Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Genes Dev ; 35(5-6): 354-366, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33602871

RESUMEN

Polycomb repressive complex 1 (PRC1) and PRC2 are critical epigenetic developmental regulators. PRC1 and PRC2 largely overlap in their genomic binding and cooperate to establish repressive chromatin domains demarcated by H2AK119ub and H3K27me3. However, the functional contribution of each complex to gene repression has been a subject of debate, and understanding of its physiological significance requires further studies. Here, using the developing murine epidermis as a paradigm, we uncovered a previously unappreciated functional redundancy between Polycomb complexes. Coablation of PRC1 and PRC2 in embryonic epidermal progenitors resulted in severe defects in epidermal stratification, a phenotype not observed in the single PRC1-null or PRC2-null epidermis. Molecular dissection indicated a loss of epidermal identity that was coupled to a strong derepression of nonlineage transcription factors, otherwise repressed by either PRC1 or PRC2 in the absence of its counterpart. Ectopic expression of subsets of PRC1/2-repressed nonepidermal transcription factors in wild-type epidermal stem cells was sufficient to suppress epidermal identity genes, highlighting the importance of functional redundancy between PRC1 and PRC2. Altogether, our studies show how PRC1 and PRC2 function as two independent counterparts, thereby providing a repressive safety net that protects and preserves lineage identity.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/citología , Células Epidérmicas/citología , Complejo Represivo Polycomb 1/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Animales , Células Madre Embrionarias/metabolismo , Células Epidérmicas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Humanos , Ratones , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 2/genética , Proteínas del Grupo Polycomb/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
2.
Genes Dev ; 33(1-2): 55-60, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30567998

RESUMEN

Polycomb-repressive complex 1 (PRC1) and PRC2 are critical chromatin regulators of gene expression and tissue development. Here, we show that despite extensive genomic cobinding, PRC1 is essential for epidermal integrity, whereas PRC2 is dispensable. Loss of PRC1 resulted in blistering skin, reminiscent of human skin fragility syndromes. Conversely, PRC1 does not restrict epidermal stratification during skin morphogenesis, whereas PRC2 does. Molecular dissection demonstrated that PRC1 functions with PRC2 to silence/dampen expression of adhesion genes. In contrast, PRC1 promotes expression of critical epidermal adhesion genes independently of PRC2-mediated H3K27me3. Together, we demonstrate a functional link between epigenetic regulation and skin diseases.


Asunto(s)
Células Epidérmicas/fisiología , Epidermis/fisiología , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica/genética , Complejo Represivo Polycomb 1/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Enfermedades de la Piel/genética , Animales , Adhesión Celular/genética , Epidermis/crecimiento & desarrollo , Histonas/metabolismo , Ratones , Complejo Represivo Polycomb 1/genética , Enfermedades de la Piel/fisiopatología
3.
Genes Dev ; 31(6): 553-566, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28404630

RESUMEN

The female mammary gland is a very dynamic organ that undergoes continuous tissue remodeling during adulthood. Although it is well established that the number of menstrual cycles and pregnancy (in this case transiently) increase the risk of breast cancer, the reasons are unclear. Growing clinical and experimental evidence indicates that improper involution plays a role in the development of this malignancy. Recently, we described the miR-424(322)/503 cluster as an important regulator of mammary epithelial involution after pregnancy. Here, through the analysis of ∼3000 primary tumors, we show that miR-424(322)/503 is commonly lost in a subset of aggressive breast cancers and describe the genetic aberrations that inactivate its expression. Furthermore, through the use of a knockout mouse model, we demonstrate for the first time that loss of miR-424(322)/503 promotes breast tumorigenesis in vivo. Remarkably, we found that loss of miR-424(322)/503 promotes chemoresistance due to the up-regulation of two of its targets: BCL-2 and insulin-like growth factor-1 receptor (IGF1R). Importantly, targeted therapies blocking the aberrant activity of these targets restore sensitivity to chemotherapy. Overall, our studies reveal miR-424(322)/503 as a tumor suppressor in breast cancer and provide a link between mammary epithelial involution, tumorigenesis, and the phenomenon of chemoresistance.


