Skip to main content
Erschienen in: Journal of Mammary Gland Biology and Neoplasia 4/2020

Open Access 24.02.2021

How to Use Online Tools to Generate New Hypotheses for Mammary Gland Biology Research: A Case Study for Wnt7b

verfasst von: Yorick Bernardus Cornelis van de Grift, Nika Heijmans, Renée van Amerongen

Erschienen in: Journal of Mammary Gland Biology and Neoplasia | Ausgabe 4/2020

Abstract

An increasing number of ‘-omics’ datasets, generated by labs all across the world, are becoming available. They contain a wealth of data that are largely unexplored. Not every scientist, however, will have access to the required resources and expertise to analyze such data from scratch. Fortunately, a growing number of investigators is dedicating their time and effort to the development of user friendly, online applications that allow researchers to use and investigate these datasets. Here, we will illustrate the usefulness of such an approach. Using regulation of Wnt7b expression as an example, we will highlight a selection of accessible tools and resources that are available to researchers in the area of mammary gland biology. We show how they can be used for in silico analyses of gene regulatory mechanisms, resulting in new hypotheses and providing leads for experimental follow up. We also call out to the mammary gland community to join forces in a coordinated effort to generate and share additional tissue-specific ‘-omics’ datasets and thereby expand the in silico toolbox.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

The experimental technology that allows genome wide analyses at the molecular level (genomics, epigenomics, transcriptomics, metabolomics and proteomics – hereafter combinedly referred to as ‘omics’ approaches) continues to evolve at breathtaking speed. Despite the fact that these techniques are becoming more affordable and therefore more widely available for scientists worldwide, they are still quite expensive – a prohibitory factor for those with limited financial resources. This is especially true for sophisticated approaches such as single-cell RNA sequencing (scRNAseq) and other-single cell approaches that are still being developed. Moreover, not everyone will have local access to the required infrastructure. Of course, scientific collaborations can offer a solution. Even then, it can be a challenge to integrate a variety of these technologies into one’s research program [1].
As can be gleaned from the published literature, all too frequently only a few hits or top candidates are followed up in instances where genome-wide datasets are generated. As a consequence, a wealth of data remains unexplored. These datasets constitute a rich and valuable resource for the larger scientific community. As an example, we have previously used published microarray data to identify the most stably rather than the most differentially expressed genes, resulting in a new set of reference genes for qRT-PCR studies in the developing mouse mammary gland [2].
Most ‘omics’ datasets are deposited in public repositories such as the NCBI Gene Expression Omnibus (https://​www.​ncbi.​nlm.​nih.​gov/​geo/​), either in raw format or in a more processed form. While this makes them available to all scientists in theory, in practice not everyone has the bioinformatics skills and expertise to analyze these data from scratch. Fortunately, multiple labs are dedicating their time and effort to the development of online tools that allow easy and intuitive access to these datasets, allowing researchers to explore them via a user friendly graphical interface.
Here we will highlight a selection of these online tools and demonstrate how they can be used to generate hypotheses and answer biological questions in the context of mammary gland biology. To illustrate this approach, we will build a case study around Wnt7b, a gene that has been implicated in mammary gland development and breast cancer, but whose precise activity and mode of regulation remain unknown.
We assume that the reader is familiar with the basic principles behind the different techniques (e.g. scRNAseq, snATACseq, Hi-C), as well as with the way in which these data are commonly presented (e.g. tSNE plots). Please note that for all figures we have kept the exact style and color schemes as generated by the different online tools to aid the reader in recognizing the output when they try out these tools for themselves.

WNT7B in Mammary Gland Development and Breast Cancer

WNT7B is expressed in human breast tissue and its expression has been reported to be altered in breast cancer [3, 4]. Its overexpression has been associated with a poor prognosis and reduced overall survival of breast cancer patients [5]. In breast cancer, WNT7B has not only been shown to be expressed by the tumor cells, but also by myeloid cells present in the local microenvironment. The latter promotes angiogenesis, invasion and metastasis [6].
Its murine counterpart, Wnt7b, is expressed in the ductal epithelium of the mouse mammary gland [7]. The levels of Wnt7b remain unaltered following ovariectomy, suggesting that regulation of Wnt7b expression is estrogen and progesterone independent [7]. During puberty, expression of Wnt7b is enriched in the terminal end bud epithelium, suggesting a role in branching morphogenesis [8]. Wnt7b has been reported to have mild transforming activities in vitro [9, 10] and in vivo [11] although not all studies agree on the extent of this effect [10, 12].
The precise role and regulation of Wnt7b/WNT7B in the mammary gland or breast remain unknown. So far, evidence that WNT7B protein can promote the activation of CTNNB1/TCF transcriptional complexes is lacking, despite the fact that Wnt7b is readily detected and shows prominent expression in luminal cells [13]. This is in contrast to other tissues, such as the skin, where the activities of WNT7B have been linked to CTNNB1/TCF driven processes [14].

Exploring Spatiotemporal Patterns of Wnt7b Expression Using scRNAseq Data

Public scRNAseq datasets are an ideal platform to start investigating spatiotemporal gene expression in the mammary gland [15, 16]. We want to highlight three user friendly scRNAseq tools that allow analysis of the in vivo expression patterns of a gene of interest in the postnatal stages of mouse mammary gland development (Box 1). Their combined use reveals extensive details about the expression pattern of any given gene across different stages and cell populations.
Wnt7b expression is absent (or at least below the limit of detection) in the fetal mammary gland (E18, Fig. 1a), but emerges postnatally (Fig. 1b-d, Fig. 2a,d, Fig. 3a-d). Its expression is cell type specific, displaying high gene expression in the luminal compartment, and low or absent expression in basal cells and supportive tissues (fat, endothelial, immune and stromal cells) (Fig. 1b-d, Fig. 2b-e, Fig. 3a-d).
Spatiotemporal expression is dynamically regulated throughout the adult reproductive cycle (Fig. 2a,d, Fig. 3a-d). In nulliparous mice, Wnt7b is expressed in luminal progenitor cells, as well as in more differentiated, hormone-sensing luminal progeny (Fig. 2b-e). During gestation and lactation Wnt7b expression is (mostly) switched off, but it re-emerges post-involution (Fig. 2a,d, Fig. 3a-d). Thus, it is exclusively expressed in the ‘resting’ state, be it nulliparous or post-involution. Of note, although the luminal progenitor population itself re-appears post-involution, Wnt7b expression is lost in this population, becoming restricted to the hormone-sensing luminal lineage post-pregnancy (Fig. 2b-f).
From these analyses we would conclude that Wnt7b is expressed exclusively in the luminal compartment of the nulliparous mammary gland, is lost during pregnancy, and is re-established post-involution (Fig. 4). Indeed, this is supported by other studies showing that Wnt7b is expressed in the virgin mammary gland, but drops at Preg12.5 of pregnancy to undetectable levels [7]. This underscores the validity of this approach and illustrates the usefulness of interactive in silico tools to determine spatiotemporal patterns of in vivo gene expression.
Table 1
Compilation of publicly available online tools that are outside the scope of the current case study. These tools are not specific for mammary gland biology and/or do not always include mammary gland datasets. Many more tools and resources are available as a stepping stone for those willing to invest the time in developing more advanced bioinformatics and data analysis skills, such as those maintained by the Broad Institute
Tool
Description
Reference
dbSuper is an interactive database containing more than 80,000 putative super enhancers for 25 mouse and >100 human tissues and cell lines. The database has migrated from its original reported location (http://​bioinfo.​au.​tsinghua.​edu.​cn/​dbsuper/​) and while functional and highly intuitive, it is not clear whether it has been updated since 2017.
[19]
SEA version 3.0 was updated in 2019 and promises to be a comprehensive resource that stores predicted super-enhancers and enhancers from 11 different species and more than 200 types of cells, tissues and diseases.
[20]
A large compendium of single cell transcriptome data from the model organism Mus musculus that contains scRNAseq datasets of 23 organs and tissues, including the mammary gland at 6 different timepoints (1 month, 3 months, 18 months, 21 months, 24 months, 30 months). This online dataset explicitly includes stromal cells and other cell types from the supportive tissue (e.g. endothelial and immune cells). Of note, all tissues have been processed and analysed by two different protocols: cells were either FACS sorted, or single-cell sorted using microfluidic droplet-capture techniques and thus sequenced using two different methodologies, providing an innate technical validation of the data when using this tool.
[21]
Also part of the Tabula Muris Senis effort. Offers extensive statistical analysis and visualization of bulk RNA seq datasets from 17 organs of Mus musculus at 10 different timepoints.
[22]
3DIV collects human Hi-C data from 80 cells lines or tissues (including HMEC, MCF7, MCF10A) and promoter capture Hi-C from 27 tissues. Chromatin conformation data from the locus of a gene or location of interest can be either displayed as a Hi-C heatmap and as a virtual 4C (with the location of interest as viewpoint). If applicable, it also predicts the boundaries of local TADs based on the provided datasets. 3DIV offers more flexibility to its users as it allows the user to select the algorithm used to predict TADs, define the cut-off for positive interactions in the virtual 4C and it is straightforward to extract the coordinates of positive hits.
[23, 24]
Single Cell Expression Atlas & Gene Expression Atlas: A database that compiles and visualizes published RNA & scRNA-seq datasets from Human, Mouse & a wide variety of model organisms. Selected datasets are plotted as a tSNE, and a heatmap highlighting marker genes for each annotated cluster is displayed. The database can be searched by gene across species, experiments, tissues and cell lines to reveal where this gene is expressed.
[25]
HACER is an atlas of Human ACtive Enhancer to interpret Regulatory variants, which includes active, transcribed enhancers derived from GRO-seq, PRO-seq and CAGE data. HACER not only compiles cell type specific enhancers but also integrates transcription factor-enhancer binding prediction, validated chromatin interactions and links GWAS SNPs and eQTL variants to enhancer regions. The database includes the MCF10A and MCF7 cell lines.
[26]
An online database that compiles published spatial transcriptomic datasets and offers a web interface for spatially resolved transcriptomic data visualisation and comparison. Includes a human breast cancer dataset.
[27]
tSNE visualisation of gene expression during mammary gland development: from E16 to Adult.
[17]
PanglaoDB is a database that collects and integrates scRNAseq data from human and mouse and presents them through an unified framework.
[28]
The database provides enhancer annotation in nine species, including human (hg19), mouse (mm9), fly (dm3), worm (ce10), zebrafish (danRer10), rat (rn5), yeast (sacCer3), chicken (galGal4), and boar (susScr3). The consensus enhancers were predicted based on multiple high throughput experimental datasets (e.g. histone modification, CAGE, GRO-seq, transcription factor binding and DHS). This database includes the HMEC cell line.
[29, 30]
A database visualized by an intuitive shiny app that allows for an interactive exploration of gene expression profiles across tissues, developmental stages and species. This does not only include protein coding genes but also putative LncRNAs. The mammary gland is not included in this dataset.
[31, 32]
ARCHS4 is a web resource that compiles the majority of RNA-seq data published from both human and mouse datasets and makes that available at the gene and transcript levels. It provides a web-interface that allows exploration of the processed data. Moreover, individual genes can be searched for their average expression across cell lines and tissues, top co-expressed genes, and predicted biological functions and protein-protein interactions.
[33]
Cistrome DB is a comprehensive database (~47.000 sets) for curated ChIP and DNase-seq data. It provides an uniform platform that contains manually curated information for each ChIP-seq and DNase-seq dataset, including species, factors, biological source, publication etc, the analysis results of each dataset from human and mouse, and comprehensive quality control checks across the complete database. By using the CistromeDB toolkit, epigenetic features or transcription factors that regulate your gene of interest can be predicted based on the datasets present in Cistrome DB.
[34, 35]
The Human Cell Landscape offers a large compendium of human scRNA-seq data. Mammary gland tissue is not included in the original dataset, but scRNA-seq data from Nguyen et al. 2018 has been integrated in the online visualisation tool. Gene expression can be visualised superimposed on a tSNE plot.
[36]
A ‘gene atlas for structural immunity’. This multi-omics dataset profiles the immunological potential of epithelial cells, endothelial cells and fibroblasts from 12 different mouse tissues. The mammary gland is not included in this dataset. Aggregated ATAC-seq, ChIP-seq and RNAseq can be visualised in the UCSC genome browser.
[37]
The cBioPortal for cancer genomics is an open-access resource for exploring and visualizing multidimensional cancer genomics datasets. cBioPortal compiles a wide variety of datasets, including TCGA, that can contain non-synonymous mutations, DNA copy-number variation, mRNA and microRNA expression data, protein-level and phosphoprotein level data, DNA methylation, and de-identified clinical data.
[38, 39]
The Kaplan-meier plotter is a tool that can be used to assess the effect of 54k genes (mRNA and protein levels) on survival across 21 cancer types, including breast, ovarian, lung and gastric cancer. Sources for these datasets include GEO, TCGA, and EGA. This is a valuable and easy to use tool for discovering and validating cancer survival biomarkers.
[81]
Enrichr is a comprehensive online tool or gene set enrichment analysis that includes over 30 gene-set libraries. It offers interactive and intuitive visualisation of the results via clustergrams. Note that while highly useful this tool requires predefined gene sets from e.g. RNA-seq and is not as useful for e.g. searching KEGG or GO terms for a gene of interest. For these kind of queries ARCHS4 is better suited.
[40, 41]

Identifying Putative Regulatory Elements

Little is known about the molecular signals and cis-regulatory elements that control mouse Wnt7b or human WNT7B gene expression. In ER-/HER2 + breast tumors, WNT7B was shown to be a direct transcriptional target of the androgen receptor (AR) [42] and predicted to be regulated by Nuclear respiratory factor 1 (NRF1) [43]. Although Wnt7b is also expressed in hormone-responsive cells (Fig. 2 and [13]), at present there is no experimental evidence to support that its expression is regulated by steroid hormones, in particular progesterone [44]. Wnt7b expression is not limited to the mammary gland, however. It is required for lung [45, 46], and kidney development [47] to name but a few and can therefore be regulated by a myriad of signals.
One way to gain understanding into tissue-specific gene expression, is to identify cis-acting enhancer elements. Using ChIPseq analysis, a recent study predicted 440 mammary-specific super-enhancers [48]. Super-enhancers can be classified as dense clusters of transcriptional enhancers that are likely to control genes important for cell type specification [4749]. Only one of these was followed up in more detail in that particular study. However, a supplementary file listing all 440 of these putative regulatory elements is available. We were particularly intrigued by a sequence that spans more than 24 kb on chromosome 15 (published mm9 coordinates chr15: 85,475,778–85,500,063, mm10 coordinates chr15: 85,645,348–85,669,633), which was assigned as a putative regulator of the nearest gene: Wnt7b (Fig. 5a). While it is common to do so, linear proximity alone is not an accurate measure for functional interaction between an enhancer and its putative target gene [49, 50]. Other genes in this region – including two miRNAs (Mirlet7c-2/Mirlet7b) and a protein coding gene (Ppara) – might also be regulated by this particular super-enhancer. A region on the edge of this super-enhancer (mm9 coordinates chr15:85,473,689–85,478,592, published mm10 coordinates chr15: 85,643,259–85,648,162) was recently indeed associated with Wnt7b, albeit not in the mammary gland but in a mouse model for hair-follicle derived skin tumors, and based on strain-specific polymorphisms rather than on having been shown to directly regulate Wnt7b expression [14]. These results show that association of this super-enhancer with Wnt7b in the mammary gland is worthy of follow-up analysis.
The term “super-enhancer” is used to define a larger chromatin area that contains clusters of smaller, individual enhancers and that is enriched for active chromatin marks (e.g. H3K27ac) or occupied by transcriptional activators (e.g. MED1) and master regulatory transcription factors (e.g. STAT5A) [48, 51, 52]. More than 80,000 super-enhancers (combined numbers for the mouse and human genome) can be accessed through the online dbSuper database [19]. An updated version of the Super Enhancer Archive (SEA 3.0) provides another entry point [20] (Table 1).
A first screen of the dbSuper database shows the tissue-specificity of super-enhancers: a putative Wnt7b super-enhancer has also been identified in the murine heart, lung and testis. However, this sequence does not overlap with the mammary-specific super-enhancer described by Shin et al. [48]. Instead, the dbSuper database predicts this particular location to contain two super-enhancers, identified in hair follicle stem cells, linked to Mirlet7c-2/Mirlet7b [53]. Additional super-enhancers in this region, identified in the kidney and the liver, are tentatively associated with Ppara (Fig. 5b). It should be noted that also in dbSuper, super-enhancers and their associated genes are linked based on a simple proximity rule to the nearest transcriptional start site (TSS) [19]. Out of the genes located in this ~ 500 kb area on chromosome 15, only Atxn10 and Wnt7b show prominent expression in one or more mammary gland cell subpopulations, although Ppara, Mirlet7c-2/b and a non-coding RNA, Lncppara, may be differentially expressed at low levels (Fig. 5c,d).

Determining the Boundaries of the Wnt7b Regulatory Domain

In recent years, it has become generally accepted that regulatory elements control target gene expression within the confines of larger, structurally ordered regions of the chromatin known as topologically associating domains (TADs) [54]. Specific DNA sequences (i.e. regulatory elements and their target genes) are much more likely to interact within a TAD, than across a TAD boundary. A logical next step in exploring the potential regulation of Wnt7b by the aforementioned mammary-specific super-enhancer would therefore be to determine the boundaries of the Wnt7b TAD.
We used the 3D Genome Browser (Box 2) to visualize TAD predictions of the Wnt7b locus using publicly available Hi-C datasets [55]. In this browser, TAD boundary predictions are calculated according to the so-called directionality index, which is a method that looks at the degree of up- and downstream interaction bias for DNA regions [56]. It was noted that DNA regions at the periphery of TADs are highly biased in their direction of interaction. Upstream regions in a TAD are highly biased towards interacting with downstream regions and vice versa. Using this directional bias, the boundaries of adjacent TADs can be predicted. Their coordinates are provided by the 3D Genome Browser, which also includes an intuitive visual reference (Fig. 6).
Only one mammary-specific Hi-C dataset is currently available, derived from human mammary epithelial cells (HMEC) [57]. However, TADs have been reported to be stable across cell types and even species [56, 60]. Although not all TAD boundaries are equally stable [61], TAD organization can therefore also be investigated using Hi-C datasets generated from a different tissue as input.
According to this analysis, the Wnt7b TAD boundary lies immediately upstream of the Wnt7b TSS in both HMECs and mouse lymphoma cells (Fig. 6a,b). This would imply that the mammary-specific super-enhancer identified by Shin et al. lies outside of the predicted Wnt7b TAD, which makes it less likely that this particular super-enhancer directly regulates the expression of Wnt7b. However, in other Hi-C datasets this TAD boundary is less well defined (Fig. 6c,d).

Discovering Novel Regulatory Interactions

To gain a better understanding of how the spatiotemporal expression of Wnt7b is regulated in the adult mammary gland, we can start by probing the epigenetic state of the Wnt7b locus in an R shiny app published by the Wahl lab (Box 3). This tool not only allows chromatin accessibility and relevant histone modifications to be examined, but also can be used to make predictions about specific promoters and their regulatory sequences of interest. An attractive graphical interface allows intuitive interpretation of the data (Fig. 7).
If we focus our attention on the Wnt7b promoter and gene region (i.e. the center portion of Fig. 7), snATACseq reveals that the chromatin is relatively accessible in all mammary cell type subpopulations irrespective of Wnt7b gene expression levels (Fig. 7, top 5 rows). In contrast, H3K27ac of the Wnt7b promoter and gene region is exclusively enriched in the luminal compartment (Fig. 7, bottom 4 rows in red). This suggests that Wnt7b is ‘primed’ and open in all epithelial cells in the mammary gland, but its potential for increased gene expression is only realized in the luminal compartment where the chromatin displays the proper histone acetylation marks.
Combining the Cicero algorithm (see Box 3) with snATACseq data, this online tool can also be used to infer co-accessibility of distal sites and the promoter of their putative genes in individual cells. In this manner, Cicero can predict cis-regulatory elements that would be able to interact with the Wnt7b promoter in vivo. At a co-accessibility threshold of 0.15, Cicero identifies 10 regions within 150 kb up- or downstream of the viewpoint that interact with the promoter of Wnt7b. Of these, 4 are located upstream of Wnt7b in an area dense with H3K27ac that encompasses, but extends beyond, the super-enhancer region, and 6 are located downstream of Wnt7b (Fig. 7).
The interacting regions depicted to the left of the Wnt7b promoter (regions 1–6, located 3′ distal to the TSS) all fall within in the predicted Wnt7b TAD (compare Fig. 6,7). These distal sites are either somewhat enriched for chromatin accessibility or H3K27ac, or a combination of both epigenetic features, in adult luminal progenitor and mature luminal cells compared to the adult basal subpopulation (Fig. 7,8). The 4 regions downstream of Wnt7b (7–10) do not display evident changes in chromatin accessibility or H3K27ac when luminal cells are compared to the basal compartment, except for region 8 (Fig. 7,8). Note that the distance between region 9 and 10 spans more than 60 kb, which is considerably larger than the reported size of the mammary-specific super-enhancer. This entire stretch of 60 kb shows characteristic marks of active and open chromatin, suggesting that a much larger collection of regulatory elements may exist in this area (Fig. 7).

Exploring Conservation of Putative Regulatory Enhancer Sequences

In previous studies, highly conserved sequences were associated with developmental and transcriptional regulators [6773]. Given the fundamental role of Wnt signaling not only in vertebrate development [74], but also specifically in mammary gland development and maintenance [7577], focusing on conserved sequences could be another criteria for the selection of candidate Wnt7b enhancers. To identify conserved regions in the vicinity of Wnt7b, we used the evolutionary conserved region (ECR) browser (Box 4).
Often, conservation is scored across vertebrate species. However, in an attempt to identify regions that are specifically conserved in mammals, we specifically selected candidate sequences in a region of ~ 100 kb up- and downstream of the Wnt7b TSS that are conserved across mammalian, but not necessarily in non-mammalian vertebrate species available in the ECR browser (Fig. 9).

A Working Model for Follow-Up Studies

Of course, none of these approaches (sequence conservation, histone modification, transcription factor ChIPseq), either by themselves or in combination, are sufficient to definitively link any of these putative regulatory elements to Wnt7b. This requires further experimental validation and specific follow up. However, as a prediction tool these combined analyses provide an excellent starting point for dissecting this super-enhancer in more detail. If we put all of the different pieces of information together (Fig. 10), we can draft some hypotheses regarding the regulation of Wnt7b expression in the mammary gland.
First, we propose that in mammary epithelial cells the proposed TAD boundary immediately upstream of Wnt7b (Fig. 6) is not very stable, given that the Cicero algorithm predicts four interactions between the Wnt7b promoter and regions to the right of this presumed TAD boundary (i.e. regions 7–10 in Fig. 7). Of note, two of these interactions (Cicero regions 7 and 8) occur in the direct vicinity of this presumed TAD boundary. The other two interactions (Cicero regions 9 and 10) border a large area of active chromatin, which extends beyond the super-enhancer region previously identified by Shin et al. [48]. This 60 kb area harbors an annotated lncRNA (Lncppara) and two microRNAs, MirLet7b/MirLet7c-2, which are broadly expressed and implicated in cancer formation [7880]. Moreover, this region also contains multiple conserved sequences that could represent functional enhancer elements (including ECR_6, ECR_7 and ECR_8 from Fig. 9).
Second, if we do take the TAD boundary prediction into account, it may be wise to prioritize the interactions that occur between Wnt7b and more downstream sequences (i.e. regions 1–6 in Fig. 7). Although the coordinates from the Cicero prediction algorithm deserve further scrutiny of the original datasets, these downstream interacting regions also lie in close vicinity to conserved sequence elements.
Third, in combination with the expression data analysis (Fig. 1-3), the published literature and the active enhancer marks (Fig. 7,8), we can make a further prioritization of putative Wnt7b enhancer sequences that are worthy of experimental validation and follow up. In this case, region 2 is particularly interesting as is has the highest Cicero score and displays both differential chromatin accessibility and H3K27 acetylation in the luminal compartment.
To summarize, by using publicly available online tools we assessed the genomic conformation of the Wnt7b locus, and how this relates to the previously identified putative Wnt7b super enhancer. By examining the epigenetic status of the Wnt7b locus more closely, we noticed that although the Wnt7b promoter is predicted to interact with the super-enhancer region, this is likely not cell type specific as both chromatin accessibility and H3K27ac do not change between the basal and luminal lineages in this region. However, regions downstream of Wnt7b do change their epigenetic status in accordance to Wnt7b gene expression and are also predicted to interact with the Wnt7b promoter. This entire area would be worthy of experimental follow up to definitively associate specific regulatory elements with Wnt7b and/or other nearby genes – in particular the miRNAs and Ppara.

Discussion

Using publicly available genome wide datasets and accessible online tools, we have identified several regions that might play a role in the regulation of spatiotemporal expression of Wnt7b in the mouse mammary gland. Our main goal was to show the reader how these findings provide additional information for future investigations. However, we also want to use this opportunity to highlight and stress the added value of making large datasets available to a wide audience through interactive online tools. We thank our colleagues who invest their resources to do so.
At the same time, we call for joint efforts from our community to ensure that the repertoire of tools as well as of accessible datasets continues to grow and remains of high quality and value to investigators worldwide. As others have undoubtedly noticed, mammary gland and breast tissue datasets are often notoriously absent from public, large-scale -omics efforts. Generating and curating additional genome wide datasets (e.g. Hi-C and others) for both epithelial and stromal cells of multiple species, including mouse and human, would be a tremendous resource for our community as a whole. The careful generation of such datasets in combination with user-friendly online tools provide a valuable resource for researchers, and could in the long run also help to reduce animal experimentation. Certain features will enhance the user experience and promote the wide use of such tools, including the ability to export high resolution graphs (ideally allowing further customization, e.g. PDF format as offered by [15, 55]) and the ability to easily download specific sequences or genome coordinates (as offered by [55, 65]). Given the challenges associated with keeping these databases up to date and operating smoothly, international and consortium efforts that provide sufficient support infrastructure may, in the long term, prove to be essential in this regard.
Here we have shown how the combined use of different online tools can be applied to generate novel hypotheses. Of course, the same tools can also be used to complement existing projects by providing additional data. Ideally, in the not too near future, researchers will have a broad compendium of resources available to them that are of such high quality that they will allow in vivo analyses to be performed in silico, thereby bringing such genome-wide analyses within reach of all scientists. This will only be possible, however, if sufficient tissue-specific datasets can be accessed. Especially in the case of the mammary gland, great care should be taken to include different timepoints to cover both embryonic and postnatal developmental stages, as well as the entire gestational cycle. Here, biological and computational expertise will continually need to go hand in hand to ensure that such online tools can meet the demands of the scientific questions that are being asked.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Gynäkologie

Kombi-Abonnement

Mit e.Med Gynäkologie erhalten Sie Zugang zu CME-Fortbildungen der beiden Fachgebiete, den Premium-Inhalten der Fachzeitschriften, inklusive einer gedruckten gynäkologischen oder urologischen Zeitschrift Ihrer Wahl.

Literatur
3.
Zurück zum Zitat Huguet EL, McMahon JA, McMahon AP, Bicknell R, Harris AL. Differential Expression of Human Wnt Genes 2, 3, 4, and 7B in Human Breast Cell Lines and Normal and Disease States of Human Breast Tissue. Cancer Res. 1994;54:2615–21.PubMed Huguet EL, McMahon JA, McMahon AP, Bicknell R, Harris AL. Differential Expression of Human Wnt Genes 2, 3, 4, and 7B in Human Breast Cell Lines and Normal and Disease States of Human Breast Tissue. Cancer Res. 1994;54:2615–21.PubMed
4.
Zurück zum Zitat Milovanovic T, Planutis K, Nguyen A, Marsh JL, Lin F, Hope C. Expression of Wnt genes and frizzled 1 and 2 receptors in normal breast epithelium and infiltrating breast carcinoma. Int J Oncol. 2004;25:1337–42.PubMed Milovanovic T, Planutis K, Nguyen A, Marsh JL, Lin F, Hope C. Expression of Wnt genes and frizzled 1 and 2 receptors in normal breast epithelium and infiltrating breast carcinoma. Int J Oncol. 2004;25:1337–42.PubMed
5.
Zurück zum Zitat Chen J, Liu T-Y, Peng H-T, Wu Y-Q, Zhang L-L, Lin X-H. Up-regulation of Wnt7b rather than Wnt1, Wnt7a, and Wnt9a indicates poor prognosis in breast cancer. Int J Clin Exp Pathol. 2018;11:4552–61.PubMedPubMedCentral Chen J, Liu T-Y, Peng H-T, Wu Y-Q, Zhang L-L, Lin X-H. Up-regulation of Wnt7b rather than Wnt1, Wnt7a, and Wnt9a indicates poor prognosis in breast cancer. Int J Clin Exp Pathol. 2018;11:4552–61.PubMedPubMedCentral
6.
Zurück zum Zitat Yeo EJ, Cassetta L, Qian BZ, Lewkowich I, Li JF, Stefater JA. Myeloid wnt7b mediates the angiogenic switch and metastasis in breast cancer. Cancer Res. 2014;74:2962–73.PubMedPubMedCentralCrossRef Yeo EJ, Cassetta L, Qian BZ, Lewkowich I, Li JF, Stefater JA. Myeloid wnt7b mediates the angiogenic switch and metastasis in breast cancer. Cancer Res. 2014;74:2962–73.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Kouros-Mehr H, Werb Z. Candidate regulators of mammary branching morphogenesis identified by genome-wide transcript analysis. Dev Dyn. 2006;235:3404–12.PubMedPubMedCentralCrossRef Kouros-Mehr H, Werb Z. Candidate regulators of mammary branching morphogenesis identified by genome-wide transcript analysis. Dev Dyn. 2006;235:3404–12.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Wong GT, Gavin BJ, McMahon AP. Differential transformation of mammary epithelial cells by Wnt genes. Mol Cell Biol. 1994;14:6278–86.PubMedPubMedCentral Wong GT, Gavin BJ, McMahon AP. Differential transformation of mammary epithelial cells by Wnt genes. Mol Cell Biol. 1994;14:6278–86.PubMedPubMedCentral
10.
Zurück zum Zitat Naylor S, Smalley MJ, Robertson D, Gusterson BA, Edwards PAW, Dale TC. Retroviral expression of Wnt-1 and Wnt-7b produces different effects in mouse mammary epithelium. J Cell Sci [Internet]. 2000 [cited 2020 Jul 7];113:2129–38. Naylor S, Smalley MJ, Robertson D, Gusterson BA, Edwards PAW, Dale TC. Retroviral expression of Wnt-1 and Wnt-7b produces different effects in mouse mammary epithelium. J Cell Sci [Internet]. 2000 [cited 2020 Jul 7];113:2129–38.
12.
Zurück zum Zitat Shimizu H, Julius MA, Giarré M, Zheng Z, Brown AMC, Kitajewski J. Transformation by wnt family proteins correlates with regulation of β- catenin. Cell Growth Differ. 1997;8:1349–58.PubMed Shimizu H, Julius MA, Giarré M, Zheng Z, Brown AMC, Kitajewski J. Transformation by wnt family proteins correlates with regulation of β- catenin. Cell Growth Differ. 1997;8:1349–58.PubMed
13.
Zurück zum Zitat Cai C, Yu QC, Jiang W, Liu W, Song W, Yu H. R-spondin1 is a novel hormone mediator for mammary stem cell self-renewal. Genes Dev. 2014;28:2205–18.PubMedPubMedCentralCrossRef Cai C, Yu QC, Jiang W, Liu W, Song W, Yu H. R-spondin1 is a novel hormone mediator for mammary stem cell self-renewal. Genes Dev. 2014;28:2205–18.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Schaum N, Karkanias J, Neff NF, May AP, Quake SR, Wyss-Coray T. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature. 2018;562:367–72.PubMedCentralCrossRef Schaum N, Karkanias J, Neff NF, May AP, Quake SR, Wyss-Coray T. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature. 2018;562:367–72.PubMedCentralCrossRef
17.
Zurück zum Zitat Giraddi RR, Chung C-Y, Heinz RE, Perou CM, Wahl GM, Spike BT. Single-Cell Transcriptomes Distinguish Stem Cell State Changes and Lineage Specification Programs in Early Mammary Gland Development. CellReports [Internet]. 2018 [cited 2020 Jul 15];24:1653–1666.e7. Available from: https://doi.org/10.1016/j.celrep.2018.07.025 Giraddi RR, Chung C-Y, Heinz RE, Perou CM, Wahl GM, Spike BT. Single-Cell Transcriptomes Distinguish Stem Cell State Changes and Lineage Specification Programs in Early Mammary Gland Development. CellReports [Internet]. 2018 [cited 2020 Jul 15];24:1653–1666.e7. Available from: https://​doi.​org/​10.​1016/​j.​celrep.​2018.​07.​025
18.
Zurück zum Zitat Han X, Wang R, Zhou Y, Fei L, Sun H, Lai S, Saadatpour A, Zhou Z, Chen H, Ye F, Huang D, Xu Y, Huang W, Jiang M, Jiang X, Mao J, Chen Y, Lu C, Xie J, Fang Q, Wang Y, Yue R, Li T, Huang H, Orkin SH, Yuan G-C, Chen M, Guo G. Mapping the Mouse Cell Atlas by Microwell-Seq. Cell. 2018;172(5):1091-1107.e17 Han X, Wang R, Zhou Y, Fei L, Sun H, Lai S, Saadatpour A, Zhou Z, Chen H, Ye F, Huang D, Xu Y, Huang W, Jiang M, Jiang X, Mao J, Chen Y, Lu C, Xie J, Fang Q, Wang Y, Yue R, Li T, Huang H, Orkin SH, Yuan G-C, Chen M, Guo G. Mapping the Mouse Cell Atlas by Microwell-Seq. Cell. 2018;172(5):1091-1107.e17
19.
Zurück zum Zitat Khan A, Zhang X. DbSUPER: A database of Super-enhancers in mouse and human genome. Nucleic Acids Res [Internet]. 2016 [cited 2020 Jul 25];44:D164–71. Khan A, Zhang X. DbSUPER: A database of Super-enhancers in mouse and human genome. Nucleic Acids Res [Internet]. 2016 [cited 2020 Jul 25];44:D164–71.
21.
Zurück zum Zitat Almanzar N, Antony J, Baghel AS, Bakerman I, Bansal I, Barres BA. A single-cell transcriptomic atlas characterizes ageing tissues in the mouse. Nature. 2020;583:590–5.CrossRef Almanzar N, Antony J, Baghel AS, Bakerman I, Bansal I, Barres BA. A single-cell transcriptomic atlas characterizes ageing tissues in the mouse. Nature. 2020;583:590–5.CrossRef
22.
Zurück zum Zitat Schaum N, Lehallier B, Hahn O, Hosseinzadeh S, Lee SE, Sit R, et al. The murine transcriptome reveals global aging nodes with organ-specific phase and amplitude. bioRxiv. 2019; Schaum N, Lehallier B, Hahn O, Hosseinzadeh S, Lee SE, Sit R, et al. The murine transcriptome reveals global aging nodes with organ-specific phase and amplitude. bioRxiv. 2019;
24.
Zurück zum Zitat Jung I, Schmitt A, Diao Y, Lee AJ, Liu T, Yang D. A compendium of promoter-centered long-range chromatin interactions in the human genome. Nat Genet. 2019;51:1442–9.PubMedPubMedCentralCrossRef Jung I, Schmitt A, Diao Y, Lee AJ, Liu T, Yang D. A compendium of promoter-centered long-range chromatin interactions in the human genome. Nat Genet. 2019;51:1442–9.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Papatheodorou I, Moreno P, Manning J, Fuentes AMP, George N, Fexova S. Expression Atlas update: From tissues to single cells. Nucleic Acids Res. 2020;48:D77-83.PubMed Papatheodorou I, Moreno P, Manning J, Fuentes AMP, George N, Fexova S. Expression Atlas update: From tissues to single cells. Nucleic Acids Res. 2020;48:D77-83.PubMed
26.
Zurück zum Zitat Wang J, Dai X, Berry LD, Cogan JD, Liu Q, Shyr Y. HACER: An atlas of human active enhancers to interpret regulatory variants. Nucleic Acids Res. 2019;47:D106-12.PubMedCrossRef Wang J, Dai X, Berry LD, Cogan JD, Liu Q, Shyr Y. HACER: An atlas of human active enhancers to interpret regulatory variants. Nucleic Acids Res. 2019;47:D106-12.PubMedCrossRef
27.
Zurück zum Zitat Fan Z, Chen R, Chen X. SpatialDB: A database for spatially resolved transcriptomes. Nucleic Acids Res. 2020;48:D233-7.PubMed Fan Z, Chen R, Chen X. SpatialDB: A database for spatially resolved transcriptomes. Nucleic Acids Res. 2020;48:D233-7.PubMed
29.
Zurück zum Zitat Gao T, He B, Liu S, Zhu H, Tan K, Qian J. EnhancerAtlas: A resource for enhancer annotation and analysis in 105 human cell/tissue types. Bioinformatics. 2016;32:3543–51.PubMedPubMedCentral Gao T, He B, Liu S, Zhu H, Tan K, Qian J. EnhancerAtlas: A resource for enhancer annotation and analysis in 105 human cell/tissue types. Bioinformatics. 2016;32:3543–51.PubMedPubMedCentral
30.
Zurück zum Zitat Gao T, Qian J. EnhancerAtlas 2.0: An updated resource with enhancer annotation in 586 tissue/cell types across nine species. Nucleic Acids Res. 2020;48:D58–64. Gao T, Qian J. EnhancerAtlas 2.0: An updated resource with enhancer annotation in 586 tissue/cell types across nine species. Nucleic Acids Res. 2020;48:D58–64.
31.
Zurück zum Zitat Cardoso-Moreira M, Halbert J, Valloton D, Velten B, Chen C, Shao Y. Gene expression across mammalian organ development. Nature. 2019;571:505–9.PubMedPubMedCentralCrossRef Cardoso-Moreira M, Halbert J, Valloton D, Velten B, Chen C, Shao Y. Gene expression across mammalian organ development. Nature. 2019;571:505–9.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Sarropoulos I, Marin R, Cardoso-Moreira M, Kaessmann H. Developmental dynamics of lncRNAs across mammalian organs and species. Nature. 2019;571:510–4.PubMedPubMedCentralCrossRef Sarropoulos I, Marin R, Cardoso-Moreira M, Kaessmann H. Developmental dynamics of lncRNAs across mammalian organs and species. Nature. 2019;571:510–4.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Mei S, Qin Q, Wu Q, Sun H, Zheng R, Zang C. Cistrome Data Browser: A data portal for ChIP-Seq and chromatin accessibility data in human and mouse. Nucleic Acids Res. 2017;45:D658-62.PubMedCrossRef Mei S, Qin Q, Wu Q, Sun H, Zheng R, Zang C. Cistrome Data Browser: A data portal for ChIP-Seq and chromatin accessibility data in human and mouse. Nucleic Acids Res. 2017;45:D658-62.PubMedCrossRef
35.
Zurück zum Zitat Zheng R, Wan C, Mei S, Qin Q, Wu Q, Sun H. Cistrome Data Browser: Expanded datasets and new tools for gene regulatory analysis. Nucleic Acids Res. 2019;47:D729-35.PubMedCrossRef Zheng R, Wan C, Mei S, Qin Q, Wu Q, Sun H. Cistrome Data Browser: Expanded datasets and new tools for gene regulatory analysis. Nucleic Acids Res. 2019;47:D729-35.PubMedCrossRef
36.
Zurück zum Zitat Han X, Zhou Z, Fei L, Sun H, Wang R, Chen Y. Construction of a human cell landscape at single-cell level. Nature. 2020;581:303–9.PubMedCrossRef Han X, Zhou Z, Fei L, Sun H, Wang R, Chen Y. Construction of a human cell landscape at single-cell level. Nature. 2020;581:303–9.PubMedCrossRef
38.
Zurück zum Zitat Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA. The cBio Cancer Genomics Portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2:401–4.PubMedCrossRef Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA. The cBio Cancer Genomics Portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2:401–4.PubMedCrossRef
41.
Zurück zum Zitat Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z. Enrichr: A comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 2016;44:W90-7.PubMedPubMedCentralCrossRef Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z. Enrichr: A comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 2016;44:W90-7.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Ramos J, Das J, Felty Q, Yoo C, Poppiti R, Murrell D. NRF1 motif sequence-enriched genes involved in ER/PR −ve HER2 +ve breast cancer signaling pathways Breast Cancer. Res Treat. 2018;172:469–85. Ramos J, Das J, Felty Q, Yoo C, Poppiti R, Murrell D. NRF1 motif sequence-enriched genes involved in ER/PR −ve HER2 +ve breast cancer signaling pathways Breast Cancer. Res Treat. 2018;172:469–85.
44.
Zurück zum Zitat Fernandez-Valdivia R, Mukherjee A, Creighton CJ, Buser AC, DeMayo FJ, Edwards DP. Transcriptional response of the murine mammary gland to acute progesterone exposure. Endocrinology. 2008;149:6236–50.PubMedPubMedCentralCrossRef Fernandez-Valdivia R, Mukherjee A, Creighton CJ, Buser AC, DeMayo FJ, Edwards DP. Transcriptional response of the murine mammary gland to acute progesterone exposure. Endocrinology. 2008;149:6236–50.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Shu W, Jiang YQ, Lu MM, Morrisey EE. Wnt7b regulates mesenchymal proliferation and vascular development in the lung. Development. 2002;129:4831–42.PubMedCrossRef Shu W, Jiang YQ, Lu MM, Morrisey EE. Wnt7b regulates mesenchymal proliferation and vascular development in the lung. Development. 2002;129:4831–42.PubMedCrossRef
46.
Zurück zum Zitat Rajagopal J, Carroll TJ, Guseh JS, Bores SA, Blank LJ, Anderson WJ. Wnt7b stimulates embryonic lung growth by coordinately increasing the replication of epithelium and mesenchyme. Development. 2008;135:1625–34.PubMedCrossRef Rajagopal J, Carroll TJ, Guseh JS, Bores SA, Blank LJ, Anderson WJ. Wnt7b stimulates embryonic lung growth by coordinately increasing the replication of epithelium and mesenchyme. Development. 2008;135:1625–34.PubMedCrossRef
47.
Zurück zum Zitat Yu J, Carroll TJ, Rajagopal J, Kobayashi A, Ren Q, McMahon AP. A Wnt7b-dependent pathway regulates the orientation of epithelial cell division and establishes the cortico-medullary axis of the mammalian kidney. Development. 2009;136:161–71.PubMedCrossRef Yu J, Carroll TJ, Rajagopal J, Kobayashi A, Ren Q, McMahon AP. A Wnt7b-dependent pathway regulates the orientation of epithelial cell division and establishes the cortico-medullary axis of the mammalian kidney. Development. 2009;136:161–71.PubMedCrossRef
49.
Zurück zum Zitat Dowen JM, Fan ZP, Hnisz D, Ren G, Abraham BJ, Zhang LN, et al. Control of cell identity genes occurs in insulated neighborhoods in mammalian chromosomes. Cell [Internet]. 2014;159:374–87. Dowen JM, Fan ZP, Hnisz D, Ren G, Abraham BJ, Zhang LN, et al. Control of cell identity genes occurs in insulated neighborhoods in mammalian chromosomes. Cell [Internet]. 2014;159:374–87.
50.
Zurück zum Zitat Novo CL, Javierre BM, Cairns J, Segonds-Pichon A, Wingett SW, Freire-Pritchett P. Long-Range Enhancer Interactions Are Prevalent in Mouse Embryonic Stem Cells and Are Reorganized upon Pluripotent State Transition. Cell Rep. 2018;22:2615–27.PubMedPubMedCentralCrossRef Novo CL, Javierre BM, Cairns J, Segonds-Pichon A, Wingett SW, Freire-Pritchett P. Long-Range Enhancer Interactions Are Prevalent in Mouse Embryonic Stem Cells and Are Reorganized upon Pluripotent State Transition. Cell Rep. 2018;22:2615–27.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Whyte WA, Orlando DA, Hnisz D, Abraham BJ, Lin CY, Kagey MH. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell. 2013;153:307–19.PubMedPubMedCentralCrossRef Whyte WA, Orlando DA, Hnisz D, Abraham BJ, Lin CY, Kagey MH. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell. 2013;153:307–19.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Pott S, Lieb JD. What are super-enhancers? Nat. Genet. 2015. p. 8–12. Pott S, Lieb JD. What are super-enhancers? Nat. Genet. 2015. p. 8–12.
53.
Zurück zum Zitat Adam RC, Yang H, Rockowitz S, Larsen SB, Nikolova M, Oristian DS. Pioneer factors govern super-enhancer dynamics in stem cell plasticity and lineage choice. Nature. 2015;521:366–70.PubMedPubMedCentralCrossRef Adam RC, Yang H, Rockowitz S, Larsen SB, Nikolova M, Oristian DS. Pioneer factors govern super-enhancer dynamics in stem cell plasticity and lineage choice. Nature. 2015;521:366–70.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Wang Y, Song F, Zhang B, Zhang L, Xu J, Kuang D, et al. The 3D Genome Browser: A web-based browser for visualizing 3D genome organization and long-range chromatin interactions. Genome Biol Genome Biology. 2018;19:1–12. Wang Y, Song F, Zhang B, Zhang L, Xu J, Kuang D, et al. The 3D Genome Browser: A web-based browser for visualizing 3D genome organization and long-range chromatin interactions. Genome Biol Genome Biology. 2018;19:1–12.
56.
Zurück zum Zitat Dixon JR, Selvaraj S, Yue F, Kim A, Li Y, Shen Y, et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature Nature Publishing Group. 2012;485:376–80. Dixon JR, Selvaraj S, Yue F, Kim A, Li Y, Shen Y, et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature Nature Publishing Group. 2012;485:376–80.
57.
Zurück zum Zitat Rao SSP, Huntley MH, Durand NC, Stamenova EK, Bochkov ID, Robinson JT, et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell. Elsevier Inc.; 2014;159:1665–80. Rao SSP, Huntley MH, Durand NC, Stamenova EK, Bochkov ID, Robinson JT, et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell. Elsevier Inc.; 2014;159:1665–80.
58.
Zurück zum Zitat Bonev B, Mendelson Cohen N, Szabo Q, Fritsch L, Papadopoulos GL, Lubling Y. Multiscale 3D Genome Rewiring during Mouse Neural Development. Cell. 2017;171:557–72.PubMedPubMedCentralCrossRef Bonev B, Mendelson Cohen N, Szabo Q, Fritsch L, Papadopoulos GL, Lubling Y. Multiscale 3D Genome Rewiring during Mouse Neural Development. Cell. 2017;171:557–72.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat McArthur E, Capra J. Topologically associating domain (TAD) boundaries stable across diverse cell types are evolutionarily constrained and enriched for heritability. bioRxiv. 2020; McArthur E, Capra J. Topologically associating domain (TAD) boundaries stable across diverse cell types are evolutionarily constrained and enriched for heritability. bioRxiv. 2020;
62.
Zurück zum Zitat Chung CY, Ma Z, Dravis C, Preissl S, Poirion O, Luna G. Single-Cell Chromatin Analysis of Mammary Gland Development Reveals Cell-State Transcriptional Regulators and Lineage Relationships. Cell Rep. 2019;29:495–510.PubMedPubMedCentralCrossRef Chung CY, Ma Z, Dravis C, Preissl S, Poirion O, Luna G. Single-Cell Chromatin Analysis of Mammary Gland Development Reveals Cell-State Transcriptional Regulators and Lineage Relationships. Cell Rep. 2019;29:495–510.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Pliner HA, Packer JS, McFaline-Figueroa JL, Cusanovich DA, Daza RM, Aghamirzaie D. Cicero Predicts cis-Regulatory DNA Interactions from Single-Cell Chromatin Accessibility Data. Mol Cell. 2018;71:858–71.PubMedPubMedCentralCrossRef Pliner HA, Packer JS, McFaline-Figueroa JL, Cusanovich DA, Daza RM, Aghamirzaie D. Cicero Predicts cis-Regulatory DNA Interactions from Single-Cell Chromatin Accessibility Data. Mol Cell. 2018;71:858–71.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat James Kent W, Sugnet CW, Furey TS, Roskin KM, Pringle TH, Zahler AM. The human genome browser at UCSC. Genome Res. 2002;12:996–1006.PubMedCrossRef James Kent W, Sugnet CW, Furey TS, Roskin KM, Pringle TH, Zahler AM. The human genome browser at UCSC. Genome Res. 2002;12:996–1006.PubMedCrossRef
67.
Zurück zum Zitat Ahituv N, Prabhakar S, Poulin F, Rubin EM, Couronne O. Mapping cis-regulatory domains in the human genome using multi-species conservation of synteny. Hum Mol Genet. 2005;14:3057–63.PubMedCrossRef Ahituv N, Prabhakar S, Poulin F, Rubin EM, Couronne O. Mapping cis-regulatory domains in the human genome using multi-species conservation of synteny. Hum Mol Genet. 2005;14:3057–63.PubMedCrossRef
68.
Zurück zum Zitat Boffelli D, Nobrega MA, Rubin EM. Comparative genomics at the vertebrate extremes. Nat Rev Genet. 2004;5:456–65.PubMedCrossRef Boffelli D, Nobrega MA, Rubin EM. Comparative genomics at the vertebrate extremes. Nat Rev Genet. 2004;5:456–65.PubMedCrossRef
70.
Zurück zum Zitat Bejerano G, Pheasant M, Makunin I, Stephen S, Kent WJ, Mattick JS, et al. Ultraconserved elements in the human genome. Science (80-). 2004;304:1321–5. Bejerano G, Pheasant M, Makunin I, Stephen S, Kent WJ, Mattick JS, et al. Ultraconserved elements in the human genome. Science (80-). 2004;304:1321–5.
71.
Zurück zum Zitat Pennacchio LA, Ahituv N, Moses AM, Prabhakar S, Nobrega MA, Shoukry M. In vivo enhancer analysis of human conserved non-coding sequences. Nature. 2006;444:499–502.PubMedCrossRef Pennacchio LA, Ahituv N, Moses AM, Prabhakar S, Nobrega MA, Shoukry M. In vivo enhancer analysis of human conserved non-coding sequences. Nature. 2006;444:499–502.PubMedCrossRef
72.
Zurück zum Zitat Chen J, Sun M, Hurst LD, Carmichael GG, Rowley JD. Human antisense genes have unusually short introns: Evidence for selection for rapid transcription. Trends Genet. 2005;21:203–7.PubMedCrossRef Chen J, Sun M, Hurst LD, Carmichael GG, Rowley JD. Human antisense genes have unusually short introns: Evidence for selection for rapid transcription. Trends Genet. 2005;21:203–7.PubMedCrossRef
74.
76.
Zurück zum Zitat Wend P, Holland JD, Ziebold U, Birchmeier W. Wnt signaling in stem and cancer stem cells. Semin Cell Dev Biol. 2010;21:855–63.PubMedCrossRef Wend P, Holland JD, Ziebold U, Birchmeier W. Wnt signaling in stem and cancer stem cells. Semin Cell Dev Biol. 2010;21:855–63.PubMedCrossRef
77.
Zurück zum Zitat Incassati A, Chandramouli A, Eelkema R, Cowin P. Key signaling nodes in mammary gland development and cancer: β-catenin. Breast Cancer Res. 2010;2:1–14. Incassati A, Chandramouli A, Eelkema R, Cowin P. Key signaling nodes in mammary gland development and cancer: β-catenin. Breast Cancer Res. 2010;2:1–14.
Metadaten
Titel
How to Use Online Tools to Generate New Hypotheses for Mammary Gland Biology Research: A Case Study for Wnt7b
verfasst von
Yorick Bernardus Cornelis van de Grift
Nika Heijmans
Renée van Amerongen
Publikationsdatum
24.02.2021
Verlag
Springer US
Erschienen in
Journal of Mammary Gland Biology and Neoplasia / Ausgabe 4/2020
Print ISSN: 1083-3021
Elektronische ISSN: 1573-7039
DOI
https://doi.org/10.1007/s10911-020-09474-z

Weitere Artikel der Ausgabe 4/2020

Journal of Mammary Gland Biology and Neoplasia 4/2020 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.