Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
Cell Death Dis ; 15(3): 232, 2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38519456

RESUMEN

Unlike the intense research effort devoted to exploring the significance of heparanase in cancer, very little attention was given to Hpa2, a close homolog of heparanase. Here, we explored the role of Hpa2 in breast cancer. Unexpectedly, we found that patients endowed with high levels of Hpa2 exhibited a higher incidence of tumor metastasis and survived less than patients with low levels of Hpa2. Immunohistochemical examination revealed that in normal breast tissue, Hpa2 localizes primarily in the cell nucleus. In striking contrast, in breast carcinoma, Hpa2 expression is not only decreased but also loses its nuclear localization and appears diffuse in the cell cytoplasm. Importantly, breast cancer patients in which nuclear localization of Hpa2 is retained exhibited reduced lymph-node metastasis, suggesting that nuclear localization of Hpa2 plays a protective role in breast cancer progression. To examine this possibility, we engineered a gene construct that directs Hpa2 to the cell nucleus (Hpa2-Nuc). Notably, overexpression of Hpa2 in breast carcinoma cells resulted in bigger tumors, whereas targeting Hpa2 to the cell nucleus attenuated tumor growth and tumor metastasis. RNAseq analysis was performed to reveal differentially expressed genes (DEG) in Hpa2-Nuc tumors vs. control. The analysis revealed, among others, decreased expression of genes associated with the hallmark of Kras, beta-catenin, and TNF-alpha (via NFkB) signaling. Our results imply that nuclear localization of Hpa2 prominently regulates gene transcription, resulting in attenuation of breast tumorigenesis. Thus, nuclear Hpa2 may be used as a predictive parameter in personalized medicine for breast cancer patients.


Asunto(s)
Neoplasias de la Mama , Glucuronidasa , Humanos , Femenino , Glucuronidasa/genética , Glucuronidasa/metabolismo , Neoplasias de la Mama/genética , Transducción de Señal , Núcleo Celular/metabolismo
2.
Elife ; 122023 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-37934570

RESUMEN

ZMYM2 is a zinc finger transcriptional regulator that plays a key role in promoting and maintaining cell identity. It has been implicated in several diseases such as congenital anomalies of the kidney where its activity is diminished and cancer where it participates in oncogenic fusion protein events. ZMYM2 is thought to function through promoting transcriptional repression and here we provide more evidence to support this designation. Here we studied ZMYM2 function in human cells and demonstrate that ZMYM2 is part of distinct chromatin-bound complexes including the established LSD1-CoREST-HDAC1 corepressor complex. We also identify new functional and physical interactions with ADNP and TRIM28/KAP1. The ZMYM2-TRIM28 complex forms in a SUMO-dependent manner and is associated with repressive chromatin. ZMYM2 and TRIM28 show strong functional similarity and co-regulate a large number of genes. However, there are no strong links between ZMYM2-TRIM28 binding events and nearby individual gene regulation. Instead, ZMYM2-TRIM28 appears to regulate genes in a more regionally defined manner within TADs where it can directly regulate co-associated retrotransposon expression. We find that different types of ZMYM2 binding complex associate with and regulate distinct subclasses of retrotransposons, with ZMYM2-ADNP complexes at SINEs and ZMYM2-TRIM28 complexes at LTR elements. We propose a model whereby ZMYM2 acts directly through retrotransposon regulation, which may then potentially affect the local chromatin environment and associated coding gene expression.


Asunto(s)
Elementos Transponibles de ADN , Retroelementos , Humanos , Dedos de Zinc , Cromatina , Proteínas Co-Represoras , Proteínas de Unión al ADN , Factores de Transcripción
3.
Elife ; 122023 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-36803948

RESUMEN

Cancer is driven by both genetic and epigenetic changes that impact on gene expression profiles and the resulting tumourigenic phenotype. Enhancers are transcriptional regulatory elements that are key to our understanding of how this rewiring of gene expression is achieved in cancer cells. Here, we have harnessed the power of RNA-seq data from hundreds of patients with oesophageal adenocarcinoma (OAC) or its precursor state Barrett's oesophagus coupled with open chromatin maps to identify potential enhancer RNAs and their associated enhancer regions in this cancer. We identify ~1000 OAC-specific enhancers and use these data to uncover new cellular pathways that are operational in OAC. Among these are enhancers for JUP, MYBL2, and CCNE1, and we show that their activity is required for cancer cell viability. We also demonstrate the clinical utility of our dataset for identifying disease stage and patient prognosis. Our data therefore identify an important set of regulatory elements that enhance our molecular understanding of OAC and point to potential new therapeutic directions.


Asunto(s)
Adenocarcinoma , Esófago de Barrett , Neoplasias Esofágicas , Humanos , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Adenocarcinoma/genética , Adenocarcinoma/patología , Esófago de Barrett/genética , Esófago de Barrett/patología , Secuencias Reguladoras de Ácidos Nucleicos , Elementos de Facilitación Genéticos/genética
4.
NAR Cancer ; 5(1): zcad001, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36694726

RESUMEN

Oesophageal adenocarcinoma (OAC) is a deadly disease with poor survival statistics and few targeted therapies available. One of the most common molecular aberrations in OAC is amplification or activation of the gene encoding the receptor tyrosine kinase ERBB2, and ERBB2 is targeted in the clinic for this subset of patients. However, the downstream consequences of these ERBB2 activating events are not well understood. Here we used a combination of phosphoproteomics, open chromatin profiling and transcriptome analysis on cell line models and patient-derived datasets to interrogate the molecular pathways operating downstream from ERBB2. Integrated analysis of these data sets converge on a model where dysregulated ERBB2 signalling is mediated at the transcriptional level by the transcription factor AP-1. AP-1 in turn controls cell behaviour by acting on cohorts of genes that regulate cell migration and adhesion, features often associated with EMT. Our study therefore provides a valuable resource for the cancer cell signalling community and reveals novel molecular determinants underlying the dysregulated behaviour of OAC cells.

5.
Oncogene ; 41(43): 4808-4822, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36153371

RESUMEN

Oesophageal adenocarcinoma (OAC) patients show poor survival rates and there are few targeted molecular therapies available. However, components of the receptor tyrosine kinase (RTK) driven pathways are commonly mutated in OAC, typified by high frequency amplifications of the RTK ERBB2. ERBB2 can be therapeutically targeted, but this has limited clinical benefit due to the acquisition of drug resistance. Here we examined how OAC cells adapt to ERBB2 inhibition as they transition to a drug resistant state. ERBB2 inhibition triggers widespread remodelling of the accessible chromatin landscape and the underlying gene regulatory networks. The transcriptional regulators HNF4A and PPARGC1A play a key role in this network rewiring. Initially, inhibition of cell cycle associated gene expression programmes is observed, with compensatory increases in the programmes driving changes in metabolic activity. Both PPARGC1A and HNF4A are required for the acquisition of resistance to ERBB2 inhibition and PPARGC1A is instrumental in promoting a switch to dependency on oxidative phosphorylation. Our work therefore reveals the molecular pathways that support the acquisition of a resistant state and points to potential new therapeutic strategies to combat cellular adaptation and ensuing drug resistance.


Asunto(s)
Adenocarcinoma , Resistencia a Antineoplásicos , Humanos , Resistencia a Antineoplásicos/genética , Cromatina/genética , Receptor ErbB-2/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Adenocarcinoma/tratamiento farmacológico , Línea Celular Tumoral
6.
Nucleic Acids Res ; 49(22): 12744-12756, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34850951

RESUMEN

High levels of histone acetylation are associated with the regulatory elements of active genes, suggesting a link between acetylation and gene activation. We revisited this model, in the context of EGF-inducible gene expression and found that rather than a simple unifying model, there are two broad classes of genes; one in which high lysine acetylation activity is required for efficient gene activation, and a second group where the opposite occurs and high acetylation activity is inhibitory. We examined the latter class in more detail using EGR2 as a model gene and found that lysine acetylation levels are critical for several activation parameters, including the timing of expression onset, and overall amplitudes of the transcriptional response. In contrast, DUSP1 responds in the canonical manner and its transcriptional activity is promoted by acetylation. Single cell approaches demonstrate heterogenous activation kinetics of a given gene in response to EGF stimulation. Acetylation levels modify these heterogenous patterns and influence both allele activation frequencies and overall expression profile parameters. Our data therefore point to a complex interplay between acetylation equilibria and target gene induction where acetylation level thresholds are an important determinant of transcriptional induction dynamics that are sensed in a gene-specific manner.


Asunto(s)
Código de Histonas , Activación Transcripcional , Acetilación/efectos de los fármacos , Línea Celular , Factor de Crecimiento Epidérmico/fisiología , Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Humanos , Lisina/metabolismo
7.
F1000Res ; 10: 570, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34504687

RESUMEN

Many biological studies of transcriptional control mechanisms produce lists of genes and non-coding genomic intervals from corresponding gene expression and epigenomic assays. In higher organisms, such as eukaryotes, genes may be regulated by distal elements, with these elements lying 10s-100s of kilobases away from a gene transcription start site. To gain insight into these distal regulatory mechanisms, it is important to determine comparative enrichment of genes of interest in relation to genomic regions of interest, and to be able to do so at a range of distances. Existing bioinformatics tools can annotate genomic regions to nearest known genes, or look for transcription factor binding sites in relation to gene transcription start sites. Here, we present PEGS ( Peak set Enrichment in Gene Sets). This tool efficiently provides an exploratory analysis by calculating enrichment of multiple gene sets, associated with multiple non-coding elements (peak sets), at multiple genomic distances, and within topologically associated domains. We apply PEGS to gene sets derived from gene expression studies, and genomic intervals from corresponding ChIP-seq and ATAC-seq experiments to derive biologically meaningful results. We also demonstrate an extended application to tissue-specific gene sets and publicly available GWAS data, to find enrichment of sleep trait associated SNPs in relation to tissue-specific gene expression profiles.


Asunto(s)
Biología Computacional , Genómica , Regulación de la Expresión Génica , Polimorfismo de Nucleótido Simple , Unión Proteica
8.
Science ; 373(6556): 760-767, 2021 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-34385390

RESUMEN

The origin of human metaplastic states and their propensity for cancer is poorly understood. Barrett's esophagus is a common metaplastic condition that increases the risk for esophageal adenocarcinoma, and its cellular origin is enigmatic. To address this, we harvested tissues spanning the gastroesophageal junction from healthy and diseased donors, including isolation of esophageal submucosal glands. A combination of single-cell transcriptomic profiling, in silico lineage tracing from methylation, open chromatin and somatic mutation analyses, and functional studies in organoid models showed that Barrett's esophagus originates from gastric cardia through c-MYC and HNF4A-driven transcriptional programs. Furthermore, our data indicate that esophageal adenocarcinoma likely arises from undifferentiated Barrett's esophagus cell types even in the absence of a pathologically identifiable metaplastic precursor, illuminating early detection strategies.


Asunto(s)
Adenocarcinoma/patología , Esófago de Barrett/patología , Cardias/citología , Neoplasias Esofágicas/patología , Esófago/patología , Esófago de Barrett/genética , Esófago de Barrett/metabolismo , Cardias/química , Diferenciación Celular , Linaje de la Célula , Transformación Celular Neoplásica , Epigénesis Genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Esófago/citología , Esófago/metabolismo , Glándulas Exocrinas/química , Glándulas Exocrinas/citología , Factor Nuclear 4 del Hepatocito/metabolismo , Humanos , Queratina-7/análisis , Metaplasia , Fenotipo , Proteínas Proto-Oncogénicas c-myc/metabolismo , RNA-Seq , Análisis de la Célula Individual , Transcripción Genética , Transcriptoma
10.
Nucleic Acids Res ; 49(3): 1345-1363, 2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33434264

RESUMEN

Enhancers play important roles in controlling gene expression in a choreographed spatial and temporal manner during development. However, it is unclear how these regulatory regions are established during differentiation. Here we investigated the genome-wide binding profile of the forkhead transcription factor FOXK2 in human embryonic stem cells (ESCs) and downstream cell types. This transcription factor is bound to thousands of regulatory regions in human ESCs, and binding at many sites is maintained as cells differentiate to mesendodermal and neural precursor cell (NPC) types, alongside the emergence of new binding regions. FOXK2 binding is generally associated with active histone marks in any given cell type. Furthermore newly acquired, or retained FOXK2 binding regions show elevated levels of activating histone marks following differentiation to NPCs. In keeping with this association with activating marks, we demonstrate a role for FOXK transcription factors in gene activation during NPC differentiation. FOXK2 occupancy in ESCs is therefore an early mark for delineating the regulatory regions, which become activated in later lineages.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/metabolismo , Factores de Transcripción Forkhead/metabolismo , Activación Transcripcional , Linaje de la Célula/genética , Células Cultivadas , Cromatina/metabolismo , Células Madre Embrionarias/citología , Endodermo/citología , Elementos de Facilitación Genéticos , Código de Histonas , Humanos , Mesodermo/citología , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Factores de Transcripción/metabolismo
11.
STAR Protoc ; 1(3): 100187, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33377081

RESUMEN

Mapping the genomic locations of chromatin-associated proteins, such as transcription factors and histone modifications, is key to understanding the mechanisms of transcriptional regulation. ChIPmentation offers a simple and robust way of investigating the genomic binding sites of a protein using relatively low-input material. Here, we present a detailed protocol for the key steps that lead to a successful ChIPmentation experiment, as well as a quick analysis pipeline to examine the data. For complete details on the use and execution of this protocol, please refer to Schmidl et al. (2015). For example data produced by this protocol, please refer to Henriksson et al. (2019) and Zhang et al. (2019).


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Genoma , Mamíferos/metabolismo , Animales , Anticuerpos Monoclonales/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Cromatina/metabolismo , Humanos , Unión Proteica , Sonicación
12.
Elife ; 92020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32880368

RESUMEN

Oesophageal adenocarcinoma (OAC) is one of the most common causes of cancer deaths. Barrett's oesophagus (BO) is the only known precancerous precursor to OAC, but our understanding about the molecular events leading to OAC development is limited. Here, we have integrated gene expression and chromatin accessibility profiles of human biopsies and identified a strong cell cycle gene expression signature in OAC compared to BO. Through analysing associated chromatin accessibility changes, we have implicated the transcription factor KLF5 in the transition from BO to OAC. Importantly, we show that KLF5 expression is unchanged during this transition, but instead, KLF5 is redistributed across chromatin to directly regulate cell cycle genes specifically in OAC cells. This new KLF5 target gene programme has potential prognostic significance as high levels correlate with poorer patient survival. Thus, the repurposing of KLF5 for novel regulatory activity in OAC provides new insights into the mechanisms behind disease progression.


Acid fluids present in the gut can sometimes 'go up' and damage the oesophagus, the pipe that connects the mouth and the stomach. As a result, a small number of individuals can develop Barrett's oesophagus, a condition where cells in the lining of the lower oesophagus show abnormal shapes. In certain patients, these cells then become cancerous, but exactly how this happens is unknown. This lack of understanding contributes to late diagnoses, limited treatment and low survival rates. Many cancers feature 'signature' mutations in a set of genes that controls how a cell can multiply. Yet, in the case of cancers of the lower oesophagus, known genetic changes have had a limited impact on our understanding of the emergence of the disease. Here, Rogerson et al. focused instead on non-genetic changes and studied transcription factors, the proteins that bind to regulatory regions of the DNA to switch genes on and off. A close inspection of cancer cells in the lower oesophagus revealed that, in that state, a transcription factor called KLF5 controls the abnormal activation of genes involved in cell growth. This is linked to the transcription factor adopting a different pattern of binding onto regulatory regions in diseased cells. Crucially, when the cell growth genes regulated by KLF5 are activated, patients have lower survival rates. Further work is now required to examine whether this finding could help to identify patients who are most at risk from developing cancer. More broadly, the results from the work by Rogerson et al. demonstrate how transcription factors can be repurposed in a disease context.


Asunto(s)
Adenocarcinoma/genética , Esófago de Barrett/genética , Ciclo Celular/genética , Neoplasias Esofágicas/genética , Factores de Transcripción de Tipo Kruppel/genética , Adenocarcinoma/patología , Esófago de Barrett/patología , Línea Celular Tumoral , Neoplasias Esofágicas/patología , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo
13.
Nat Commun ; 11(1): 3920, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32764605

RESUMEN

How the genome activates or silences transcriptional programmes governs organ formation. Little is known in human embryos undermining our ability to benchmark the fidelity of stem cell differentiation or cell programming, or interpret the pathogenicity of noncoding variation. Here, we study histone modifications across thirteen tissues during human organogenesis. We integrate the data with transcription to build an overview of how the human genome differentially regulates alternative organ fates including by repression. Promoters from nearly 20,000 genes partition into discrete states. Key developmental gene sets are actively repressed outside of the appropriate organ without obvious bivalency. Candidate enhancers, functional in zebrafish, allow imputation of tissue-specific and shared patterns of transcription factor binding. Overlaying more than 700 noncoding mutations from patients with developmental disorders allows correlation to unanticipated target genes. Taken together, the data provide a comprehensive genomic framework for investigating normal and abnormal human development.


Asunto(s)
Discapacidades del Desarrollo/genética , Epigénesis Genética , Organogénesis/genética , Animales , Animales Modificados Genéticamente , Bases de Datos Genéticas , Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Código de Histonas/genética , Humanos , Modelos Genéticos , Mutación , Organogénesis/fisiología , Regiones Promotoras Genéticas , Distribución Tisular , Factores de Transcripción/metabolismo , Pez Cebra/embriología , Pez Cebra/genética
14.
Pigment Cell Melanoma Res ; 33(5): 695-708, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32145051

RESUMEN

A major challenge for managing melanoma is its tumour heterogeneity based on individual co-existing melanoma cell phenotypes. These phenotypes display variable responses to standard therapies, and they drive individual steps of melanoma progression; hence, understanding their behaviour is imperative. Melanoma phenotypes are defined by distinct transcriptional states, which relate to different melanocyte lineage development phases, ranging from a mesenchymal, neural crest-like to a proliferative, melanocytic phenotype. It is thought that adaptive phenotype plasticity based on transcriptional reprogramming drives melanoma progression, but at which stage individual phenotypes dominate and moreover, how they interact is poorly understood. We monitored melanocytic and mesenchymal phenotypes throughout melanoma progression and detected transcriptional reprogramming at different stages, with a gain in mesenchymal traits in circulating melanoma cells (CTCs) and proliferative features in metastatic tumours. Intriguingly, we found that distinct phenotype populations interact in a cooperative manner, which generates tumours of greater "fitness," supports CTCs and expands organotropic cues in metastases. Fibronectin, expressed in mesenchymal cells, acts as key player in cooperativity and promotes survival of melanocytic cells. Our data reveal an important role for inter-phenotype communications at various stages of disease progression, suggesting these communications could act as therapeutic target.


Asunto(s)
Adaptación Fisiológica , Comunicación Celular , Progresión de la Enfermedad , Melanoma/patología , Animales , Línea Celular Tumoral , Proliferación Celular , Fibronectinas/metabolismo , Humanos , Melanocitos/patología , Mesodermo/patología , Ratones , Metástasis de la Neoplasia , Células Neoplásicas Circulantes/patología , Fenotipo
15.
J Integr Bioinform ; 16(3)2019 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-31301672

RESUMEN

The interconversion of sequences that constitute the genome and the proteome is becoming increasingly important due to the generation of large amounts of DNA sequence data. Following mapping of DNA segments to the genome, one fundamentally important task is to find the amino acid sequences which are coded within a list of genomic sections. Conversely, given a series of protein segments, an important task is to find the genomic loci which code for a list of protein regions. To perform these tasks on a region by region basis is extremely laborious when a large number of regions are being studied. We have therefore implemented an R package geno2proteo which performs the two mapping tasks and subsequent sequence retrieval in a batch fashion. In order to make the tool more accessible to users, we have created a web interface of the R package which allows the users to perform the mapping tasks by going to the web page http://sharrocksresources.manchester.ac.uk/tofigaps and using the web service.


Asunto(s)
Bases de Datos de Ácidos Nucleicos , Genoma , Proteínas/genética , Análisis de Secuencia de ADN , Análisis de Secuencia de Proteína , Programas Informáticos
16.
Wellcome Open Res ; 4: 41, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31346550

RESUMEN

Background: The ERK MAPK pathway plays a pivotal role in regulating numerous cellular processes during normal development and in the adult but is often deregulated in disease scenarios. One of its key nuclear targets is the transcription factor ELK1, which has been shown to play an important role in controlling gene expression in human embryonic stem cells (hESCs). ELK1 is known to act as a transcriptional activator in response to ERK pathway activation but repressive roles have also been uncovered, including a putative interaction with the PRC2 complex. Methods: Here we probe the activity of ELK1 in hESCs by using a combination of gene expression analysis in hESCs and during differentiation following ELK1 depletion and also analysis of chromatin occupancy of transcriptional regulators and histone mark deposition that accompany changes in gene expression. Results: We find that ELK1 can exert its canonical activating activity downstream from the ERK pathway but also possesses additional repressive activities. Despite its co-binding to PRC2 occupied regions, we could not detect any ELK1-mediated repression at these regions. Instead, we find that ELK1 has a repressive role at a subset of co-occupied SRF binding regions. This latter repressive role appears not to be exerted through competition with MRTF family co-activators. Conclusions: ELK1 should therefore be viewed as a dichotomous transcriptional regulator that can act through SRF to generate both activating and repressing properties at different genomic loci.

17.
Cell Rep ; 27(11): 3215-3227.e6, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31189106

RESUMEN

Embryonic stem cells (ESCs) must transition through a series of intermediate cell states before becoming terminally differentiated. Here, we investigated the early events in this transition by determining the changes in the open chromatin landscape as naive mouse ESCs transition to epiblast-like cells (EpiLCs). Motif enrichment analysis of the newly opening regions coupled with expression analysis identified ZIC3 as a potential regulator of this cell fate transition. Chromatin binding and genome-wide transcriptional profiling following Zic3 depletion confirmed ZIC3 as an important regulatory transcription factor, and among its targets are genes encoding a number of transcription factors. Among these is GRHL2, which acts through enhancer switching to maintain the expression of a subset of genes from the ESC state. Our data therefore place ZIC3 upstream of a set of pro-differentiation transcriptional regulators and provide an important advance in our understanding of the regulatory factors governing the early steps in ESC differentiation.


Asunto(s)
Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Ensamble y Desensamble de Cromatina , Elementos de Facilitación Genéticos , Proteínas de Homeodominio/genética , Ratones , Células Madre Embrionarias de Ratones/citología , Factores de Transcripción/genética , Transcriptoma
18.
Genome Res ; 29(5): 723-736, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30962179

RESUMEN

Esophageal adenocarcinoma (EAC) is one of the most frequent causes of cancer death, and yet compared to other common cancers, we know relatively little about the molecular composition of this tumor type. To further our understanding of this cancer, we have used open chromatin profiling to decipher the transcriptional regulatory networks that are operational in EAC. We have uncovered a transcription factor network that is usually found in primitive intestinal cells during embryonic development, centered on HNF4A and GATA6. These transcription factors work together to control the EAC transcriptome. We show that this network is activated in Barrett's esophagus, the putative precursor state to EAC, thereby providing novel molecular evidence in support of stepwise malignant transition. Furthermore, we show that HNF4A alone is sufficient to drive chromatin opening and activation of a Barrett's-like chromatin signature when expressed in normal human epithelial cells. Collectively, these data provide a new way to categorize EAC at a genome scale and implicate HNF4A activation as a potential pivotal event in its malignant transition from healthy cells.


Asunto(s)
Adenocarcinoma/genética , Esófago de Barrett/genética , Neoplasias Esofágicas/genética , Factor de Transcripción GATA6/metabolismo , Redes Reguladoras de Genes/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Adenocarcinoma/metabolismo , Biomarcadores de Tumor/genética , Progresión de la Enfermedad , Neoplasias Esofágicas/metabolismo , Femenino , Células HEK293 , Humanos , Masculino , Transcriptoma
20.
Nucleic Acids Res ; 47(2): e10, 2019 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-30335168

RESUMEN

ATAC-seq is a recently developed method to identify the areas of open chromatin in a cell. These regions usually correspond to active regulatory elements and their location profile is unique to a given cell type. When done at single-cell resolution, ATAC-seq provides an insight into the cell-to-cell variability that emerges from otherwise identical DNA sequences by identifying the variability in the genomic location of open chromatin sites in each of the cells. This paper presents Scasat (single-cell ATAC-seq analysis tool), a complete pipeline to process scATAC-seq data with simple steps. Scasat treats the data as binary and applies statistical methods that are especially suitable for binary data. The pipeline is developed in a Jupyter notebook environment that holds the executable code along with the necessary description and results. It is robust, flexible, interactive and easy to extend. Within Scasat we developed a novel differential accessibility analysis method based on information gain to identify the peaks that are unique to a cell. The results from Scasat showed that open chromatin locations corresponding to potential regulatory elements can account for cellular heterogeneity and can identify regulatory regions that separates cells from a complex population.


Asunto(s)
Análisis de Secuencia de ADN/métodos , Análisis de la Célula Individual/métodos , Programas Informáticos , Animales , Ontologías Biológicas , Células/clasificación , Cromatina/química , Análisis por Conglomerados , Enfermedad/genética , Genómica , Humanos , Células K562 , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...