Asunto(s)
Neoplasias de la Mama/genética , MicroARNs/genética , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/mortalidad , Línea Celular Tumoral , Resistencia a Antineoplásicos , Femenino , Eliminación de Gen , Genes Supresores de Tumor , Humanos , Neoplasias Mamarias Experimentales/genética , Ratones , Embarazo , Complicaciones Neoplásicas del Embarazo/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Receptor IGF Tipo 1 , Receptores de Somatomedina/genética , Fosfatasas cdc25/genética
4.
Cell ; 136(6): 1122-35, 2009 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-19303854

RESUMEN

Although in vitro studies of embryonic stem cells have identified polycomb repressor complexes (PRCs) as key regulators of differentiation, it remains unclear as to how PRC-mediated mechanisms control fates of multipotent progenitors in developing tissues. Here, we show that an essential PRC component, Ezh2, is expressed in epidermal progenitors but diminishes concomitant with embryonic differentiation and with postnatal decline in proliferative activity. We show that Ezh2 controls proliferative potential of basal progenitors by repressing the Ink4A-Ink4B locus and tempers the developmental rate of differentiation by preventing premature recruitment of AP1 transcriptional activator to the structural genes that are required for epidermal differentiation. Together, our studies reveal that PRCs control epigenetic modifications temporally and spatially in tissue-restricted stem cells. They maintain their proliferative potential and globally repressing undesirable differentiation programs while selectively establishing a specific terminal differentiation program in a stepwise fashion.


Asunto(s)
Diferenciación Celular , Células Epidérmicas , Epidermis/metabolismo , Regulación del Desarrollo de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/metabolismo , Células Madre/metabolismo , Animales , Núcleo Celular/metabolismo , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Proteína Potenciadora del Homólogo Zeste 2 , Histonas/metabolismo , Humanos , Metilación , Ratones , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Proteínas Represoras/metabolismo
5.
Cell ; 137(6): 1047-61, 2009 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-19524508

RESUMEN

Identification of bona fide tumor suppressors is often challenging because of the large number of genetic alterations present in most human cancers. To evaluate candidate genes present within chromosomal regions recurrently deleted in human cancers, we coupled high-resolution genomic analysis with a two-stage genetic study using RNA interference (RNAi). We found that Cyfip1, a subunit of the WAVE complex, which regulates cytoskeletal dynamics, is commonly deleted in human epithelial cancers. Reduced expression of CYFIP1 is commonly observed during invasion of epithelial tumors and is associated with poor prognosis in this setting. Silencing of Cyfip1 disturbed normal epithelial morphogenesis in vitro and cooperated with oncogenic Ras to produce invasive carcinomas in vivo. Mechanistically, we have linked alterations in WAVE-regulated actin dynamics with impaired cell-cell adhesion and cell-ECM interactions. Thus, we propose Cyfip1 as an invasion suppressor gene.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinoma/metabolismo , Invasividad Neoplásica , Animales , Carcinoma/diagnóstico , Carcinoma/patología , Línea Celular Tumoral , Células Cultivadas , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Queratinocitos/metabolismo , Ratones , Trasplante de Neoplasias , Trasplante Heterólogo
6.
PLoS Genet ; 17(12): e1009948, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34905545

RESUMEN

Hair follicle stem cells (HFSCs) are multipotent cells that cycle through quiescence and activation to continuously fuel the production of hair follicles. Prior genome mapping studies had shown that tri-methylation of histone H3 at lysine 27 (H3K27me3), the chromatin mark mediated by Polycomb Repressive Complex 2 (PRC2), is dynamic between quiescent and activated HFSCs, suggesting that transcriptional changes associated with H3K27me3 might be critical for proper HFSC function. However, functional in vivo studies elucidating the role of PRC2 in adult HFSCs are lacking. In this study, by using in vivo loss-of-function studies we show that, surprisingly, PRC2 plays a non-instructive role in adult HFSCs and loss of PRC2 in HFSCs does not lead to loss of HFSC quiescence or changes in cell identity. Interestingly, RNA-seq and immunofluorescence analyses of PRC2-null quiescent HFSCs revealed upregulation of genes associated with activated state of HFSCs. Altogether, our findings show that transcriptional program under PRC2 regulation is dispensable for maintaining HFSC quiescence and hair regeneration.


