Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
Cell Rep ; 42(5): 112511, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37195865

RESUMEN

Several methods for generating human-skin-equivalent (HSE) organoid cultures are in use to study skin biology; however, few studies thoroughly characterize these systems. To fill this gap, we use single-cell transcriptomics to compare in vitro HSEs, xenograft HSEs, and in vivo epidermis. By combining differential gene expression, pseudotime analyses, and spatial localization, we reconstruct HSE keratinocyte differentiation trajectories that recapitulate known in vivo epidermal differentiation pathways and show that HSEs contain major in vivo cellular states. However, HSEs also develop unique keratinocyte states, an expanded basal stem cell program, and disrupted terminal differentiation. Cell-cell communication modeling shows aberrant epithelial-to-mesenchymal transition (EMT)-associated signaling pathways that alter upon epidermal growth factor (EGF) supplementation. Last, xenograft HSEs at early time points post transplantation significantly rescue many in vitro deficits while undergoing a hypoxic response that drives an alternative differentiation lineage. This study highlights the strengths and limitations of organoid cultures and identifies areas for potential innovation.


Asunto(s)
Piel , Transcriptoma , Humanos , Transcriptoma/genética , Piel/metabolismo , Queratinocitos/metabolismo , Epidermis/metabolismo , Diferenciación Celular , Organoides
3.
Stem Cells ; 40(4): 435-445, 2022 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-35325240

RESUMEN

Proper differentiation of the epidermis is essential to prevent water loss and to protect the body from the outside environment. Perturbations in this process can lead to a variety of skin diseases that impacts 1 in 5 people. While transcription factors that control epidermal differentiation have been well characterized, other aspects of transcription control such as elongation are poorly understood. Here we show that of the two cyclin-dependent kinases (CDK12 and CDK13), that are known to regulate transcription elongation, only CDK12 is necessary for epidermal differentiation. Depletion of CDK12 led to loss of differentiation gene expression and absence of skin barrier formation in regenerated human epidermis. CDK12 binds to genes that code for differentiation promoting transcription factors (GRHL3, KLF4, and OVOL1) and is necessary for their elongation. CDK12 is necessary for elongation by promoting Ser2 phosphorylation on the C-terminal domain of RNA polymerase II and the stabilization of binding of the elongation factor SPT6 to target genes. Our results suggest that control of transcription elongation by CDK12 plays a prominent role in adult cell fate decisions.


Asunto(s)
Quinasas Ciclina-Dependientes , ARN Polimerasa II , Diferenciación Celular/genética , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Humanos , Fosforilación , ARN Polimerasa II/química , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
PLoS Biol ; 19(9): e3001378, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34543262

RESUMEN

Stratified epithelia such as the epidermis require coordinated regulation of stem and progenitor cell proliferation, survival, and differentiation to maintain homeostasis. Integrin-mediated anchorage of the basal layer stem cells of the epidermis to the underlying dermis through extracellular matrix (ECM) proteins is crucial for this process. It is currently unknown how the expression of these integrins and ECM genes are regulated. Here, we show that the RNA-binding protein (RBP) heterogeneous nuclear ribonucleoprotein L (HNRNPL) binds to these genes on chromatin to promote their expression. HNRNPL recruits RNA polymerase II (Pol II) to integrin/ECM genes and is required for stabilizing Pol II transcription through those genes. In the absence of HNRNPL, the basal layer of the epidermis where the stem cells reside prematurely differentiates and detaches from the underlying dermis due to diminished integrin/ECM expression. Our results demonstrate a critical role for RBPs on chromatin to maintain stem and progenitor cell fate by dictating the expression of specific classes of genes.


Asunto(s)
Células Epidérmicas/metabolismo , Matriz Extracelular/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo L/metabolismo , Integrinas/metabolismo , Diferenciación Celular , Células Cultivadas , Cromatina , Epidermis/crecimiento & desarrollo , Matriz Extracelular/genética , Humanos , Integrinas/genética , Células Madre
6.
Nat Commun ; 12(1): 784, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33542242

RESUMEN

In adult tissue, stem and progenitor cells must tightly regulate the balance between proliferation and differentiation to sustain homeostasis. How this exquisite balance is achieved is an area of active investigation. Here, we show that epidermal genes, including ~30% of induced differentiation genes already contain stalled Pol II at the promoters in epidermal stem and progenitor cells which is then released into productive transcription elongation upon differentiation. Central to this process are SPT6 and PAF1 which are necessary for the elongation of these differentiation genes. Upon SPT6 or PAF1 depletion there is a loss of human skin differentiation and stratification. Unexpectedly, loss of SPT6 also causes the spontaneous transdifferentiation of epidermal cells into an intestinal-like phenotype due to the stalled transcription of the master regulator of epidermal fate P63. Our findings suggest that control of transcription elongation through SPT6 plays a prominent role in adult somatic tissue differentiation and the inhibition of alternative cell fate choices.


Asunto(s)
Diferenciación Celular/genética , Epidermis/fisiología , Elongación de la Transcripción Genética , Factores de Transcripción/metabolismo , Células Madre Adultas/fisiología , Transdiferenciación Celular/genética , Células Cultivadas , Secuenciación de Inmunoprecipitación de Cromatina , Técnicas de Silenciamiento del Gen , Humanos , Recién Nacido , Queratinocitos , Masculino , Cultivo Primario de Células , Regiones Promotoras Genéticas , ARN Polimerasa II/metabolismo , ARN Interferente Pequeño/metabolismo , RNA-Seq , Técnicas de Cultivo de Tejidos , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/metabolismo
8.
J Cell Sci ; 133(23)2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-33172988

RESUMEN

Proper epithelial development and homeostasis depends on strict control of oriented cell division. Current evidence shows that this process is regulated by intrinsic polarity factors and external spatial cues. Owing to the lack of an appropriate model system that can recapitulate the architecture of the skin, deregulation of spindle orientation in human epithelial carcinoma has never been investigated. Here, using an inducible model of human squamous cell carcinoma (SCC), we demonstrate that RAS-dependent suppression of PAR3 (encoded by PARD3) accelerates epithelial disorganization during early tumorigenesis. Diminished PAR3 led to loss of E-cadherin-mediated cell adhesion, which in turn contributed to misoriented cell division. Pharmacological inhibition of the MAPK pathway downstream of RAS activation reversed the defects in PAR3 expression, E-cadherin-mediated cell adhesion and mitotic spindle orientation. Thus, temporal analysis of human neoplasia provides a powerful approach to study cellular and molecular transformations during early oncogenesis, which allowed identification of PAR3 as a critical regulator of tissue architecture during initial human SCC development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Carcinoma de Células Escamosas , Proteínas de Ciclo Celular , Proteínas ras , Carcinogénesis/genética , Carcinoma de Células Escamosas/genética , Adhesión Celular , Proteínas de Ciclo Celular/metabolismo , División Celular , Polaridad Celular , Humanos , Hiperplasia , Huso Acromático/metabolismo
9.
iScience ; 23(7): 101320, 2020 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-32659720

RESUMEN

Impairments in the differentiation process can lead to skin diseases that can afflict ∼20% of the population. Thus, it is of utmost importance to understand the factors that promote the differentiation process. Here we identify the transcription factor KLF3 as a regulator of epidermal differentiation. Knockdown of KLF3 results in reduced differentiation gene expression and increased cell cycle gene expression. Over half of KLF3's genomic binding sites occur at active enhancers. KLF3 binds to active enhancers proximal to differentiation genes that are dependent upon KLF3 for expression. KLF3's genomic binding sites also highly overlaps with CBP, a histone acetyltransferase necessary for activating enhancers. Depletion of KLF3 causes reduced CBP localization at enhancers proximal to differentiation gene clusters, which leads to loss of enhancer activation but not priming. Our results suggest that KLF3 is necessary to recruit CBP to activate enhancers and drive epidermal differentiation gene expression.

12.
Nat Commun ; 10(1): 4198, 2019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-31519929

RESUMEN

Maintenance of high-turnover tissues such as the epidermis requires a balance between stem cell proliferation and differentiation. The molecular mechanisms governing this process are an area of investigation. Here we show that HNRNPK, a multifunctional protein, is necessary to prevent premature differentiation and sustains the proliferative capacity of epidermal stem and progenitor cells. To prevent premature differentiation of progenitor cells, HNRNPK is necessary for DDX6 to bind a subset of mRNAs that code for transcription factors that promote differentiation. Upon binding, these mRNAs such as GRHL3, KLF4, and ZNF750 are degraded through the mRNA degradation pathway, which prevents premature differentiation. To sustain the proliferative capacity of the epidermis, HNRNPK is necessary for RNA Polymerase II binding to proliferation/self-renewal genes such as MYC, CYR61, FGFBP1, EGFR, and cyclins to promote their expression. Our study establishes a prominent role for HNRNPK in maintaining adult tissue self-renewal through both transcriptional and post-transcriptional mechanisms.


Asunto(s)
Diferenciación Celular/fisiología , Ribonucleoproteína Heterogénea-Nuclear Grupo K/metabolismo , ARN Mensajero/metabolismo , Diferenciación Celular/genética , Proliferación Celular/genética , Proliferación Celular/fisiología , Células Cultivadas , Proteína 61 Rica en Cisteína/genética , Proteína 61 Rica en Cisteína/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Epidermis/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo K/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Estabilidad del ARN/genética , Estabilidad del ARN/fisiología , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor
13.
ACS Nano ; 13(6): 6605-6617, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31188564

RESUMEN

Stem cell therapy in heart disease is challenged by mis-injection, poor survival, and low cell retention. Here, we describe a biocompatible multifunctional silica-iron oxide nanoparticle to help solve these issues. The nanoparticles were made via an in situ growth of Fe3O4 nanoparticles on both the external surfaces and pore walls of mesocellular foam silica nanoparticles. In contrast to previous work, this approach builds a magnetic moiety inside the pores of a porous silica structure. These materials serve three roles: drug delivery, magnetic manipulation, and imaging. The addition of Fe3O4 to the silica nanoparticles increased their colloidal stability, T2-based magnetic resonance imaging contrast, and superparamagnetism. We then used the hybrid materials as a sustained release vehicle of insulin-like growth factor-a pro-survival agent that can increase cell viability. In vivo rodent studies show that labeling stem cells with this nanoparticle increased the efficacy of stem cell therapy in a ligation/reperfusion model. The nanoparticle-labeled cells increase the mean left ventricular ejection fraction by 11 and 21% and the global longitudinal strain by 24 and 34% on days 30 and 60, respectively. In summary, this multifunctional nanomedicine improves stem cell survival via the sustained release of pro-survival agents.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Trasplante de Células Madre Mesenquimatosas/métodos , Nanopartículas/química , Nanomedicina Teranóstica/métodos , Animales , Células Cultivadas , Medios de Contraste/química , Liberación de Fármacos , Compuestos Férricos/química , Humanos , Factor I del Crecimiento Similar a la Insulina/farmacología , Imagen por Resonancia Magnética/métodos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/terapia , Dióxido de Silicio/química
14.
Biomaterials ; 179: 60-70, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29980075

RESUMEN

Silicon carbide has been shown to be biocompatible and is used as a coating material for implanted medical devices to prevent biofilms. Silicon carbide nanomaterials are also promising in cell tracking due to their stable and strong luminescence, but more comprehensive studies of this material on the nanoscale are needed. Here, we studied the toxicity of silicon carbide nanomaterials on human mesenchymal stem cells in terms of metabolism, viability, adhesion, proliferation, migration, oxidative stress, and differentiation ability. We compared two different shapes and found that silicon carbide nanowires are toxic to human mesenchymal stem cells but not to cancer cell lines at the concentration of 0.1 mg/mL. Control silicon carbide nanoparticles were biocompatible to human mesenchymal stem cells at 0.1 mg/mL. We studied the potential mechanistic effect of silicon carbide nanowires on human mesenchymal stem cells' phenotype, cytokine secretion, and gene expression. These findings suggest that the toxic effect of silicon carbide nanomaterials to human mesenchymal stem cells are dependent on morphology.


Asunto(s)
Compuestos Inorgánicos de Carbono/química , Nanoestructuras/química , Nanocables/química , Compuestos de Silicona/química , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Citocinas/química , Humanos , Células Madre Mesenquimatosas/citología , Estrés Oxidativo/fisiología
15.
Cell Rep ; 20(13): 3005-3013, 2017 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-28954219

RESUMEN

Adult stem and progenitor cells are critical for replenishing lost tissue due to injury or normal turnover. How these cells maintain self-renewal and sustain the tissue they populate are areas of active investigation. Here, we show that the cohesin complex, which has previously been implicated in regulating chromosome segregation and gene expression, is necessary to promote epidermal stem and progenitor cell self-renewal through cell-autonomous mechanisms. Cohesin binds to genomic sites associated with open chromatin, including DNase-I-hypersensitive sites, RNA polymerase II, and histone marks such as H3K27ac and H3K4me3. Reduced cohesin expression results in spontaneous epidermal differentiation due to loss of open chromatin structure and expression of key self-renewal genes. Our results demonstrate a prominent role for cohesin in modulating chromatin structure to allow for enforcement of a stem and progenitor cell gene expression program.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/genética , Epidermis/metabolismo , Genes/genética , Células Madre/metabolismo , Diferenciación Celular , Células Epidérmicas , Humanos , Células Madre/citología , Cohesinas
16.
Exp Dermatol ; 26(9): 792-797, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28094886

RESUMEN

Darier disease (DD) is a genetic skin disease that is associated with mutations in the ATP2A2 gene encoding the type 2 sarco/endoplasmic reticulum (ER) Ca2+ - ATPase (SERCA2). Mutations of this gene result in alterations of calcium homoeostasis, abnormal epidermal adhesion and dyskeratosis. Silencing of ATP2A2 in monolayer cell culture of keratinocytes reduces desmoplakin expression at the borders of cells and impacts cell adhesion. Here, we report establishment of a three-dimensional (3D) epidermal model of DD and use this model to evaluate peptide therapy with tuberoinfundibular peptide of 39 residues (TIP39) to normalize calcium transport. Gene silencing of ATP2A2 in keratinocytes grown in a 3D model resulted in dyskeratosis, partial parakeratosis and suprabasal clefts that resembled the histological changes seen in skin biopsies from patients with DD. TIP39, a peptide recently identified as a regulator of keratinocyte calcium transport, was then applied to this ATP2A2-silenced 3D epidermal model. In normal keratinocytes, TIP39 increased [Ca2+ ]i through the inositol trisphosphate (IP3) receptor pathway and stimulated differentiation. In monolayer ATP2A2-silenced keratinocytes, although TIP39 increased cytosolic calcium from the ER, the response was incomplete compared with its control. TIP39 was observed to reduce intercellular clefts of the gene-silenced epidermal model but did not significantly upregulate keratinocyte differentiation genes such as keratin 10 and filaggrin. These findings indicate that TIP39 is a modulator of ER calcium signalling and may be used as a potential strategy for improving aspects of DD.


Asunto(s)
Calcio/metabolismo , Enfermedad de Darier/metabolismo , Neuropéptidos/metabolismo , Receptor de Hormona Paratiroídea Tipo 2/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Células Cultivadas , Retículo Endoplásmico/metabolismo , Epidermis/metabolismo , Proteínas Filagrina , Humanos , Queratinocitos/metabolismo
17.
Cell Stem Cell ; 19(4): 415-417, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27716517

RESUMEN

In this issue of Cell Stem Cell, Rinaldi et al. (2016) find an unexpected role for the de novo DNA methyltransferases Dnmt3a and Dnmt3b in the regulation of enhancers. Their findings provide new insight into how regulation of enhancer activity through DNA methylation can have dramatic consequences on epidermal stem cell fate decisions.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Células Madre , Metilación de ADN
18.
Immunity ; 45(1): 119-30, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27438769

RESUMEN

Type 1 interferons (IFNs) promote inflammation in the skin but the mechanisms responsible for inducing these cytokines are not well understood. We found that IFN-ß was abundantly produced by epidermal keratinocytes (KCs) in psoriasis and during wound repair. KC IFN-ß production depended on stimulation of mitochondrial antiviral-signaling protein (MAVS) by the antimicrobial peptide LL37 and double stranded-RNA released from necrotic cells. MAVS activated downstream TBK1 (TANK-Binding Kinase 1)-AKT (AKT serine/threonine kinase 1)-IRF3 (interferon regulatory factor 3) signaling cascade leading to IFN-ß production and then promoted maturation of dendritic cells. In mice, the production of epidermal IFN-ß by LL37 required MAVS, and human wounded and/or psoriatic skin showed activation of MAVS-associated IRF3 and induction of MAVS and IFN-ß gene signatures. These findings show that KCs are an important source of IFN-ß and MAVS is critical to this function, and demonstrates how the epidermis triggers unwanted skin inflammation under disease conditions.


Asunto(s)
Catelicidinas/metabolismo , Células Dendríticas/fisiología , Epidermis/patología , Queratinocitos/inmunología , Mitocondrias/metabolismo , Psoriasis/inmunología , Heridas y Lesiones/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos , Catelicidinas/genética , Diferenciación Celular , Células Cultivadas , Humanos , Interferón beta/metabolismo , Ratones , Ratones Noqueados , ARN Interferente Pequeño/genética , Transducción de Señal , Cicatrización de Heridas
19.
J Invest Dermatol ; 136(4): 746-752, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26875726

RESUMEN

Epidermal stem and progenitor cells exist within the basal layer of the epidermis and serve to replenish the loss of differentiated cells because of normal turnover or injury. Current efforts have focused on elucidating the transcriptional regulation of epidermal stem cell self-renewal and differentiation. However, recent studies have pointed to an emerging and prominent role for post-transcriptional regulation of epidermal cell fate decisions. In this review, we will focus on post-transcriptional mechanisms including noncoding RNAs, RNA binding proteins, and mRNA decay-mediated control of epidermal stem and progenitor cell function in the skin.


Asunto(s)
Autorrenovación de las Células , Células Epidérmicas , Procesamiento Postranscripcional del ARN , Células Madre/citología , Regiones no Traducidas 3' , Animales , Diferenciación Celular , Proliferación Celular , ARN Helicasas DEAD-box/metabolismo , Exosomas/metabolismo , Regulación de la Expresión Génica , Humanos , Ratones , MicroARNs/genética , Mutación , Proteínas Proto-Oncogénicas/metabolismo , ARN Largo no Codificante/genética
20.
J Vis Exp ; (106): e53280, 2015 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-26709715

RESUMEN

Organotypic cultures allow the reconstitution of a 3D environment critical for cell-cell contact and cell-matrix interactions which mimics the function and physiology of their in vivo tissue counterparts. This is exemplified by organotypic skin cultures which faithfully recapitulates the epidermal differentiation and stratification program. Primary human epidermal keratinocytes are genetically manipulable through retroviruses where genes can be easily overexpressed or knocked down. These genetically modified keratinocytes can then be used to regenerate human epidermis in organotypic skin cultures providing a powerful model to study genetic pathways impacting epidermal growth, differentiation, and disease progression. The protocols presented here describe methods to prepare devitalized human dermis as well as to genetically manipulate primary human keratinocytes in order to generate organotypic skin cultures. Regenerated human skin can be used in downstream applications such as gene expression profiling, immunostaining, and chromatin immunoprecipitations followed by high throughput sequencing. Thus, generation of these genetically modified organotypic skin cultures will allow the determination of genes that are critical for maintaining skin homeostasis.


Asunto(s)
Técnicas de Cultivo de Órganos/métodos , Piel/citología , Piel/crecimiento & desarrollo , Comunicación Celular/fisiología , Diferenciación Celular/fisiología , Dermis/citología , Epidermis/metabolismo , Humanos , Queratinocitos/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...