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










Base de datos
Intervalo de año de publicación
1.
Gigascience ; 9(5)2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32401319

RESUMEN

BACKGROUND: Infinium Human Methylation BeadChip is an array platform for complex evaluation of DNA methylation at an individual CpG locus in the human genome based on Illumina's bead technology and is one of the most common techniques used in epigenome-wide association studies. Finding associations between epigenetic variation and phenotype is a significant challenge in biomedical research. The newest version, HumanMethylationEPIC, quantifies the DNA methylation level of 850,000 CpG sites, while the previous versions, HumanMethylation450 and HumanMethylation27, measured >450,000 and 27,000 loci, respectively. Although a number of bioinformatics tools have been developed to analyse this assay, they require some programming skills and experience in order to be usable. RESULTS: We have developed a pipeline for the Galaxy platform for those without experience aimed at DNA methylation analysis using the Infinium Human Methylation BeadChip. Our tool is integrated into Galaxy (http://galaxyproject.org), a web-based platform. This allows users to analyse data from the Infinium Human Methylation BeadChip in the easiest possible way. CONCLUSIONS: The pipeline provides a group of integrated analytical methods wrapped into an easy-to-use interface. Our tool is available from the Galaxy ToolShed, GitHub repository, and also as a Docker image. The aim of this project is to make Infinium Human Methylation BeadChip analysis more flexible and accessible to everyone.


Asunto(s)
Biología Computacional/métodos , Epigénesis Genética , Epigenómica/métodos , Programas Informáticos , Biología Computacional/normas , Metilación de ADN , Epigenómica/normas , Genética de Población/métodos , Genoma Humano , Estudio de Asociación del Genoma Completo , Humanos , Anotación de Secuencia Molecular , Interfaz Usuario-Computador
2.
F1000Res ; 92020.
Artículo en Inglés | MEDLINE | ID: mdl-34367618

RESUMEN

Copy number variations (CNVs) are major causative contributors both in the genesis of genetic diseases and human neoplasias. While "High-Throughput" sequencing technologies are increasingly becoming the primary choice for genomic screening analysis, their ability to efficiently detect CNVs is still heterogeneous and remains to be developed. The aim of this white paper is to provide a guiding framework for the future contributions of ELIXIR's recently established human CNV Community, with implications beyond human disease diagnostics and population genomics. This white paper is the direct result of a strategy meeting that took place in September 2018 in Hinxton (UK) and involved representatives of 11 ELIXIR Nodes. The meeting led to the definition of priority objectives and tasks, to address a wide range of CNV-related challenges ranging from detection and interpretation to sharing and training. Here, we provide suggestions on how to align these tasks within the ELIXIR Platforms strategy, and on how to frame the activities of this new ELIXIR Community in the international context.


Asunto(s)
Biología Computacional , Variaciones en el Número de Copia de ADN , Variaciones en el Número de Copia de ADN/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos
4.
PLoS Pathog ; 14(9): e1007276, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30188954

RESUMEN

Merkel cell carcinoma (MCC) is an aggressive skin cancer with a high propensity for recurrence and metastasis. Merkel cell polyomavirus (MCPyV) is recognised as the causative factor in the majority of MCC cases. The MCPyV small tumour antigen (ST) is considered to be the main viral transforming factor, however potential mechanisms linking ST expression to the highly metastatic nature of MCC are yet to be fully elucidated. Metastasis is a complex process, with several discrete steps required for the formation of secondary tumour sites. One essential trait that underpins the ability of cancer cells to metastasise is how they interact with adjoining tumour cells and the surrounding extracellular matrix. Here we demonstrate that MCPyV ST expression disrupts the integrity of cell-cell junctions, thereby enhancing cell dissociation and implicate the cellular sheddases, A disintegrin and metalloproteinase (ADAM) 10 and 17 proteins in this process. Inhibition of ADAM 10 and 17 activity reduced MCPyV ST-induced cell dissociation and motility, attributing their function as critical to the MCPyV-induced metastatic processes. Consistent with these data, we confirm that ADAM 10 and 17 are upregulated in MCPyV-positive primary MCC tumours. These novel findings implicate cellular sheddases as key host cell factors contributing to virus-mediated cellular transformation and metastasis. Notably, ADAM protein expression may be a novel biomarker of MCC prognosis and given the current interest in cellular sheddase inhibitors for cancer therapeutics, it highlights ADAM 10 and 17 activity as a novel opportunity for targeted interventions for disseminated MCC.


Asunto(s)
Antígenos Virales de Tumores/fisiología , Carcinoma de Células de Merkel/etiología , Poliomavirus de Células de Merkel/patogenicidad , Infecciones por Polyomavirus/etiología , Neoplasias Cutáneas/etiología , Infecciones Tumorales por Virus/etiología , Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Carcinoma de Células de Merkel/enzimología , Carcinoma de Células de Merkel/secundario , Movimiento Celular , Células HEK293 , Humanos , Uniones Intercelulares/patología , Uniones Intercelulares/fisiología , Proteínas de la Membrana/metabolismo , Poliomavirus de Células de Merkel/inmunología , Poliomavirus de Células de Merkel/fisiología , Invasividad Neoplásica/patología , Invasividad Neoplásica/fisiopatología , Infecciones por Polyomavirus/enzimología , Infecciones por Polyomavirus/patología , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/patología , Infecciones Tumorales por Virus/enzimología , Infecciones Tumorales por Virus/patología
5.
J Invest Dermatol ; 138(11): 2343-2354, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29777657

RESUMEN

Virus-encoded microRNAs are emerging as key regulators of persistent infection and host-cell immune evasion. Merkel cell polyomavirus, the predominant etiological agent of Merkel cell carcinoma, encodes a single microRNA, MCV-miR-M1, which targets the oncogenic Merkel cell polyomavirus large T antigen. MCV-miR-M1 has previously been shown to play an important role in the establishment of long-term infection, however, the underlying mechanism is not fully understood. A key unanswered question is whether, in addition to autoregulating large T antigen, MCV-miR-M1 also targets cellular transcripts to orchestrate an environment conducive to persistent infection. To address this, we adopted an RNA sequencing-based approach to identify cellular targets of MCV-miR-M1. Intriguingly, bioinformatics analysis of transcripts that are differentially expressed in cells expressing MCV-miR-M1 revealed several genes implicated in immune evasion. Subsequent target validation led to the identification of the innate immunity protein, SP100, as a direct target of MCV-miR-M1. Moreover, MCV-miR-M1-mediated modulation of SP100 was associated with a significant decrease in CXCL8 secretion, resulting in the attenuation of neutrophil chemotaxis toward Merkel cells harboring synthetic Merkel cell polyomavirus. Based on these observations, we propose that MCV-miR-M1 targets key immune response regulators to help facilitate persistent infection, which is a prerequisite for cellular transformation in Merkel cell carcinoma.


Asunto(s)
Carcinoma de Células de Merkel/inmunología , Poliomavirus de Células de Merkel/fisiología , MicroARNs/genética , Neutrófilos/inmunología , Infecciones por Polyomavirus/inmunología , ARN Viral/genética , Infecciones Tumorales por Virus/inmunología , Antígenos Nucleares/genética , Antígenos Virales de Tumores/genética , Autoantígenos/genética , Carcinoma de Células de Merkel/genética , Quimiotaxis , Células HEK293 , Humanos , Evasión Inmune , Inmunidad Innata/genética , Interleucina-8/metabolismo , Infecciones por Polyomavirus/genética , Infecciones Tumorales por Virus/genética
6.
J Biol Chem ; 293(12): 4582-4590, 2018 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-29462791

RESUMEN

Ion channels regulate many aspects of cell physiology, including cell proliferation, motility, and migration, and aberrant expression and activity of ion channels is associated with various stages of tumor development, with K+ and Cl- channels now being considered the most active during tumorigenesis. Accordingly, emerging in vitro and preclinical studies have revealed that pharmacological manipulation of ion channel activity offers protection against several cancers. Merkel cell polyomavirus (MCPyV) is a major cause of Merkel cell carcinoma (MCC), primarily because of the expression of two early regulatory proteins termed small and large tumor antigens (ST and LT, respectively). Several molecular mechanisms have been attributed to MCPyV-mediated cancer formation but, thus far, no studies have investigated any potential link to cellular ion channels. Here we demonstrate that Cl- channel modulation can reduce MCPyV ST-induced cell motility and invasiveness. Proteomic analysis revealed that MCPyV ST up-regulates two Cl- channels, CLIC1 and CLIC4, which when silenced, inhibit MCPyV ST-induced motility and invasiveness, implicating their function as critical to MCPyV-induced metastatic processes. Consistent with these data, we confirmed that CLIC1 and CLIC4 are up-regulated in primary MCPyV-positive MCC patient samples. We therefore, for the first time, implicate cellular ion channels as a key host cell factor contributing to virus-mediated cellular transformation. Given the intense interest in ion channel modulating drugs for human disease. This highlights CLIC1 and CLIC4 activity as potential targets for MCPyV-induced MCC.


Asunto(s)
Carcinoma de Células de Merkel/patología , Movimiento Celular , Canales de Cloruro/metabolismo , Poliomavirus de Células de Merkel/fisiología , Infecciones por Polyomavirus/complicaciones , Neoplasias Cutáneas/secundario , Infecciones Tumorales por Virus/complicaciones , Antígenos Virales de Tumores/genética , Antígenos Virales de Tumores/metabolismo , Carcinoma de Células de Merkel/epidemiología , Carcinoma de Células de Merkel/virología , Proliferación Celular , Canales de Cloruro/genética , Cloruros/metabolismo , Células HEK293 , Humanos , Incidencia , Invasividad Neoplásica , Infecciones por Polyomavirus/patología , Infecciones por Polyomavirus/virología , Proteoma/análisis , Neoplasias Cutáneas/epidemiología , Neoplasias Cutáneas/virología , Infecciones Tumorales por Virus/patología , Infecciones Tumorales por Virus/virología
7.
Oncotarget ; 8(52): 89566-89579, 2017 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-29163771

RESUMEN

The HOX genes encode a family of transcription factors that have key roles in both development and malignancy. Disrupting the interaction between HOX proteins and their binding partner, PBX, has been shown to cause apoptotic cell death in a range of solid tumors. However, despite HOX proteins playing a particularly significant role in acute myeloid leukemia (AML), the relationship between HOX gene expression and patient survival has not been evaluated (with the exception of HOXA9), and the mechanism by which HOX/PBX inhibition induces cell death in this malignancy is not well understood. In this study, we show that the expression of HOXA5, HOXB2, HOXB4, HOXB9, and HOXC9, but not HOXA9, in primary AML samples is significantly related to survival. Furthermore, the previously described inhibitor of HOX/PBX dimerization, HXR9, is cytotoxic to both AML-derived cell lines and primary AML cells from patients. The mechanism of cell death is not dependent on apoptosis but instead involves a regulated form of necrosis referred to as necroptosis. HXR9-induced necroptosis is enhanced by inhibitors of protein kinase C (PKC) signaling, and HXR9 combined with the PKC inhibitor Ro31 causes a significantly greater reduction in tumor growth compared to either reagent alone.

8.
PLoS Genet ; 13(9): e1006966, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28863138

RESUMEN

Mammalian genomes contain several dozens of large (>0.5 Mbp) lineage-specific gene loci harbouring functionally related genes. However, spatial chromatin folding, organization of the enhancer-promoter networks and their relevance to Topologically Associating Domains (TADs) in these loci remain poorly understood. TADs are principle units of the genome folding and represents the DNA regions within which DNA interacts more frequently and less frequently across the TAD boundary. Here, we used Chromatin Conformation Capture Carbon Copy (5C) technology to characterize spatial chromatin interaction network in the 3.1 Mb Epidermal Differentiation Complex (EDC) locus harbouring 61 functionally related genes that show lineage-specific activation during terminal keratinocyte differentiation in the epidermis. 5C data validated by 3D-FISH demonstrate that the EDC locus is organized into several TADs showing distinct lineage-specific chromatin interaction networks based on their transcription activity and the gene-rich or gene-poor status. Correlation of the 5C results with genome-wide studies for enhancer-specific histone modifications (H3K4me1 and H3K27ac) revealed that the majority of spatial chromatin interactions that involves the gene-rich TADs at the EDC locus in keratinocytes include both intra- and inter-TAD interaction networks, connecting gene promoters and enhancers. Compared to thymocytes in which the EDC locus is mostly transcriptionally inactive, these interactions were found to be keratinocyte-specific. In keratinocytes, the promoter-enhancer anchoring regions in the gene-rich transcriptionally active TADs are enriched for the binding of chromatin architectural proteins CTCF, Rad21 and chromatin remodeler Brg1. In contrast to gene-rich TADs, gene-poor TADs show preferential spatial contacts with each other, do not contain active enhancers and show decreased binding of CTCF, Rad21 and Brg1 in keratinocytes. Thus, spatial interactions between gene promoters and enhancers at the multi-TAD EDC locus in skin epithelial cells are cell type-specific and involve extensive contacts within TADs as well as between different gene-rich TADs, forming the framework for lineage-specific transcription.


Asunto(s)
Diferenciación Celular/genética , Cromatina/genética , ADN Helicasas/genética , Proteínas Nucleares/genética , Fosfoproteínas/genética , Proteínas Represoras/genética , Factores de Transcripción/genética , Animales , Factor de Unión a CCCTC , Proteínas de Ciclo Celular , Ensamble y Desensamble de Cromatina/genética , Proteínas de Unión al ADN/genética , Elementos de Facilitación Genéticos , Epidermis/metabolismo , Epigénesis Genética , Genoma , Queratinocitos , Ratones , Regiones Promotoras Genéticas , Piel/metabolismo
9.
J Invest Dermatol ; 137(10): 2157-2167, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28595999

RESUMEN

The maintenance of a proper nuclear architecture and three-dimensional organization of the genes, enhancer elements, and transcription machinery plays an essential role in tissue development and regeneration. Here we show that in the developing skin, epidermal progenitor cells of mice lacking p63 transcription factor display alterations in the nuclear shape accompanied by a marked decrease in expression of several nuclear envelope-associated components (Lamin B1, Lamin A/C, Sun1, Nesprin-3, Plectin) compared with controls. Furthermore, chromatin immunoprecipitation-quantitative PCR assay showed enrichment of p63 on Sun1, Syne3, and Plec promoters, suggesting them as p63 targets. Alterations in the nuclei shape and expression of nuclear envelope-associated proteins were accompanied by altered distribution patterns of the repressive histone marks trimethylation on lysine 27 of histone H3, trimethylation on lysine 9 of histone H3, and heterochromatin protein 1-alpha in p63-null keratinocytes. These changes were also accompanied by downregulation of the transcriptional activity and relocation of the keratinocyte-specific gene loci away from the sites of active transcription toward the heterochromatin-enriched repressive nuclear compartments in p63-null cells. These data demonstrate functional links between the nuclear envelope organization, chromatin architecture, and gene expression in keratinocytes and suggest nuclear envelope-associated genes as important targets mediating p63-regulated gene expression program in the epidermis.


Asunto(s)
Epidermis/metabolismo , Regulación del Desarrollo de la Expresión Génica , Queratinocitos/metabolismo , Fosfoproteínas/genética , Transactivadores/genética , Animales , Diferenciación Celular , Núcleo Celular/metabolismo , Epidermis/patología , Humanos , Queratinocitos/patología , Ratones , Modelos Animales , Membrana Nuclear/genética , Membrana Nuclear/metabolismo , Fosfoproteínas/biosíntesis , ARN/genética , Transactivadores/biosíntesis , Factores de Transcripción/genética , Transcripción Genética
10.
J Cell Biol ; 212(1): 77-89, 2016 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-26711500

RESUMEN

During development, multipotent progenitor cells establish lineage-specific programmers of gene activation and silencing underlying their differentiation into specialized cell types. We show that the Polycomb component Cbx4 serves as a critical determinant that maintains the epithelial identity in the developing epidermis by repressing nonepidermal gene expression programs. Cbx4 ablation in mice results in a marked decrease of the epidermal thickness and keratinocyte (KC) proliferation associated with activation of numerous neuronal genes and genes encoding cyclin-dependent kinase inhibitors (p16/p19 and p57). Furthermore, the chromodomain- and SUMO E3 ligase-dependent Cbx4 activities differentially regulate proliferation, differentiation, and expression of nonepidermal genes in KCs. Finally, Cbx4 expression in KCs is directly regulated by p63 transcription factor, whereas Cbx4 overexpression is capable of partially rescuing the effects of p63 ablation on epidermal development. These data demonstrate that Cbx4 plays a crucial role in the p63-regulated program of epidermal differentiation, maintaining the epithelial identity and proliferative activity in KCs via repression of the selected nonepidermal lineage and cell cycle inhibitor genes.


Asunto(s)
Linaje de la Célula , Células Epiteliales/citología , Células Epiteliales/metabolismo , Epitelio/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Epitelio/crecimiento & desarrollo , Ligasas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejo Represivo Polycomb 1/deficiencia , Complejo Represivo Polycomb 1/genética , Células Madre/citología , Células Madre/metabolismo , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética
11.
J Cell Biol ; 207(4): 549-67, 2014 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-25422376

RESUMEN

Skin development is governed by complex programs of gene activation and silencing, including microRNA-dependent modulation of gene expression. Here, we show that miR-214 regulates skin morphogenesis and hair follicle (HF) cycling by targeting ß-catenin, a key component of the Wnt signaling pathway. miR-214 exhibits differential expression patterns in the skin epithelium, and its inducible overexpression in keratinocytes inhibited proliferation, which resulted in formation of fewer HFs with decreased hair bulb size and thinner hair production. The inhibitory effects of miR-214 on HF development and cycling were associated with altered activities of multiple signaling pathways, including decreased expression of key Wnt signaling mediators ß-catenin and Lef-1, and were rescued by treatment with pharmacological Wnt activators. Finally, we identify ß-catenin as one of the conserved miR-214 targets in keratinocytes. These data provide an important foundation for further analyses of miR-214 as a key regulator of Wnt pathway activity and stem cell functions during normal tissue homeostasis, regeneration, and aging.


Asunto(s)
Folículo Piloso/crecimiento & desarrollo , Factor de Unión 1 al Potenciador Linfoide/genética , MicroARNs/fisiología , Vía de Señalización Wnt , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Senescencia Celular/genética , Genotipo , Folículo Piloso/metabolismo , Queratina-10/biosíntesis , Queratina-14/biosíntesis , Queratinocitos/citología , Factor de Unión 1 al Potenciador Linfoide/biosíntesis , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Transgénicos , MicroARNs/genética , Regeneración/genética , Piel/crecimiento & desarrollo , Piel/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/biosíntesis , beta Catenina/genética
12.
J Invest Dermatol ; 134(12): 2873-2882, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24999588

RESUMEN

Chemotherapy has severe side effects in normal rapidly proliferating organs, such as hair follicles, and causes massive apoptosis in hair matrix keratinocytes followed by hair loss. To define the molecular signature of hair follicle response to chemotherapy, human scalp hair follicles cultured ex vivo were treated with doxorubicin (DXR), and global microarray analysis was performed 3 hours after treatment. Microarray data revealed changes in expression of 504 genes in DXR-treated hair follicles versus controls. Among these genes, upregulations of several tumor necrosis factor family of apoptotic receptors (FAS, TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) receptors 1/2), as well as of a large number of keratin-associated protein genes, were seen after DXR treatment. Hair follicle apoptosis induced by DXR was significantly inhibited by either TRAIL-neutralizing antibody or caspase-8 inhibitor, thus suggesting a previously unreported role for TRAIL receptor signaling in mediating DXR-induced hair loss. These data demonstrate that the early phase of the hair follicle response to DXR includes upregulation of apoptosis-associated markers, as well as substantial reorganization of the terminal differentiation programs in hair follicle keratinocytes. These data provide an important platform for further studies toward the design of effective approaches for the management of chemotherapy-induced hair loss.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Doxorrubicina/farmacología , Folículo Piloso/citología , Alopecia/inducido químicamente , Alopecia/metabolismo , Alopecia/patología , Antineoplásicos/efectos adversos , Caspasa 8/efectos de los fármacos , Caspasa 8/metabolismo , Células Cultivadas , Daño del ADN/efectos de los fármacos , Doxorrubicina/efectos adversos , Folículo Piloso/metabolismo , Folículo Piloso/patología , Humanos , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/efectos de los fármacos , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor fas/efectos de los fármacos , Receptor fas/metabolismo
13.
J Invest Dermatol ; 134(3): 827-837, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24126843

RESUMEN

Bone morphogenetic protein (BMP) signaling plays a key role in the control of skin development and postnatal remodeling by regulating keratinocyte proliferation, differentiation, and apoptosis. To study the role of BMPs in wound-induced epidermal repair, we used transgenic mice overexpressing the BMP downstream component Smad1 under the control of a K14 promoter as an in vivo model, as well as ex vivo and in vitro assays. K14-caSmad1 (transgenic mice overexpressing a constitutively active form of Smad1 under K14 promoter) mice exhibited retarded wound healing associated with significant inhibition of proliferation and increased apoptosis in healing wound epithelium. Furthermore, microarray and quantitative real-time reverse-transcriptase-PCR (qRT-PCR) analyses revealed decreased expression of a number of cytoskeletal/cell motility-associated genes including wound-associated keratins (Krt16, Krt17) and Myosin VA (Myo5a), in the epidermis of K14-caSmad1 mice versus wild-type (WT) controls during wound healing. BMP treatment significantly inhibited keratinocyte migration ex vivo, and primary keratinocytes of K14-caSmad1 mice showed retarded migration compared with WT controls. Finally, small interfering RNA (siRNA)-mediated silencing of BMPR-1B in primary mouse keratinocytes accelerated cell migration and was associated with increased expression of Krt16, Krt17, and Myo5a compared with controls. Thus, this study demonstrates that BMPs inhibit keratinocyte proliferation, cytoskeletal organization, and migration in regenerating skin epithelium during wound healing, and raises a possibility for using BMP antagonists for the management of chronic wounds.


Asunto(s)
Apoptosis/fisiología , Proteínas Morfogenéticas Óseas/metabolismo , Epidermis/fisiología , Queratinocitos/fisiología , Transducción de Señal/fisiología , Cicatrización de Heridas/fisiología , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas Morfogenéticas Óseas/genética , Movimiento Celular/fisiología , Proliferación Celular , Células Cultivadas , Células Epidérmicas , Humanos , Queratina-14/genética , Queratinocitos/citología , Ratones , Ratones Endogámicos , Ratones Transgénicos , ARN Interferente Pequeño/genética , Proteína Smad1/genética , Proteína Smad1/metabolismo
14.
Development ; 141(1): 101-11, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24346698

RESUMEN

Chromatin structural states and their remodelling, including higher-order chromatin folding and three-dimensional (3D) genome organisation, play an important role in the control of gene expression. The role of 3D genome organisation in the control and execution of lineage-specific transcription programmes during the development and differentiation of multipotent stem cells into specialised cell types remains poorly understood. Here, we show that substantial remodelling of the higher-order chromatin structure of the epidermal differentiation complex (EDC), a keratinocyte lineage-specific gene locus on mouse chromosome 3, occurs during epidermal morphogenesis. During epidermal development, the locus relocates away from the nuclear periphery towards the nuclear interior into a compartment enriched in SC35-positive nuclear speckles. Relocation of the EDC locus occurs prior to the full activation of EDC genes involved in controlling terminal keratinocyte differentiation and is a lineage-specific, developmentally regulated event controlled by transcription factor p63, a master regulator of epidermal development. We also show that, in epidermal progenitor cells, p63 directly regulates the expression of the ATP-dependent chromatin remodeller Brg1, which binds to distinct domains within the EDC and is required for relocation of the EDC towards the nuclear interior. Furthermore, Brg1 also regulates gene expression within the EDC locus during epidermal morphogenesis. Thus, p63 and its direct target Brg1 play an essential role in remodelling the higher-order chromatin structure of the EDC and in the specific positioning of this locus within the landscape of the 3D nuclear space, as required for the efficient expression of EDC genes in epidermal progenitor cells during skin development.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , ADN Helicasas/metabolismo , Células Madre Multipotentes/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Cromatina/metabolismo , ADN Helicasas/genética , Células Epidérmicas , Epidermis/embriología , Epidermis/metabolismo , Factor de Transcripción de la Proteína de Unión a GA/genética , Regulación del Desarrollo de la Expresión Génica , Queratinocitos/citología , Queratinocitos/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/genética , Fosfoproteínas/genética , Unión Proteica , Pliegue de Proteína , Interferencia de ARN , ARN Interferente Pequeño , Ribonucleoproteínas/metabolismo , Factores de Empalme Serina-Arginina , Transactivadores/genética , Factores de Transcripción/genética , Transcripción Genética
15.
J Invest Dermatol ; 133(9): 2191-201, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23407401

RESUMEN

The nucleus of epidermal keratinocytes (KCs) is a complex and highly compartmentalized organelle, whose structure is markedly changed during terminal differentiation and transition of the genome from a transcriptionally active state seen in the basal and spinous epidermal cells to a fully inactive state in the keratinized cells of the cornified layer. Here, using multicolor confocal microscopy, followed by computational image analysis and mathematical modeling, we demonstrate that in normal mouse footpad epidermis, transition of KCs from basal epidermal layer to the granular layer is accompanied by marked differences in nuclear architecture and microenvironment including the following: (i) decrease in the nuclear volume; (ii) decrease in expression of the markers of transcriptionally active chromatin; (iii) internalization and decrease in the number of nucleoli; (iv) increase in the number of pericentromeric heterochromatic clusters; and (v) increase in the frequency of associations between the pericentromeric clusters, chromosomal territory 3, and nucleoli. These data suggest a role for nucleoli and pericentromeric heterochromatin clusters as organizers of nuclear microenvironment required for proper execution of gene expression programs in differentiating KCs, and provide important background information for further analyses of alterations in the topological genome organization seen in pathological skin conditions, including disorders of epidermal differentiation and epidermal tumors.


Asunto(s)
Diferenciación Celular/fisiología , Nucléolo Celular/fisiología , Núcleo Celular/fisiología , Células Epidérmicas , Queratinocitos/citología , Modelos Biológicos , Animales , Microambiente Celular/fisiología , Pie , Marcadores Genéticos/fisiología , Heterocromatina/fisiología , Imagenología Tridimensional/métodos , Ratones , Ratones Endogámicos C57BL , Transcripción Genética/fisiología
16.
Semin Cancer Biol ; 23(2): 72-9, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22771615

RESUMEN

When cells change functions or activities (such as during differentiation, response to extracellular stimuli, or migration), gene expression undergoes large-scale reprogramming, in cell type- and function-specific manners. Large changes in gene regulation require changes in chromatin architecture, which involve recruitment of chromatin remodeling enzymes and epigenomic modification enzymes to specific genomic loci. Transcription factors must also be accurately assembled at these loci. SATB1 is a genome organizer protein that facilitates these processes, providing a nuclear architectural platform that anchors hundreds of genes, through its interaction with specific genomic sequences; this activity allows expression of all these genes to be regulated in parallel, and enables cells to thereby alter their function. We review and describe future perspectives on SATB1 function in higher-order chromatin structure and gene regulation, and its role in metastasis of breast cancer and other tumor types.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Proteínas de Unión a la Región de Fijación a la Matriz/fisiología , Neoplasias/genética , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Cromatina/química , Cromatina/metabolismo , Progresión de la Enfermedad , Epigénesis Genética/genética , Femenino , Genoma Humano/fisiología , Humanos , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Modelos Biológicos
17.
FASEB J ; 26(10): 3946-56, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22730438

RESUMEN

The relevance of preconceptional and prenatal toxicant exposures for genomic stability in offspring is difficult to analyze in human populations, because gestational exposures usually cannot be separated from preconceptional exposures. To analyze the roles of exposures during gestation and conception on genomic stability in the offspring, stability was assessed via the Comet assay and highly sensitive, semiautomated confocal laser scans of γH2AX foci in cord, maternal, and paternal blood as well as spermatozoa from 39 families in Crete, Greece, and the United Kingdom. With use of multivariate linear regression analysis with backward selection, preconceptional paternal smoking (% tail DNA: P>0.032; γH2AX foci: P>0.018) and gestational maternal (% tail DNA: P>0.033) smoking were found to statistically significantly predict DNA damage in the cord blood of F1 offspring. Maternal passive smoke exposure was not identified as a predictor of DNA damage in cord blood, indicating that the effect of paternal smoking may be transmitted via the spermatozoal genome. Taken together, these studies reveal a role for cigarette smoke in the induction of DNA alterations in human F1 offspring via exposures of the fetus in utero or the paternal germline. Moreover, the identification of transgenerational DNA alterations in the unexposed F1 offspring of smoking-exposed fathers supports the claim that cigarette smoke is a human germ cell mutagen.


Asunto(s)
Sangre Fetal/metabolismo , Inestabilidad Genómica/efectos de los fármacos , Inestabilidad Genómica/genética , Exposición Materna/efectos adversos , Fumar/efectos adversos , Adolescente , Adulto , Ensayo Cometa , Cotinina/orina , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Femenino , Humanos , Recién Nacido , Masculino , Análisis Multivariante , Embarazo , Adulto Joven
18.
Development ; 138(22): 4843-52, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22028024

RESUMEN

The Lhx2 transcription factor plays essential roles in morphogenesis and patterning of ectodermal derivatives as well as in controlling stem cell activity. Here, we show that during murine skin morphogenesis, Lhx2 is expressed in the hair follicle (HF) buds, whereas in postnatal telogen HFs Lhx2(+) cells reside in the stem cell-enriched epithelial compartments (bulge, secondary hair germ) and co-express selected stem cell markers (Sox9, Tcf4 and Lgr5). Remarkably, Lhx2(+) cells represent the vast majority of cells in the bulge and secondary hair germ that proliferate in response to skin injury. This is functionally important, as wound re-epithelization is significantly retarded in heterozygous Lhx2 knockout (+/-) mice, whereas anagen onset in the HFs located closely to the wound is accelerated compared with wild-type mice. Cell proliferation in the bulge and the number of Sox9(+) and Tcf4(+) cells in the HFs closely adjacent to the wound in Lhx2(+/-) mice are decreased in comparison with wild-type controls, whereas expression of Lgr5 and cell proliferation in the secondary hair germ are increased. Furthermore, acceleration of wound-induced anagen development in Lhx2(+/-) mice is inhibited by administration of Lgr5 siRNA. Finally, Chip-on-chip/ChIP-qPCR and reporter assay analyses identified Sox9, Tcf4 and Lgr5 as direct Lhx2 targets in keratinocytes. These data strongly suggest that Lhx2 positively regulates Sox9 and Tcf4 in the bulge cells, and promotes wound re-epithelization, whereas it simultaneously negatively regulates Lgr5 in the secondary hair germ and inhibits HF cycling. Thus, Lhx2 operates as an important regulator of epithelial stem cell activity in the skin response to injury.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Epidermis/fisiología , Folículo Piloso/metabolismo , Proteínas con Homeodominio LIM/fisiología , Receptores Acoplados a Proteínas G/genética , Regeneración/genética , Factor de Transcripción SOX9/genética , Células Madre/fisiología , Factores de Transcripción/fisiología , Animales , Animales Recién Nacidos , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Células Cultivadas , Embrión de Mamíferos , Epidermis/lesiones , Epidermis/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Folículo Piloso/citología , Humanos , Proteínas con Homeodominio LIM/antagonistas & inhibidores , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Ratones , Ratones Transgénicos , ARN Interferente Pequeño/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Factor de Transcripción SOX9/metabolismo , Células Madre/metabolismo , Factor de Transcripción 4 , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/genética
19.
J Cell Biol ; 194(6): 825-39, 2011 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-21930775

RESUMEN

During development, multipotent progenitor cells establish tissue-specific programs of gene expression. In this paper, we show that p63 transcription factor, a master regulator of epidermal morphogenesis, executes its function in part by directly regulating expression of the genome organizer Satb1 in progenitor cells. p63 binds to a proximal regulatory region of the Satb1 gene, and p63 ablation results in marked reduction in the Satb1 expression levels in the epidermis. Satb1(-/-) mice show impaired epidermal morphology. In Satb1-null epidermis, chromatin architecture of the epidermal differentiation complex locus containing genes associated with epidermal differentiation is altered primarily at its central domain, where Satb1 binding was confirmed by chromatin immunoprecipitation-on-chip analysis. Furthermore, genes within this domain fail to be properly activated upon terminal differentiation. Satb1 expression in p63(+/-) skin explants treated with p63 small interfering ribonucleic acid partially restored the epidermal phenotype of p63-deficient mice. These data provide a novel mechanism by which Satb1, a direct downstream target of p63, contributes in epidermal morphogenesis via establishing tissue-specific chromatin organization and gene expression in epidermal progenitor cells.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Cromatina/metabolismo , Epidermis/embriología , Regulación del Desarrollo de la Expresión Génica , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Fosfoproteínas/genética , Transactivadores/genética , Animales , Diferenciación Celular , Células Epidérmicas , Genoma , Hibridación Fluorescente in Situ , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Fosfoproteínas/metabolismo , Transactivadores/metabolismo
20.
J Invest Dermatol ; 130(2): 398-404, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19710687

RESUMEN

BMP signaling has a crucial role in skin development and homeostasis, whereas molecular mechanisms underlying its involvement in regulating gene expression programs in keratinocytes and fibroblasts remain largely unknown. We show here that several BMP ligands, all BMP receptors, and BMP-associated Smad1/5/8 are expressed in human primary epidermal keratinocytes and dermal fibroblasts. Treatment of both cell types by BMP-4 resulted in the activation of the BMP-Smad, but not BMP-MAPK pathways. Global microarray analysis revealed that BMP-4 treatment induces distinct and cell type-specific changes in gene expression programs in keratinocytes and fibroblasts, which are far more complex than the effects of BMPs on cell proliferation/differentiation described earlier. Furthermore, our data suggest that the potential modulation of cell adhesion, extracellular matrix remodeling, motility, metabolism, signaling, and transcription by BMP-4 in keratinocytes and fibroblasts is likely to be achieved by the distinct and cell-type-specific sets of molecules. Thus, these data provide an important basis for delineating mechanisms that underlie the distinct effects of the BMP pathway on different cell populations in the skin, and will be helpful in further establishing molecular signaling networks regulating skin homeostasis in health and disease.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Fibroblastos/citología , Perfilación de la Expresión Génica , Queratinocitos/citología , Activinas/metabolismo , Adhesión Celular , Células Cultivadas , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Humanos , Ligandos , Modelos Biológicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...