Asunto(s)
Folículo Piloso/crecimiento & desarrollo , Cabello/crecimiento & desarrollo , Histonas/genética , Complejo Represivo Polycomb 2/genética , Regeneración/genética , Células Madre Adultas/metabolismo , Animales , Cromatina/genética , Cabello/metabolismo , Folículo Piloso/metabolismo , Humanos , Metilación , Ratones , RNA-Seq , Transducción de Señal/genética
7.
Genes Dev ; 30(20): 2325-2338, 2016 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-27807033

RESUMEN

Growth and regeneration of one tissue within an organ compels accommodative changes in the surrounding tissues. However, the molecular nature and operating logic governing these concurrent changes remain poorly defined. The dermal adipose layer expands concomitantly with hair follicle downgrowth, providing a paradigm for studying coordinated changes of surrounding lineages with a regenerating tissue. Here, we discover that hair follicle transit-amplifying cells (HF-TACs) play an essential role in orchestrating dermal adipogenesis through secreting Sonic Hedgehog (SHH). Depletion of Shh from HF-TACs abrogates both dermal adipogenesis and hair follicle growth. Using cell type-specific deletion of Smo, a gene required in SHH-receiving cells, we found that SHH does not act on hair follicles, adipocytes, endothelial cells, and hematopoietic cells for adipogenesis. Instead, SHH acts directly on adipocyte precursors, promoting their proliferation and their expression of a key adipogenic gene, peroxisome proliferator-activated receptor γ (Pparg), to induce dermal adipogenesis. Our study therefore uncovers a critical role for TACs in orchestrating the generation of both their own progeny and a neighboring lineage to achieve concomitant tissue production across lineages.


Asunto(s)
Adipogénesis/fisiología , Folículo Piloso/citología , Folículo Piloso/metabolismo , Proteínas Hedgehog/metabolismo , Piel/metabolismo , Adipogénesis/genética , Animales , Proliferación Celular/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Folículo Piloso/embriología , Folículo Piloso/crecimiento & desarrollo , Masculino , Ratones , Transducción de Señal , Piel/embriología , Piel/crecimiento & desarrollo
8.
Development ; 147(22)2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-33191273

RESUMEN

Cell divisions and cell-fate decisions require stringent regulation for proper tissue development and homeostasis. The mammalian epidermis is a highly organized tissue structure that is sustained by epidermal stem cells (ESCs) that balance self-renewal and cell-fate decisions to establish a protective barrier, while replacing dying cells during homeostasis and in response to injury. Extensive work over past decades has provided insights into the regulatory mechanisms that control ESC specification, self-renewal and maintenance during different stages of the lifetime of an organism. In this Review, we discuss recent findings that have furthered our understanding of key regulatory features that allow ESCs to establish a functional barrier during development and to maintain tissue homeostasis in adults.


Asunto(s)
Células Epidérmicas/metabolismo , Epidermis/embriología , Epidermis/crecimiento & desarrollo , Homeostasis/genética , Células Madre/metabolismo , Animales , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Proliferación Celular/fisiología , Autorrenovación de las Células/fisiología , Humanos , Transcripción Genética , Cicatrización de Heridas/fisiología
9.
Genes Dev ; 29(2): 144-56, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25547114

RESUMEN

Altered epidermal differentiation characterizes numerous skin diseases affecting >25% of the human population. Here we identified Fra-2/AP-1 as a key regulator of terminal epidermal differentiation. Epithelial-restricted, ectopic expression of Fra-2 induced expression of epidermal differentiation genes located within the epidermal differentiation complex (EDC). Moreover, in a papilloma-prone background, a reduced tumor burden was observed due to precocious keratinocyte differentiation by Fra-2 expression. Importantly, loss of Fra-2 in suprabasal keratinocytes is sufficient to cause skin barrier defects due to reduced expression of differentiation genes. Mechanistically, Fra-2 binds and transcriptionally regulates EDC gene promoters, which are co-occupied by the transcriptional repressor Ezh2. Fra-2 remains transcriptionally inactive in nondifferentiated keratinocytes, where it was found monomethylated and dimethylated on Lys104 and interacted with Ezh2. Upon keratinocyte differentiation, Fra-2 is C-terminally phosphorylated on Ser320 and Thr322 by ERK1/2, leading to transcriptional activation. Thus, the induction of epidermal differentiation by Fra-2 is controlled by a dual mechanism involving Ezh2-dependent methylation and activation by ERK1/2-dependent phosphorylation.


Asunto(s)
Diferenciación Celular , Antígeno 2 Relacionado con Fos/metabolismo , Queratinocitos/citología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Factor de Transcripción AP-1/metabolismo , Animales , Células Cultivadas , Embrión de Mamíferos , Proteína Potenciadora del Homólogo Zeste 2 , Regulación del Desarrollo de la Expresión Génica , Lisina/metabolismo , Metilación , Ratones , Fosforilación
10.
Bioessays ; 42(5): e1900192, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32196702

RESUMEN

Polycomb group proteins are evolutionary conserved chromatin-modifying complexes, essential for the regulation of developmental and cell-identity genes. Polycomb-mediated transcriptional regulation is provided by two multi-protein complexes known as Polycomb repressive complex 1 (PRC1) and 2 (PRC2). Recent studies positioned PRC1 as a foremost executer of Polycomb-mediated transcriptional control. Mammalian PRC1 complexes can form multiple sub-complexes that vary in their core and accessory subunit composition, leading to fascinating and diverse transcriptional regulatory mechanisms employed by PRC1 complexes. These mechanisms include PRC1-catalytic activity toward monoubiquitination of histone H2AK119, a well-established hallmark of PRC1 complexes, whose importance has been long debated. In this review, the central roles that PRC1-catalytic activity plays in transcriptional repression are emphasized and the recent evidence supporting a role for PRC1 complexes in gene activation is discussed.


Asunto(s)
Histonas , Complejo Represivo Polycomb 1 , Animales , Proteínas de Ciclo Celular , Cromatina , Histonas/metabolismo , Humanos , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Ubiquitinación
11.
Proc Natl Acad Sci U S A ; 116(40): 20104-20114, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31527246

RESUMEN

Viral cancers show oncogene addiction to viral oncoproteins, which are required for survival and proliferation of the dedifferentiated cancer cell. Human Merkel cell carcinomas (MCCs) that harbor a clonally integrated Merkel cell polyomavirus (MCV) genome have low mutation burden and require viral T antigen expression for tumor growth. Here, we showed that MCV+ MCC cells cocultured with keratinocytes undergo neuron-like differentiation with neurite outgrowth, secretory vesicle accumulation, and the generation of sodium-dependent action potentials, hallmarks of a neuronal cell lineage. Cocultured keratinocytes are essential for induction of the neuronal phenotype. Keratinocyte-conditioned medium was insufficient to induce this phenotype. Single-cell RNA sequencing revealed that T antigen knockdown inhibited cell cycle gene expression and reduced expression of key Merkel cell lineage/MCC marker genes, including HES6, SOX2, ATOH1, and KRT20 Of these, T antigen knockdown directly inhibited Sox2 and Atoh1 expression. MCV large T up-regulated Sox2 through its retinoblastoma protein-inhibition domain, which in turn activated Atoh1 expression. The knockdown of Sox2 in MCV+ MCCs mimicked T antigen knockdown by inducing MCC cell growth arrest and neuron-like differentiation. These results show Sox2-dependent conversion of an undifferentiated, aggressive cancer cell to a differentiated neuron-like phenotype and suggest that the ontology of MCC arises from a neuronal cell precursor.


Asunto(s)
Antígenos Virales de Tumores/genética , Carcinoma de Células de Merkel/etiología , Carcinoma de Células de Merkel/metabolismo , Poliomavirus de Células de Merkel/genética , Fenotipo , Infecciones por Polyomavirus/complicaciones , Factores de Transcripción SOXB1/genética , Antígenos Virales de Tumores/inmunología , Antígenos Virales de Tumores/metabolismo , Carcinoma de Células de Merkel/patología , Ciclo Celular/genética , Línea Celular Tumoral , Linaje de la Célula/genética , Transformación Celular Viral , Técnicas de Silenciamiento del Gen , Humanos , Queratinocitos , Células de Merkel/metabolismo , Poliomavirus de Células de Merkel/inmunología , Neuritas/metabolismo , Neuronas/metabolismo , Infecciones por Polyomavirus/inmunología , Infecciones por Polyomavirus/virología , Factores de Transcripción SOXB1/metabolismo , Infecciones Tumorales por Virus/complicaciones , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/virología
12.
Genes Dev ; 28(7): 765-82, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24636986

RESUMEN

The mammary gland is a very dynamic organ that undergoes continuous remodeling. The critical regulators of this process are not fully understood. Here we identify the microRNA cluster miR-424(322)/503 as an important regulator of epithelial involution after pregnancy. Through the generation of a knockout mouse model, we found that regression of the secretory acini of the mammary gland was compromised in the absence of miR-424(322)/503. Mechanistically, we show that miR-424(322)/503 orchestrates cell life and death decisions by targeting BCL-2 and IGF1R (insulin growth factor-1 receptor). Furthermore, we demonstrate that the expression of this microRNA cluster is regulated by TGF-ß, a well-characterized regulator of mammary involution. Overall, our data suggest a model in which activation of the TGF-ß pathway after weaning induces the transcription of miR-424(322)/503, which in turn down-regulates the expression of key genes. Here, we unveil a previously unknown, multilayered regulation of epithelial tissue remodeling coordinated by the microRNA cluster miR-424(322)/503.


Asunto(s)
Epitelio/metabolismo , Regulación del Desarrollo de la Expresión Génica , Glándulas Mamarias Animales/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Animales , Muerte Celular/genética , Línea Celular , Femenino , Técnicas de Inactivación de Genes , Humanos , Glándulas Mamarias Animales/citología , Ratones Noqueados , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptor IGF Tipo 1/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Destete
13.
Exp Dermatol ; 28(4): 374-382, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30758073

RESUMEN

Merkel cells are mechanosensory cells involved in tactile discrimination. Merkel cells have been primarily studied in the murine back skin, where they are found in specialized structures called touch domes located around primary hair follicles. Yet, little is known about the morphogenesis of Merkel cells in areas of the skin devoid of hair, such as the glabrous paw skin. Here, we describe Merkel cell formation in the glabrous paw skin during embryogenesis. We first found in the glabrous paw skin that Merkel cells were specified at E15.5, 24 hours later, compared to in the back skin. Additionally, by performing lineage-tracing experiments, we found that unlike in the back skin, SOX9(+) cells do not give rise to Merkel cells in the glabrous paw skin. Finally, we compared the transcriptomes of Merkel cells in the back and the glabrous paw skin and showed that they are similar. Genetic and transcriptome studies showed that the formation of Merkel cells in both regions was controlled by similar regulators. Among them was FGFR2, an upstream factor of MAPK signalling that was reported to have a critical function in Merkel cell formation in the back skin. Here, we showed that FGFR2 is also required for Merkel cell development in the glabrous paw skin. Taken together, our results demonstrate that Merkel cells in the murine back skin and glabrous paw skin are similar, and even though their formation is controlled by a common genetic programme, their precursor cells might differ.


Asunto(s)
Células de Merkel , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Piel/embriología , Animales , Femenino , Ratones , Embarazo , Piel/citología , Transcriptoma
14.
PLoS Genet ; 12(7): e1006151, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27414999

RESUMEN

An increasing amount of evidence indicates that developmental programs are tightly regulated by the complex interplay between signaling pathways, as well as transcriptional and epigenetic processes. Here, we have uncovered coordination between transcriptional and morphogen cues to specify Merkel cells, poorly understood skin cells that mediate light touch sensations. In murine dorsal skin, Merkel cells are part of touch domes, which are skin structures consisting of specialized keratinocytes, Merkel cells, and afferent neurons, and are located exclusively around primary hair follicles. We show that the developing primary hair follicle functions as a niche required for Merkel cell specification. We find that intraepidermal Sonic hedgehog (Shh) signaling, initiated by the production of Shh ligand in the developing hair follicles, is required for Merkel cell specification. The importance of Shh for Merkel cell formation is further reinforced by the fact that Shh overexpression in embryonic epidermal progenitors leads to ectopic Merkel cells. Interestingly, Shh signaling is common to primary, secondary, and tertiary hair follicles, raising the possibility that there are restrictive mechanisms that regulate Merkel cell specification exclusively around primary hair follicles. Indeed, we find that loss of Polycomb repressive complex 2 (PRC2) in the epidermis results in the formation of ectopic Merkel cells that are associated with all hair types. We show that PRC2 loss expands the field of epidermal cells competent to differentiate into Merkel cells through the upregulation of key Merkel-differentiation genes, which are known PRC2 targets. Importantly, PRC2-mediated repression of the Merkel cell differentiation program requires inductive Shh signaling to form mature Merkel cells. Our study exemplifies how the interplay between epigenetic and morphogen cues regulates the complex patterning and formation of the mammalian skin structures.


Asunto(s)
Proteínas Hedgehog/fisiología , Células de Merkel/citología , Complejo Represivo Polycomb 2/fisiología , Transducción de Señal , Piel/embriología , Animales , Linaje de la Célula , Proliferación Celular , Epidermis/embriología , Epidermis/metabolismo , Epigénesis Genética , Femenino , Perfilación de la Expresión Génica , Folículo Piloso/embriología , Queratinocitos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Piel/metabolismo , Células Madre/citología , Transcripción Genética
15.
Genes Dev ; 25(5): 485-98, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21317239

RESUMEN

Polycomb protein group (PcG)-dependent trimethylation on H3K27 (H3K27me3) regulates identity of embryonic stem cells (ESCs). How H3K27me3 governs adult SCs and tissue development is unclear. Here, we conditionally target H3K27 methyltransferases Ezh2 and Ezh1 to address their roles in mouse skin homeostasis. Postnatal phenotypes appear only in doubly targeted skin, where H3K27me3 is abolished, revealing functional redundancy in EZH1/2 proteins. Surprisingly, while Ezh1/2-null hair follicles (HFs) arrest morphogenesis and degenerate due to defective proliferation and increased apoptosis, epidermis hyperproliferates and survives engraftment. mRNA microarray studies reveal that, despite these striking phenotypic differences, similar genes are up-regulated in HF and epidermal Ezh1/2-null progenitors. Featured prominently are (1) PcG-controlled nonskin lineage genes, whose expression is still significantly lower than in native tissues, and (2) the PcG-regulated Ink4a/Inkb/Arf locus. Interestingly, when EZH1/2 are absent, even though Ink4a/Arf/Ink4b genes are fully activated in HF cells, they are only partially so in epidermal progenitors. Importantly, transduction of Ink4b/Ink4a/Arf shRNAs restores proliferation/survival of Ezh1/2-null HF progenitors in vitro, pointing toward the relevance of this locus to the observed HF phenotypes. Our findings reveal new insights into Polycomb-dependent tissue control, and provide a new twist to how different progenitors within one tissue respond to loss of H3K27me3.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Folículo Piloso/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Homeostasis/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Factores de Transcripción/metabolismo , Cicatrización de Heridas/genética , Factor 1 de Ribosilacion-ADP/genética , Apoptosis/genética , Proliferación Celular , Supervivencia Celular/genética , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Proteínas de Unión al ADN/genética , Proteína Potenciadora del Homólogo Zeste 2 , Células Epidérmicas , Epidermis/trasplante , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Folículo Piloso/citología , N-Metiltransferasa de Histona-Lisina/genética , Metilación , Complejo Represivo Polycomb 2 , Trasplante de Piel , Células Madre/metabolismo , Factores de Transcripción/genética
16.
EMBO J ; 32(14): 1990-2000, 2013 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-23673358

RESUMEN

While the Polycomb complex is known to regulate cell identity in ES cells, its role in controlling tissue-specific stem cells is not well understood. Here we show that removal of Ezh1 and Ezh2, key Polycomb subunits, from mouse skin results in a marked change in fate determination in epidermal progenitor cells, leading to an increase in the number of lineage-committed Merkel cells, a specialized subtype of skin cells involved in mechanotransduction. By dissecting the genetic mechanism, we showed that the Polycomb complex restricts differentiation of epidermal progenitor cells by repressing the transcription factor Sox2. Ablation of Sox2 results in a dramatic loss of Merkel cells, indicating that Sox2 is a critical regulator of Merkel cell specification. We show that Sox2 directly activates Atoh1, the obligate regulator of Merkel cell differentiation. Concordantly, ablation of Sox2 attenuated the Ezh1/2-null phenotype, confirming the importance of Polycomb-mediated repression of Sox2 in maintaining the epidermal progenitor cell state. Together, these findings define a novel regulatory network by which the Polycomb complex maintains the progenitor cell state and governs differentiation in vivo.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Células de Merkel/citología , Células de Merkel/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proteína Potenciadora del Homólogo Zeste 2 , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Complejo Represivo Polycomb 2/deficiencia , Complejo Represivo Polycomb 2/genética , Embarazo , Factores de Transcripción SOXB1/deficiencia , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
Development ; 141(24): 4690-6, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25468937

RESUMEN

Merkel cell-neurite complexes are located in touch-sensitive areas of the mammalian skin and are involved in recognition of the texture and shape of objects. Merkel cells are essential for these tactile discriminations, as they generate action potentials in response to touch stimuli and induce the firing of innervating afferent nerves. It has been shown that Merkel cells originate from epidermal stem cells, but the cellular and molecular mechanisms of their development are largely unknown. In this study, we analyzed Merkel cell differentiation during development and found that it is a temporally regulated maturation process characterized by a sequential activation of Merkel cell-specific genes. We uncovered key transcription factors controlling this process and showed that the transcription factor Atoh1 is required for initial Merkel cell specification. The subsequent maturation steps of Merkel cell differentiation are controlled by cooperative function of the transcription factors Sox2 and Isl1, which physically interact and work to sustain Atoh1 expression. These findings reveal the presence of a robust transcriptional network required to produce functional Merkel cells that are required for tactile discrimination.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Redes Reguladoras de Genes/fisiología , Células de Merkel/fisiología , Piel/embriología , Animales , Western Blotting , Técnica del Anticuerpo Fluorescente , Redes Reguladoras de Genes/genética , Humanos , Inmunoprecipitación , Indoles , Proteínas con Homeodominio LIM/metabolismo , Ratones , Microscopía Fluorescente , Factores de Transcripción SOXB1/metabolismo , Piel/citología , Factores de Transcripción/metabolismo
18.
Methods Mol Biol ; 2736: 9-21, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37615890

RESUMEN

Cleavage Under Targets & Release Using Nuclease (CUT&RUN) has swiftly become the preferred procedure over the past few years for genomic mapping and detecting interactions between chromatin and its bound proteins. CUT&RUN is now being widely used for characterizing the epigenetic landscape in many cell types as it utilizes far less cell numbers when compared to Chromatin Immunoprecipitation-sequencing (ChIP-seq), thereby making it a powerful tool for researchers working with limited material. This protocol has been specifically optimized for detecting histone modifications in fluorescence-activated cell sorting (FACS)-isolated epidermal stem cells from adult mice.


Asunto(s)
Cromatina , Código de Histonas , Ratones , Animales , Procesamiento Proteico-Postraduccional , Células Madre , Epigenómica/métodos
19.
Dev Cell ; 59(1): 1-3, 2024 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-38194909

RESUMEN

Eccrine glands secrete water onto the surface of human skin to regulate body temperature. In this issue of Developmental Cell, Dingwall et al. dissect the transcriptional signature of developing eccrine glands, and they also uncover a unique dermal niche that is responsible for promoting eccrine gland developmental progression.


Asunto(s)
Glándulas Ecrinas , Piel , Humanos
20.
Sci Adv ; 10(3): eadi5791, 2024 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-38241368

RESUMEN

The touch dome (TD) keratinocytes are specialized epidermal cells that intimately associate with the light touch sensing Merkel cells (MCs). The TD keratinocytes function as a niche for the MCs and can induce de novo hair follicles upon stimulation; however, how the TD keratinocytes are maintained during homeostasis remains unclear. scRNA-seq identified a specific TD keratinocyte marker, Tenascin-C (TNC). Lineage tracing of Tnc-expressing TD keratinocytes revealed that these cells maintain themselves as an autonomous epidermal compartment and give rise to MCs upon injury. Molecular characterization uncovered that, while the transcriptional and chromatin landscape of the TD keratinocytes is remarkably similar to that of the interfollicular epidermal keratinocytes, it also shares certain molecular signatures with the hair follicle keratinocytes. Our study highlights that the TD keratinocytes in the adult skin have molecular characteristics of keratinocytes of diverse epidermal lineages.


Asunto(s)
Queratinocitos , Tenascina , Tenascina/genética , Epidermis , Piel , Células de Merkel/fisiología , Folículo Piloso
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA