for sweet, bitter and umami, but does not alter Type III sour detector cells. Beta-catenin activation in post-mitotic taste bud precursors likewise regulates cell differentiation; forced activation of ß-catenin in these Shh+ cells promotes Type I cell fate in both FF and CV taste buds, but likely does so non-cell autonomously. Our data are consistent with a model where ß-catenin signaling levels within lingual epithelial progenitors dictate cell fate prior to or during entry of new cells into taste buds; high signaling induces Type I cells, intermediate levels drive Type II cell differentiation, while low levels may drive differentiation of Type III cells.


Asunto(s)
Diferenciación Celular/genética , Papilas Gustativas/crecimiento & desarrollo , Gusto/genética , beta Catenina/genética , Animales , Linaje de la Célula/genética , Células Epiteliales/metabolismo , Ratones , Ratones Transgénicos , Transducción de Señal , Papilas Gustativas/metabolismo , beta Catenina/metabolismo
11.
Nat Genet ; 40(9): 1130-5, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19165927

RESUMEN

Constitutive Hedgehog (Hh) signaling underlies several human tumors, including basal cell carcinoma (BCC) and basaloid follicular hamartoma in skin. Intriguingly, superficial BCCs arise as de novo epithelial buds resembling embryonic hair germs, collections of epidermal cells whose development is regulated by canonical Wnt/beta-catenin signaling. Similar to embryonic hair germs, human BCC buds showed increased levels of cytoplasmic and nuclear beta-catenin and expressed early hair follicle lineage markers. We also detected canonical Wnt/ beta-catenin signaling in epithelial buds and hamartomas from mice expressing an oncogene, M2SMO, leading to constitutive Hh signaling in skin. Conditional overexpression of the Wnt pathway antagonist Dkk1 in M2SMO-expressing mice potently inhibited epithelial bud and hamartoma development without affecting Hh signaling. Our findings uncover a hitherto unknown requirement for ligand-driven, canonical Wnt/ beta-catenin signaling for Hh pathway-driven tumorigenesis, identify a new pharmacological target for these neoplasms and establish the molecular basis for the well-known similarity between early superficial BCCs and embryonic hair germs.


Asunto(s)
Carcinoma Basocelular/genética , Proteínas Hedgehog/genética , Neoplasias Cutáneas/genética , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Linaje de la Célula , Células Epiteliales/metabolismo , Folículo Piloso/embriología , Hamartoma/genética , Humanos , Ratones , Proteínas Oncogénicas/genética , Transducción de Señal
12.
Development ; 140(8): 1655-64, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23487314

RESUMEN

The mammalian hair follicle relies on adult resident stem cells and their progeny to fuel and maintain hair growth throughout the life of an organism. The cyclical and initially synchronous nature of hair growth makes the hair follicle an ideal system with which to define homeostatic mechanisms of an adult stem cell population. Recently, we demonstrated that Hopx is a specific marker of intestinal stem cells. Here, we show that Hopx specifically labels long-lived hair follicle stem cells residing in the telogen basal bulge. Hopx(+) cells contribute to all lineages of the mature hair follicle and to the interfollicular epidermis upon epidermal wounding. Unexpectedly, our analysis identifies a previously unappreciated progenitor population that resides in the lower hair bulb of anagen-phase follicles and expresses Hopx. These cells co-express Lgr5, do not express Shh and escape catagen-induced apoptosis. They ultimately differentiate into the cytokeratin 6-positive (K6) inner bulge cells in telogen, which regulate the quiescence of adjacent hair follicle stem cells. Although previous studies have suggested that K6(+) cells arise from Lgr5-expressing lower outer root sheath cells in anagen, our studies indicate an alternative origin, and a novel role for Hopx-expressing lower hair bulb progenitor cells in contributing to stem cell homeostasis.


Asunto(s)
Diferenciación Celular/fisiología , Células Epidérmicas , Folículo Piloso/citología , Cabello/crecimiento & desarrollo , Proteínas de Homeodominio/metabolismo , Queratina-6/metabolismo , Células Madre Multipotentes/metabolismo , Animales , Bromodesoxiuridina , Linaje de la Célula/fisiología , Citometría de Flujo , Etiquetado Corte-Fin in Situ , Queratinocitos/metabolismo , Queratinocitos/fisiología , Ratones , Ratones Transgénicos , Tamoxifeno , beta-Galactosidasa
13.
Eur J Oral Sci ; 124(3): 221-7, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26957367

RESUMEN

Beta-catenin is a multifunctional protein that plays key roles in cadherin-based cell adherens junctions and in the Wnt signaling pathway. The canonical Wnt/ß-catenin pathway can regulate transcription factors that control cell movement/invasion. We investigated whether ß-catenin regulates ameloblast movement through canonical Wnt signaling. The morphological and physical properties of enamel were assessed in enamel from control and ß-catenin conditional knockout (cKO) mice. Ameloblast-lineage cells (ALC) were used to investigate the potential roles of ß-catenin in cell migration and in E-cadherin expression. Compared with controls, incisors from ß-catenin cKO mice were short, blunt, and where enamel was present, it was soft and malformed. Scanning electron microscopy revealed a dysplastic rod pattern within the enamel of incisors from ß-catenin cKO mice, and Vickers microhardness measurements confirmed that mice with ß-catenin ablated from their enamel organ had enamel that was significantly softer than normal. Amelogenesis was disrupted in the absence of ß-catenin and the ameloblasts did not differentiate properly. We further demonstrated that migration of ALCs was inhibited in vitro and that E-cadherin expression was significantly up-regulated when ALCs were treated with the ß-catenin inhibitor, ICG-001. Beta-catenin ablation causes enamel malformation in mice and this phenotype may occur, in part, by a lack of ameloblast differentiation and/or movement necessary to form the decussating enamel rod structure.


Asunto(s)
Ameloblastos , Amelogénesis , Esmalte Dental/crecimiento & desarrollo , beta Catenina/fisiología , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes , Ratones , Pirimidinonas
14.
Nat Genet ; 39(1): 106-12, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17128274

RESUMEN

Fungiform taste papillae form a regular array on the dorsal tongue. Taste buds arise from papilla epithelium and, unusually for epithelial derivatives, synapse with neurons, release neurotransmitters and generate receptor and action potentials. Despite the importance of taste as one of our five senses, genetic analyses of taste papilla and bud development are lacking. We demonstrate that Wnt-beta-catenin signaling is activated in developing fungiform placodes and taste bud cells. A dominant stabilizing mutation of epithelial beta-catenin causes massive overproduction of enlarged fungiform papillae and taste buds. Likewise, genetic deletion of epithelial beta-catenin or inhibition of Wnt-beta-catenin signaling by ectopic dickkopf1 (Dkk1) blocks initiation of fungiform papilla morphogenesis. Ectopic papillae are innervated in the stabilizing beta-catenin mutant, whereas ectopic Dkk1 causes absence of lingual epithelial innervation. Thus, Wnt-beta-catenin signaling is critical for fungiform papilla and taste bud development. Altered regulation of this pathway may underlie evolutionary changes in taste papilla patterning.


Asunto(s)
Papilas Gustativas/embriología , Proteínas Wnt/fisiología , beta Catenina/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Femenino , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Transgénicos , Morfogénesis/genética , Embarazo , Transducción de Señal/genética , Papilas Gustativas/crecimiento & desarrollo , beta Catenina/genética
15.
Development ; 139(8): 1405-16, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22434867

RESUMEN

MicroRNAs (miRNAs) regulate the expression of many mammalian genes and play key roles in embryonic hair follicle development; however, little is known of their functions in postnatal hair growth. We compared the effects of deleting the essential miRNA biogenesis enzymes Drosha and Dicer in mouse skin epithelial cells at successive postnatal time points. Deletion of either Drosha or Dicer during an established growth phase (anagen) caused failure of hair follicles to enter a normal catagen regression phase, eventual follicular degradation and stem cell loss. Deletion of Drosha or Dicer in resting phase follicles did not affect follicular structure or epithelial stem cell maintenance, and stimulation of anagen by hair plucking caused follicular proliferation and formation of a primitive transient amplifying matrix population. However, mutant matrix cells exhibited apoptosis and DNA damage and hair follicles rapidly degraded. Hair follicle defects at early time points post-deletion occurred in the absence of inflammation, but a dermal inflammatory response and hyperproliferation of interfollicular epidermis accompanied subsequent hair follicle degradation. These data reveal multiple functions for Drosha and Dicer in suppressing DNA damage in rapidly proliferating follicular matrix cells, facilitating catagen and maintaining follicular structures and their associated stem cells. Although Drosha and Dicer each possess independent non-miRNA-related functions, the similarity in phenotypes of the inducible epidermal Drosha and Dicer mutants indicates that these defects result primarily from failure of miRNA processing. Consistent with this, Dicer deletion resulted in the upregulation of multiple direct targets of the highly expressed epithelial miRNA miR-205.


Asunto(s)
ARN Helicasas DEAD-box/genética , Eliminación de Gen , MicroARNs/metabolismo , Ribonucleasa III/genética , Piel/crecimiento & desarrollo , Animales , Cruzamientos Genéticos , ARN Helicasas DEAD-box/fisiología , Células Epidérmicas , Folículo Piloso/metabolismo , Ratones , Microscopía Fluorescente/métodos , Fenotipo , Ribonucleasa III/fisiología , Transducción de Señal , Piel/metabolismo , Células Madre/citología , Cicatrización de Heridas
16.
Nat Mater ; 18(6): 530-531, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31114070
17.
Proc Natl Acad Sci U S A ; 109(29): 11758-63, 2012 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-22753467

RESUMEN

The role of the circadian clock in skin and the identity of genes participating in its chronobiology remain largely unknown, leading us to define the circadian transcriptome of mouse skin at two different stages of the hair cycle, telogen and anagen. The circadian transcriptomes of telogen and anagen skin are largely distinct, with the former dominated by genes involved in cell proliferation and metabolism. The expression of many metabolic genes is antiphasic to cell cycle-related genes, the former peaking during the day and the latter at night. Consistently, accumulation of reactive oxygen species, a byproduct of oxidative phosphorylation, and S-phase are antiphasic to each other in telogen skin. Furthermore, the circadian variation in S-phase is controlled by BMAL1 intrinsic to keratinocytes, because keratinocyte-specific deletion of Bmal1 obliterates time-of-day-dependent synchronicity of cell division in the epidermis leading to a constitutively elevated cell proliferation. In agreement with higher cellular susceptibility to UV-induced DNA damage during S-phase, we found that mice are most sensitive to UVB-induced DNA damage in the epidermis at night. Because in the human epidermis maximum numbers of keratinocytes go through S-phase in the late afternoon, we speculate that in humans the circadian clock imposes regulation of epidermal cell proliferation so that skin is at a particularly vulnerable stage during times of maximum UV exposure, thus contributing to the high incidence of human skin cancers.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Proliferación Celular , Ritmo Circadiano/genética , Daño del ADN/genética , Células Epidérmicas , Redes y Vías Metabólicas/genética , Transcriptoma/genética , Factores de Transcripción ARNTL/genética , Animales , Bromodesoxiuridina , Ciclo Celular/fisiología , Ritmo Circadiano/fisiología , Colchicina , Daño del ADN/fisiología , Ensayo de Inmunoadsorción Enzimática , Epidermis/efectos de la radiación , Inmunohistoquímica , Masculino , Redes y Vías Metabólicas/fisiología , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , Reacción en Cadena de la Polimerasa , Transcriptoma/fisiología , Rayos Ultravioleta/efectos adversos
18.
Ocul Surf ; 33: 39-49, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38679196

RESUMEN

PURPOSE: To investigate the roles of HDAC1/2 and HDAC3 in adult Meibomian gland (MG) homeostasis. METHODS: HDAC1/2 or HDAC3 were inducibly deleted in MG epithelial cells of adult mice. The morphology of MG was examined. Proliferation, apoptosis, and expression of MG acinus and duct marker genes, meibocyte differentiation genes, and HDAC target genes, were analyzed via immunofluorescence, TUNEL assay, and RNA in situ hybridization. RESULTS: Co-deletion of HDAC1/2 in MG epithelium caused gradual loss of acini and formation of cyst-like structures in the central duct. These phenotypes required homozygous deletion of both HDAC1 and HDAC2, indicating that they function redundantly in the adult MG. Short-term deletion of HDAC1/2 in MG epithelium had little effect on meibocyte maturation but caused decreased proliferation of acinar basal cells, excessive DNA damage, ectopic apoptosis, and increased p53 acetylation and p16 expression in the MG. By contrast, HDAC3 deletion in MG epithelium caused dilation of central duct, atrophy of acini, defective meibocyte maturation, increased acinar basal cell proliferation, and ectopic apoptosis and DNA damage. Levels of p53 acetylation and p21 expression were elevated in HDAC3-deficient MGs, while the expression of the differentiation regulator PPARγ and the differentiation markers PLIN2 and FASN was downregulated. CONCLUSIONS: HDAC1 and HDAC2 function redundantly in adult Meibomian gland epithelial progenitor cells and are essential for their proliferation and survival, but not for acinar differentiation, while HDAC3 is required to limit acinar progenitor cell proliferation and permit differentiation. HDAC1/2 and HDAC3 have partially overlapping roles in maintaining survival of MG cells.


Asunto(s)
Apoptosis , Histona Desacetilasa 1 , Histona Desacetilasa 2 , Histona Desacetilasas , Homeostasis , Glándulas Tarsales , Animales , Glándulas Tarsales/metabolismo , Glándulas Tarsales/patología , Ratones , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 1/genética , Homeostasis/fisiología , Histona Desacetilasas/metabolismo , Histona Desacetilasas/genética , Histona Desacetilasa 2/metabolismo , Histona Desacetilasa 2/genética , Proliferación Celular/fisiología , Etiquetado Corte-Fin in Situ , Hibridación in Situ , Diferenciación Celular/fisiología
19.
Nature ; 447(7142): 316-20, 2007 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-17507982

RESUMEN

The mammalian hair follicle is a complex 'mini-organ' thought to form only during development; loss of an adult follicle is considered permanent. However, the possibility that hair follicles develop de novo following wounding was raised in studies on rabbits, mice and even humans fifty years ago. Subsequently, these observations were generally discounted because definitive evidence for follicular neogenesis was not presented. Here we show that, after wounding, hair follicles form de novo in genetically normal adult mice. The regenerated hair follicles establish a stem cell population, express known molecular markers of follicle differentiation, produce a hair shaft and progress through all stages of the hair follicle cycle. Lineage analysis demonstrated that the nascent follicles arise from epithelial cells outside of the hair follicle stem cell niche, suggesting that epidermal cells in the wound assume a hair follicle stem cell phenotype. Inhibition of Wnt signalling after re-epithelialization completely abrogates this wounding-induced folliculogenesis, whereas overexpression of Wnt ligand in the epidermis increases the number of regenerated hair follicles. These remarkable regenerative capabilities of the adult support the notion that wounding induces an embryonic phenotype in skin, and that this provides a window for manipulation of hair follicle neogenesis by Wnt proteins. These findings suggest treatments for wounds, hair loss and other degenerative skin disorders.


Asunto(s)
Folículo Piloso/citología , Folículo Piloso/crecimiento & desarrollo , Regeneración/fisiología , Proteínas Wnt/metabolismo , Cicatrización de Heridas/fisiología , Animales , Linaje de la Célula , Células Epiteliales/citología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Transducción de Señal/efectos de los fármacos , Células Madre/citología , Heridas y Lesiones/patología
20.
Ocul Surf ; 29: 486-494, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37453535

RESUMEN

PURPOSE: To investigate the role of Wnt/ß-catenin signaling in mouse eyelid development. METHODS: Wnt/ß-catenin signaling was disrupted by deleting supraorbital mesenchymal ß-catenin or epithelial Wls. p63 was removed to determine whether the expression of Wnts is affected. The eyelid morphology was examined at different stages. Proliferation, apoptosis, and expression of Wnt ligands and their target genes were analyzed via immunofluorescence staining, TUNEL assay, and in situ hybridization. RESULTS: Deletion of ß-catenin in supraorbital mesenchyme abolishes eyelid growth by causing decreased proliferation in supraorbital epithelium and underlying mesenchyme. Inhibition of Wnt secretion by deleting Wls in supraorbital epithelium results in failure of eyelid development, similar to the effects of deleting mesenchymal ß-catenin. Knockout of p63 results in formation of hypoplastic eyelids and reduced expression of several Wnt ligands in eyelid epithelium. CONCLUSIONS: Epithelial Wnt ligands activate mesenchymal Wnt/ß-catenin signaling to control eyelid growth and their expression is partially regulated by p63.


Asunto(s)
Vía de Señalización Wnt , beta Catenina , Ratones , Animales , beta Catenina/genética , beta Catenina/metabolismo , Ligandos , Ratones Noqueados , Epitelio/metabolismo , Vía de Señalización Wnt/genética , Proliferación Celular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA