Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Invest Dermatol ; 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38431220

RESUMEN

NIPP1 is a ubiquitously expressed regulatory subunit of PP1. Its embryonic deletion in keratinocytes causes chronic sterile skin inflammation, epidermal hyperproliferation, and resistance to mutagens in adult mice. To explore the primary effects of NIPP1 deletion, we first examined hair cycle progression of NIPP1 skin knockouts (SKOs). The entry of the first hair cycle in the SKOs was delayed owing to prolonged quiescence of hair follicle stem cells. In contrast, the entry of the second hair cycle in the SKOs was advanced as a result of precocious activation of hair follicle stem cells. The epidermis of SKOs progressively accumulated senescent cells, and this cell-fate switch was accelerated by DNA damage. Primary keratinocytes from SKO neonates and human NIPP1-depleted HaCaT keratinocytes failed to proliferate and showed an increase in the expression of cell cycle inhibitors (p21, p16/Ink4a, and/or p19/Arf) and senescence-associated-secretory-phenotype factors as well as in DNA damage (γH2AX and 53BP1). Our data demonstrate that the primary effect of NIPP1 deletion in keratinocytes is a cell cycle arrest and premature senescence that gradually progresse to chronic senescence and likely contribute to the decreased sensitivity of SKOs to mutagens.

2.
Methods Mol Biol ; 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38407797

RESUMEN

The mammalian integumentary system, including skin and its appendages, serves as a protective barrier for the body. During development, skin epidermis undergoes rapid cell division and differentiation to form multiple stratified layers of keratinocytes. Concurrently the epidermis also gives rise to hair follicles that invaginate into the dermis. In adult skin, the hair follicle undergoes cyclic regeneration fueled by hair follicle stem cells located in the bulge. Three-dimensional and high-resolution imaging of these structures using whole-mount immunofluorescent staining allows to better characterize epidermal progenitors and stem cells.

4.
Nat Commun ; 13(1): 4449, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35915068

RESUMEN

Hair follicles undergo cycles of regeneration fueled by hair follicle stem cells (HFSCs). While ß-catenin-dependent canonical Wnt signaling has been extensively studied and implicated in HFSC activation and fate determination, very little is known about the function of ß-catenin-independent Wnt signaling in HFSCs. In this study, we investigate the functional role of ROR2, a Wnt receptor, in HFSCs. By analyzing Ror2-depleted HFSCs, we uncover that ROR2 is not only essential to regulate Wnt-activated signaling that is responsible for HFSC activation and self-renewal, but it is also required to maintain proper ATM/ATR-dependent DNA damage response, which is indispensable for the long-term maintenance of HFSCs. In analyzing HFSCs lacking ß-catenin, we identify a compensatory role of ROR2-PKC signaling in protecting ß-catenin-null HFSCs from the loss of stem cell pool. Collectively, our study unveils a previously unrecognized role of ROR2 in regulation of stem cell self-renewal and maintenance.


Asunto(s)
Folículo Piloso , beta Catenina , Folículo Piloso/metabolismo , Células Madre/metabolismo , Vía de Señalización Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
5.
Cancers (Basel) ; 11(9)2019 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-31438551

RESUMEN

The skin functions as a barrier between the organism and the surrounding environment. Direct exposure to external stimuli and the accumulation of genetic mutations may lead to abnormal cell growth, irreversible tissue damage and potentially favor skin malignancy. Skin homeostasis is coordinated by an intricate signaling network, and its dysregulation has been implicated in the development of skin cancers. Wnt signaling is one such regulatory pathway orchestrating skin development, homeostasis, and stem cell activation. Aberrant regulation of Wnt signaling cascades not only gives rise to tumor initiation, progression and invasion, but also maintains cancer stem cells which contribute to tumor recurrence. In this review, we summarize recent studies highlighting functional evidence of Wnt-related oncology in keratinocyte carcinomas, as well as discussing preclinical and clinical approaches that target oncogenic Wnt signaling to treat cancers. Our review provides valuable insight into the significance of Wnt signaling for future interventions against keratinocyte carcinomas.

6.
Cell Stem Cell ; 22(3): 398-413.e7, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29337183

RESUMEN

Tissue regeneration relies on resident stem cells (SCs), whose activity and lineage choices are influenced by the microenvironment. Exploiting the synchronized, cyclical bouts of tissue regeneration in hair follicles (HFs), we investigate how microenvironment dynamics shape the emergence of stem cell lineages. Employing epigenetic and ChIP-seq profiling, we uncover how signal-dependent transcription factors couple spatiotemporal cues to chromatin dynamics, thereby choreographing stem cell lineages. Using enhancer-driven reporters, mutagenesis, and genetics, we show that simultaneous BMP-inhibitory and WNT signals set the stage for lineage choices by establishing chromatin platforms permissive for diversification. Mechanistically, when binding of BMP effector pSMAD1 is relieved, enhancers driving HF-stem cell master regulators are silenced. Concomitantly, multipotent, lineage-fated enhancers silent in HF-stem cells become activated by exchanging WNT effectors TCF3/4 for LEF1. Throughout regeneration, lineage enhancers continue reliance upon LEF1 but then achieve specificity by accommodating additional incoming signaling effectors. Barriers to progenitor plasticity increase when diverse, signal-sensitive transcription factors shape LEF1-regulated enhancer dynamics.


Asunto(s)
Linaje de la Célula , Ensamble y Desensamble de Cromatina , Folículo Piloso/citología , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Acetilación , Animales , Secuencia de Bases , Proteínas Morfogenéticas Óseas/metabolismo , Cromatina/metabolismo , Elementos de Facilitación Genéticos/genética , Histonas/metabolismo , Lisina/metabolismo , Ratones Endogámicos C57BL , Fosforilación , Regeneración , Proteína Smad1/metabolismo , Factores de Tiempo , Factores de Transcripción/metabolismo , Vía de Señalización Wnt
7.
Stem Cells ; 36(1): 22-35, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29047191

RESUMEN

Mammalian skin and its appendages constitute the integumentary system forming a barrier between the organism and its environment. During development, skin epidermal cells divide rapidly and stratify into a multilayered epithelium, as well as invaginate downward in the underlying mesenchyme to form hair follicles (HFs). In postnatal skin, the interfollicular epidermal (IFE) cells continuously proliferate and differentiate while HFs undergo cycles of regeneration. Epidermal regeneration is fueled by epidermal stem cells (SCs) located in the basal layer of the IFE and the outer layer of the bulge in the HF. Epidermal development and SC behavior are mainly regulated by various extrinsic cues, among which Wnt-dependent signaling pathways play crucial roles. This review not only summarizes the current knowledge of Wnt signaling pathways in the regulation of skin development and governance of SCs during tissue homeostasis, but also discusses the potential crosstalk of Wnt signaling with other pathways involved in these processes. Stem Cells 2018;36:22-35.


Asunto(s)
Epidermis/metabolismo , Piel/crecimiento & desarrollo , Células Madre/metabolismo , Diferenciación Celular , Proliferación Celular , Humanos , Vía de Señalización Wnt
8.
Dev Cell ; 41(5): 481-495.e5, 2017 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-28552558

RESUMEN

Malformations of the cerebral cortex (MCCs) are devastating developmental disorders. We report here that mice with embryonic neural stem-cell-specific deletion of Llgl1 (Nestin-Cre/Llgl1fl/fl), a mammalian ortholog of the Drosophila cell polarity gene lgl, exhibit MCCs resembling severe periventricular heterotopia (PH). Immunohistochemical analyses and live cortical imaging of PH formation revealed that disruption of apical junctional complexes (AJCs) was responsible for PH in Nestin-Cre/Llgl1fl/fl brains. While it is well known that cell polarity proteins govern the formation of AJCs, the exact mechanisms remain unclear. We show that LLGL1 directly binds to and promotes internalization of N-cadherin, and N-cadherin/LLGL1 interaction is inhibited by atypical protein kinase C-mediated phosphorylation of LLGL1, restricting the accumulation of AJCs to the basolateral-apical boundary. Disruption of the N-cadherin-LLGL1 interaction during cortical development in vivo is sufficient for PH. These findings reveal a mechanism responsible for the physical and functional connection between cell polarity and cell-cell adhesion machineries in mammalian cells.


Asunto(s)
Encéfalo/anomalías , Adhesión Celular/fisiología , Polaridad Celular/fisiología , Células Madre Embrionarias/fisiología , Proteínas de Homeodominio/fisiología , Células-Madre Neurales/fisiología , Heterotopia Nodular Periventricular/patología , Proteínas Supresoras de Tumor/fisiología , Animales , Apoptosis , Encéfalo/metabolismo , Encéfalo/patología , Cadherinas/genética , Cadherinas/metabolismo , Proliferación Celular , Células Cultivadas , Proteínas del Citoesqueleto , Células Madre Embrionarias/citología , Femenino , Ratones , Ratones Transgénicos , Nestina/genética , Nestina/metabolismo , Células-Madre Neurales/citología , Heterotopia Nodular Periventricular/metabolismo , Fosforilación
9.
Genes Dev ; 30(7): 798-811, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-27013234

RESUMEN

Cell-cell adhesion protein αE-catenin inhibits skin squamous cell carcinoma (SCC) development; however, the mechanisms responsible for this function are not completely understood. We report here that αE-catenin inhibits ß4 integrin-mediated activation of SRC tyrosine kinase.SRCis the first discovered oncogene, but the protein substrate critical for SRC-mediated transformation has not been identified. We found that YAP1, the pivotal effector of the Hippo signaling pathway, is a direct SRC phosphorylation target, and YAP1 phosphorylation at three sites in its transcription activation domain is necessary for SRC-YAP1-mediated transformation. We uncovered a marked increase in this YAP1 phosphorylation in human and mouse SCC tumors with low/negative expression of αE-catenin. We demonstrate that the tumor suppressor function of αE-catenin involves negative regulation of the ß4 integrin-SRC signaling pathway and that SRC-mediated phosphorylation and activation of YAP1 are an alternative to the canonical Hippo signaling pathway that directly connect oncogenic tyrosine kinase signaling with YAP1.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinoma de Células Escamosas/fisiopatología , Proteína Oncogénica pp60(v-src)/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal , alfa Catenina/metabolismo , Animales , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/genética , Proteínas de Ciclo Celular , Núcleo Celular/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/genética , Células Cultivadas , Regulación Neoplásica de la Expresión Génica , Humanos , Queratinocitos/citología , Queratinocitos/patología , Ratones , Fosforilación , Transporte de Proteínas , Proteínas Señalizadoras YAP
10.
Nat Commun ; 7: 10936, 2016 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-26939553

RESUMEN

Sequential generation of neurons and glial cells during development is critical for the wiring and function of the cerebral cortex. This process requires accurate coordination of neural progenitor cell (NPC) fate decisions, by NPC-autonomous mechanisms as well as by negative feedback from neurons. Here, we show that neurogenesis is protracted and gliogenesis decreased in mice with mutations of genes Celsr3 and Fzd3. This phenotype is not due to gene inactivation in progenitors, but rather in immature cortical neurons. Mutant neurons are unable to upregulate expression of Jag1 in response to cortical Wnt7, resulting in blunted activation of Notch signalling in NPC. Thus, Celsr3 and Fzd3 enable immature neurons to respond to Wnt7, upregulate Jag1 and thereby facilitate feedback signals that tune the timing of NPC fate decisions via Notch activation.


Asunto(s)
Cadherinas/metabolismo , Receptores Frizzled/metabolismo , Regulación de la Expresión Génica/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Superficie Celular/metabolismo , Transducción de Señal/fisiología , Proteínas Wnt/metabolismo , Animales , Bromodesoxiuridina , Cadherinas/genética , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/embriología , Femenino , Receptores Frizzled/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-1 , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Mutación , Neurogénesis/fisiología , Embarazo , Proteínas Proto-Oncogénicas/genética , Receptores de Superficie Celular/genética , Receptores Notch/genética , Receptores Notch/metabolismo , Proteínas Serrate-Jagged , Coloración y Etiquetado , Proteínas Wnt/genética
11.
Genes Dev ; 28(14): 1517-32, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25030692

RESUMEN

In mammals, Wnt/ß-catenin signaling features prominently in stem cells and cancers, but how and for what purposes have been matters of much debate. In this review, we summarize our current knowledge of Wnt/ß-catenin signaling and its downstream transcriptional regulators in normal and malignant stem cells. We centered this review largely on three types of stem cells--embryonic stem cells, hair follicle stem cells, and intestinal epithelial stem cells--in which the roles of Wnt/ß-catenin have been extensively studied. Using these models, we unravel how many controversial issues surrounding Wnt signaling have been resolved by dissecting the diversity of its downstream circuitry and effectors, often leading to opposite outcomes of Wnt/ß-catenin-mediated regulation and differences rooted in stage- and context-dependent effects.


Asunto(s)
Regulación de la Expresión Génica , Transducción de Señal , Células Madre/citología , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Humanos
12.
PLoS Comput Biol ; 10(6): e1003671, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24922058

RESUMEN

Recent studies have shown that the transcriptional functions of REST are much broader than repressing neuronal genes in non-neuronal systems. Whether REST occupies similar chromatin regions in different cell types and how it interacts with other transcriptional regulators to execute its functions in a context-dependent manner has not been adequately investigated. We have applied ChIP-seq analysis to identify the REST cistrome in human CD4+ T cells and compared it with published data from 15 other cell types. We found that REST cistromes were distinct among cell types, with REST binding to several tumor suppressors specifically in cancer cells, whereas 7% of the REST peaks in non-neuronal cells were ubiquitously called and <25% were identified for ≥ 5 cell types. Nevertheless, using a quantitative metric directly comparing raw ChIP-seq signals, we found the majority (∼80%) was shared by ≥ 2 cell types. Integration with RNA-seq data showed that REST binding was generally correlated with low gene expression. Close examination revealed that multiple contexts were correlated with reduced expression of REST targets, e.g., the presence of a cognate RE1 motif and cellular specificity of REST binding. These contexts were shown to play a role in differential corepressor recruitment. Furthermore, transcriptional outcome was highly influenced by REST cofactors, e.g., SIN3 and EZH2 co-occupancy marked higher and lower expression of REST targets, respectively. Unexpectedly, the REST cistrome in differentiated neurons exhibited unique features not observed in non-neuronal cells, e.g., the lack of RE1 motifs and an association with active gene expression. Finally, our analysis demonstrated how REST could differentially regulate a transcription network constituted of miRNAs, REST complex and neuronal factors. Overall, our findings of contexts playing critical roles in REST occupancy and regulatory outcome provide insights into the molecular interactions underlying REST's diverse functions, and point to novel roles of REST in differentiated neurons.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Regulación de la Expresión Génica/genética , Genómica/métodos , Neuronas/metabolismo , Proteínas Represoras/genética , Animales , Línea Celular , Inmunoprecipitación de Cromatina , Silenciador del Gen , Histonas/genética , Humanos , Ratones , MicroARNs/genética
13.
Nat Cell Biol ; 16(2): 179-90, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24463605

RESUMEN

Hair follicle stem cells (HFSCs) regenerate hair in response to Wnt signalling. Here, we unfold genome-wide transcriptional and chromatin landscapes of ß-catenin-TCF3/4-TLE circuitry, and genetically dissect their biological roles within the native HFSC niche. We show that during HFSC quiescence, TCF3, TCF4 and TLE (Groucho) bind coordinately and transcriptionally repress Wnt target genes. We also show that ß-catenin is dispensable for HFSC viability, and if TCF3/4 levels are sufficiently reduced, it is dispensable for proliferation. However, ß-catenin is essential to activate genes that launch hair follicle fate and suppress sebocyte fate determination. TCF3/4 deficiency mimics Wnt-ß-catenin-dependent activation of these hair follicle fate targets; TCF3 overexpression parallels their TLE4-dependent suppression. Our studies unveil TCF3/4-TLE histone deacetylases as a repressive rheostat, whose action can be relieved by Wnt-ß-catenin signalling. When TCF3/4 and TLE levels are high, HFSCs can maintain stemness, but remain quiescent. When these levels drop or when Wnt-ß-catenin levels rise, this balance is shifted and hair regeneration initiates.


Asunto(s)
Folículo Piloso/metabolismo , Transcripción Genética , Proteínas Wnt/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Folículo Piloso/citología , Ratones , Ratones Noqueados , Regeneración , Transducción de Señal , Factor de Transcripción 4 , beta Catenina/fisiología
14.
Proc Natl Acad Sci U S A ; 110(51): E4950-9, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24282298

RESUMEN

Hair production is fueled by stem cells (SCs), which transition between cyclical bouts of rest and activity. Here, we explore why hair growth wanes with age. We show that aged hair follicle SCs (HFSCs) in mice exhibit enhanced resting and abbreviated growth phases and are delayed in response to tissue-regenerating cues. Aged HFSCs are poor at initiating proliferation and show diminished self-renewing capacity upon extensive use. Only modestly restored by parabiosis, these features are rooted in elevated cell-intrinsic sensitivity and local elevation in bone morphogenic protein (BMP) signaling. Transcriptional profiling presents differences consistent with defects in aged HFSC activation. Notably, BMP-/calcium-regulated, nuclear factor of activated T-cell c1 (NFATc1) in HFSCs becomes recalcitrant to its normal down-regulating cues, and NFATc1 ChIP-sequencing analyses reveal a marked enrichment of NFATc1 target genes within the age-related signature. Moreover, aged HFSCs display more youthful levels of hair regeneration when BMP and/or NFATc1 are inhibited. These results provide unique insights into how skin SCs age.


Asunto(s)
Proliferación Celular , Folículo Piloso/metabolismo , Factores de Transcripción NFATC/metabolismo , Células Madre/metabolismo , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Células Cultivadas , Senescencia Celular/fisiología , Regulación de la Expresión Génica/fisiología , Folículo Piloso/citología , Ratones , Factores de Transcripción NFATC/genética , Transducción de Señal/fisiología , Envejecimiento de la Piel/fisiología , Células Madre/citología
15.
Nature ; 501(7466): 185-90, 2013 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-23945586

RESUMEN

Tissue growth is the multifaceted outcome of a cell's intrinsic capabilities and its interactions with the surrounding environment. Decoding these complexities is essential for understanding human development and tumorigenesis. Here we tackle this problem by carrying out the first genome-wide RNA-interference-mediated screens in mice. Focusing on skin development and oncogenic (Hras(G12V)-induced) hyperplasia, our screens uncover previously unknown as well as anticipated regulators of embryonic epidermal growth. Among the top oncogenic screen hits are Mllt6 and the Wnt effector ß-catenin, which maintain Hras(G12V)-dependent hyperproliferation. We also expose ß-catenin as an unanticipated antagonist of normal epidermal growth, functioning through Wnt-independent intercellular adhesion. Finally, we validate functional significance in mouse and human cancers, thereby establishing the feasibility of in vivo mammalian genome-wide investigations to dissect tissue development and tumorigenesis. By documenting some oncogenic growth regulators, we pave the way for future investigations of other hits and raise promise for unearthing new targets for cancer therapies.


Asunto(s)
Carcinogénesis/genética , Carcinogénesis/patología , Epidermis/patología , Neoplasias/genética , Neoplasias/patología , Oncogenes/genética , Interferencia de ARN , Animales , Carcinogénesis/metabolismo , Adhesión Celular , Proliferación Celular , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Epidermis/embriología , Epidermis/metabolismo , Femenino , Genoma/genética , Humanos , Hiperplasia/genética , Hiperplasia/metabolismo , Hiperplasia/patología , Masculino , Ratones , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Proteína Oncogénica p21(ras)/metabolismo , Reproducibilidad de los Resultados , Transducción de Señal , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Factores de Tiempo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , beta Catenina/deficiencia , beta Catenina/genética , beta Catenina/metabolismo
16.
Mol Cell ; 47(4): 633-47, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22841485

RESUMEN

DNA methylation is a mechanism of epigenetic regulation that is common to all vertebrates. Functional studies underscore its relevance for tissue homeostasis, but the global dynamics of DNA methylation during in vivo differentiation remain underexplored. Here we report high-resolution DNA methylation maps of adult stem cell differentiation in mouse, focusing on 19 purified cell populations of the blood and skin lineages. DNA methylation changes were locus specific and relatively modest in magnitude. They frequently overlapped with lineage-associated transcription factors and their binding sites, suggesting that DNA methylation may protect cells from aberrant transcription factor activation. DNA methylation and gene expression provided complementary information, and combining the two enabled us to infer the cellular differentiation hierarchy of the blood lineage directly from genome-scale data. In summary, these results demonstrate that in vivo differentiation of adult stem cells is associated with small but informative changes in the genomic distribution of DNA methylation.


Asunto(s)
Células Madre Adultas/citología , Células Sanguíneas/citología , Metilación de ADN , Piel/citología , Animales , Sitios de Unión , Ciclo Celular/genética , Diferenciación Celular/genética , Linaje de la Célula , Regulación hacia Abajo , Epigenómica , Expresión Génica , Genes Homeobox/genética , Sitios Genéticos , Genoma/genética , Linfocitos/citología , Ratones , Células Mieloides/citología
17.
Cell Stem Cell ; 9(3): 219-32, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21885018

RESUMEN

Using mouse skin, where bountiful reservoirs of synchronized hair follicle stem cells (HF-SCs) fuel cycles of regeneration, we explore how adult SCs remodel chromatin in response to activating cues. By profiling global mRNA and chromatin changes in quiescent and activated HF-SCs and their committed, transit-amplifying (TA) progeny, we show that polycomb-group (PcG)-mediated H3K27-trimethylation features prominently in HF-lineage progression by mechanisms distinct from embryonic-SCs. In HF-SCs, PcG represses nonskin lineages and HF differentiation. In TA progeny, nonskin regulators remain PcG-repressed, HF-SC regulators acquire H3K27me3-marks, and HF-lineage regulators lose them. Interestingly, genes poised in embryonic stem cells, active in HF-SCs, and PcG-repressed in TA progeny encode not only key transcription factors, but also signaling regulators. We document their importance in balancing HF-SC quiescence, underscoring the power of chromatin mapping in dissecting SC behavior. Our findings explain how HF-SCs cycle through quiescent and activated states without losing stemness and define roles for PcG-mediated repression in governing a fate switch irreversibly.


Asunto(s)
Células Madre Adultas/metabolismo , Folículo Piloso/metabolismo , Histonas/metabolismo , Proteínas Represoras/metabolismo , Células Madre Adultas/patología , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Ensamble y Desensamble de Cromatina , Estudio de Asociación del Genoma Completo , Regeneración Tisular Dirigida , Folículo Piloso/patología , Ratones , Proteínas del Grupo Polycomb , Procesamiento Proteico-Postraduccional , Transducción de Señal/genética , Piel/patología
18.
Sci Signal ; 4(174): ra33, 2011 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-21610251

RESUMEN

The Hippo pathway regulates contact inhibition of cell proliferation and, ultimately, organ size in diverse multicellular organisms. Inactivation of the Hippo pathway promotes nuclear localization of the transcriptional coactivator Yap1, a Hippo pathway effector, and can cause cancer. Here, we show that deletion of αE (α epithelial) catenin in the hair follicle stem cell compartment resulted in the development of skin squamous cell carcinoma in mice. Tumor formation was accelerated by simultaneous deletion of αE-catenin and the tumor suppressor-encoding gene p53. A small interfering RNA screen revealed a functional connection between αE-catenin and Yap1. By interacting with Yap1, αE-catenin promoted its cytoplasmic localization, and Yap1 showed constitutive nuclear localization in αE-catenin-null cells. We also found an inverse correlation between αE-catenin abundance and Yap1 activation in human squamous cell carcinoma tumors. These findings identify αE-catenin as a tumor suppressor that inhibits Yap1 activity and sequesters it in the cytoplasm.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinoma de Células Escamosas/metabolismo , Núcleo Celular/metabolismo , Fosfoproteínas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , alfa Catenina/metabolismo , Transporte Activo de Núcleo Celular/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Proteínas de Ciclo Celular , Línea Celular Tumoral , Núcleo Celular/genética , Proliferación Celular , Células HEK293 , Humanos , Ratones , Ratones Desnudos , Ratones Transgénicos , Fosfoproteínas/genética , Factores de Transcripción , Proteína p53 Supresora de Tumor/genética , Proteínas Señalizadoras YAP , alfa Catenina/genética
19.
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
20.
Genes Cancer ; 1(5): 465-479, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20927200

RESUMEN

Deregulated c-Myc is associated with a wide range of human cancers. In many cell types, overexpression of c-Myc potently promotes cell growth and proliferation concomitant with the induction of apoptosis. Secondary genetic events that shift this balance either by increasing growth and proliferation or limiting apoptosis are likely to cooperate with c-Myc in tumorigenesis. Here, the authors have performed large-scale insertional mutagenesis in Eµ-c-myc mice that, through mdm2 loss of function mutations, are sensitized to apoptosis. The authors chose to use this genetic background based on the hypothesis that the high level of apoptosis induced by c-Myc overexpression in MDM2-deficient mice would act as a rate-limiting barrier for lymphoma development. As a result, it was predicted that the spectrum of retroviral insertions would be shifted toward loci that harbor antiapoptotic genes. Nine novel common insertion sites (CISs) specific to mice with this sensitized genetic background were identified, suggesting the presence of novel antiapoptotic cancer genes. Moreover, cross-comparing the data to the Retroviral Tagged Cancer Gene Database, the authors identified an additional 23 novel CISs. Here, evidence is presented that 2 genes, ppp1r16b and hdac6, identified at CISs, are bona fide cellular oncogenes. This study highlights the power of combining unique sensitized genetic backgrounds with large-scale mutagenesis as an approach for identifying novel cancer genes.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...