Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
NAR Genom Bioinform ; 4(3): lqac072, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36159174

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a cellular process involved in development and disease progression. Intermediate EMT states were observed in tumors and fibrotic tissues, but previous in vitro studies focused on time-dependent responses with single doses of signals; it was unclear whether single-cell transcriptomes support stable intermediates observed in diseases. Here, we performed single-cell RNA-sequencing with human mammary epithelial cells treated with multiple doses of TGF-ß. We found that dose-dependent EMT harbors multiple intermediate states at nearly steady state. Comparisons of dose- and time-dependent EMT transcriptomes revealed that the dose-dependent data enable higher sensitivity to detect genes associated with EMT. We identified cell clusters unique to time-dependent EMT, reflecting cells en route to stable states. Combining dose- and time-dependent cell clusters gave rise to accurate prognosis for cancer patients. Our transcriptomic data and analyses uncover a stable EMT continuum at the single-cell resolution, and complementary information of two types of single-cell experiments.

2.
Med Image Anal ; 79: 102436, 2022 07.
Article in English | MEDLINE | ID: mdl-35405571

ABSTRACT

Cell detection is an important task in biomedical research. Recently, deep learning methods have made it possible to improve the performance of cell detection. However, a detection network trained with training data under a specific condition (source domain) may not work well on data under other conditions (target domains), which is called the domain shift problem. In particular, cells are cultured under different conditions depending on the purpose of the research. Characteristics, e.g., the shapes and density of the cells, change depending on the conditions, and such changes may cause domain shift problems. Here, we propose an unsupervised domain adaptation method for cell detection using a pseudo-cell-position heatmap, where the cell centroid is at the peak of a Gaussian distribution in the map and selective pseudo-labeling. In the prediction result for the target domain, even if the peak location is correct, the signal distribution around the peak often has a non-Gaussian shape. The pseudo-cell-position heatmap is thus re-generated using the peak positions in the predicted heatmap to have a clear Gaussian shape. Our method selects confident pseudo-cell-position heatmaps based on uncertainty and curriculum learning. We conducted numerous experiments showing that, compared with the existing methods, our method improved detection performance under different conditions.


Subject(s)
Normal Distribution , Humans
3.
iScience ; 25(2): 103777, 2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35146392

ABSTRACT

The analysis and interpretation of single-cell RNA sequencing (scRNA-seq) experiments are compromised by the presence of poor-quality cells. For meaningful analyses, such poor-quality cells should be excluded as they introduce noise in the data. We introduce SkewC, a quality-assessment tool, to identify skewed cells in scRNA-seq experiments. The tool's methodology is based on the assessment of gene coverage for each cell, and its skewness as a quality measure; the gene body coverage is a unique characteristic for each protocol, and different protocols yield highly different coverage profiles. This tool is designed to avoid misclustering or false clusters by identifying, isolating, and removing cells with skewed gene body coverage profiles. SkewC is capable of processing any type of scRNA-seq dataset, regardless of the protocol. We envision SkewC as a distinctive QC method to be incorporated into scRNA-seq QC processing to preclude the possibility of scRNA-seq data misinterpretation.

4.
Cell Rep ; 38(2): 110240, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35021086

ABSTRACT

Maintenance of undifferentiated, long-lived, and often quiescent stem cells in the basal compartment is important for homeostasis and regeneration of multiple epithelial tissues, but the molecular mechanisms that coordinately control basal cell fate and stem cell quiescence are elusive. Here, we report an epithelium-intrinsic requirement for Zeb1, a core transcriptional inducer of epithelial-to-mesenchymal transition, for mammary epithelial ductal side branching and for basal cell regenerative capacity. Our findings uncover an evolutionarily conserved role of Zeb1 in promoting basal cell fate over luminal differentiation. We show that Zeb1 loss results in increased basal cell proliferation at the expense of quiescence and self-renewal. Moreover, Zeb1 cooperates with YAP to activate Axin2 expression, and inhibition of Wnt signaling partially restores stem cell function to Zeb1-deficient basal cells. Thus, Zeb1 is a transcriptional regulator that maintains both basal cell fate and stem cell quiescence, and it functions in part through suppressing Wnt signaling.


Subject(s)
Cell Lineage/genetics , Stem Cells/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism , 3T3 Cells , Animals , Axin Protein/metabolism , Cell Differentiation , Cell Proliferation , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Transcription Factors , Wnt Signaling Pathway/physiology , Zinc Finger E-box-Binding Homeobox 1/genetics
5.
J Mammary Gland Biol Neoplasia ; 26(4): 357-365, 2021 12.
Article in English | MEDLINE | ID: mdl-34932179

ABSTRACT

Mammary gland is an outstanding system to study the regulatory mechanisms governing adult epithelial stem cell activity. Stem cells in the basal layer of the mammary gland fuel the morphogenesis and regeneration of a complex epithelial network during development and upon transplantation. The self-renewal of basal stem/progenitor cells is subjected to regulation by both cell-intrinsic and extrinsic mechanisms. Nfatc1 is a transcription factor that regulates breast tumorigenesis and metastasis, but its role in mammary epithelial development and stem cell function has not been investigated. Here we show that Nfatc1 is expressed in a small subset of mammary basal epithelial cells and its epithelial-specific deletion results in mild defects in side branching and basal-luminal cell balance. Moreover, Nfatc1-deficient basal cells exhibit reduced colony forming ability in vitro and somewhat compromised regenerative potential upon transplantation. Thus, our study provides evidence for a detectable yet non-essential role of Nfatc1 in mammary epithelial morphogenesis and basal stem/progenitor cell self-renewal.


Subject(s)
Mammary Glands, Animal , Stem Cells , Animals , Cell Differentiation/physiology , Epithelial Cells/pathology , Morphogenesis , Stem Cells/physiology , Transcription Factors
6.
Front Genet ; 12: 719099, 2021.
Article in English | MEDLINE | ID: mdl-34490045

ABSTRACT

Large-scale transcriptome data, such as single-cell RNA-sequencing data, have provided unprecedented resources for studying biological processes at the systems level. Numerous dimensionality reduction methods have been developed to visualize and analyze these transcriptome data. In addition, several existing methods allow inference of functional variations among samples using gene sets with known biological functions. However, it remains challenging to analyze transcriptomes with reduced dimensions that are interpretable in terms of dimensions' directionalities, transferrable to new data, and directly expose the contribution or association of individual genes. In this study, we used gene set non-negative principal component analysis (gsPCA) and non-negative matrix factorization (gsNMF) to analyze large-scale transcriptome datasets. We found that these methods provide low-dimensional information about the progression of biological processes in a quantitative manner, and their performances are comparable to existing functional variation analysis methods in terms of distinguishing multiple cell states and samples from multiple conditions. Remarkably, upon training with a subset of data, these methods allow predictions of locations in the functional space using data from experimental conditions that are not exposed to the models. Specifically, our models predicted the extent of progression and reversion for cells in the epithelial-mesenchymal transition (EMT) continuum. These methods revealed conserved EMT program among multiple types of single cells and tumor samples. Finally, we demonstrate this approach is broadly applicable to data and gene sets beyond EMT and provide several recommendations on the choice between the two linear methods and the optimal algorithmic parameters. Our methods show that simple constrained matrix decomposition can produce to low-dimensional information in functionally interpretable and transferrable space, and can be widely useful for analyzing large-scale transcriptome data.

7.
Med Image Anal ; 73: 102182, 2021 10.
Article in English | MEDLINE | ID: mdl-34340103

ABSTRACT

Cell instance segmentation is important in biomedical research. For living cell analysis, microscopy images are captured under various conditions (e.g., the type of microscopy and type of cell). Deep-learning-based methods can be used to perform instance segmentation if sufficient annotations of individual cell boundaries are prepared as training data. Generally, annotations are required for each condition, which is very time-consuming and labor-intensive. To reduce the annotation cost, we propose a weakly supervised cell instance segmentation method that can segment individual cell regions under various conditions by only using rough cell centroid positions as training data. This method dramatically reduces the annotation cost compared with the standard annotation method of supervised segmentation. We demonstrated the efficacy of our method on various cell images; it outperformed several of the conventional weakly-supervised methods on average. In addition, we demonstrated that our method can perform instance cell segmentation without any manual annotation by using pairs of phase contrast and fluorescence images in which cell nuclei are stained as training data.


Subject(s)
Image Processing, Computer-Assisted , Microscopy , Cell Nucleus , Supervised Machine Learning
8.
Cell Rep ; 33(3): 108273, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33086071

ABSTRACT

The mammary epithelial cell (MEC) system is a bilayered ductal epithelium of luminal and basal cells, maintained by a lineage of stem and progenitor populations. Here, we used integrated single-cell transcriptomics and chromatin accessibility analysis to reconstruct the cell types of the mouse MEC system and their underlying gene regulatory features in an unbiased manner. We define differentiation states within the secretory type of luminal cells, which forms a continuous spectrum of general luminal progenitor and lactation-committed progenitor cells. By integrating single-cell transcriptomics and chromatin accessibility landscapes, we identify cis- and trans-regulatory elements that are differentially activated in the specific epithelial cell types and our newly defined luminal differentiation states. Our work provides a resource to reveal cis/trans-regulatory elements associated with MEC identity and differentiation that will serve as a reference to determine how the chromatin accessibility landscape changes during breast cancer.


Subject(s)
Chromatin/genetics , Epithelial Cells/metabolism , Animals , Base Sequence , Cell Differentiation/genetics , Cell Lineage , Cell Proliferation/genetics , Chromatin/physiology , Computational Biology/methods , Epithelial Cells/physiology , Epithelium/metabolism , Female , Gene Expression Profiling/methods , Gene Expression Regulation , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Regulatory Sequences, Nucleic Acid , Single-Cell Analysis/methods , Stem Cells/metabolism , Transcriptome
9.
FEBS Lett ; 594(10): 1532-1549, 2020 05.
Article in English | MEDLINE | ID: mdl-32017069

ABSTRACT

Activation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) transcription factor, a central player in immune response regulation, is based on phosphorylation of inhibitor of kappaB alpha (IκBα) by the Inhibitor of kappaB kinase (IKK) that triggers IκBα degradation. Although inhibitor of kappaB beta (IκBß) is structurally similar to IκBα, its precise characteristics remain undefined. Herein, we report that the molecular interactivity of IκBß with the kinase-active region of IKK subunit 2 (IKK2), as well as its phosphorylation status, differs markedly from those of IκBα. A mass spectrometry analysis revealed that IκBß phosphorylation sites are distributed in its C-terminal region, whereas IκBα phosphorylation sites are located in the N-terminal region. Furthermore, IKK2 phosphorylation sites in IκBß are found in a region distinct from typical degradation signals, such as phosphodegron and proline/glutamic acid/serine/threonine-rich sequence (PEST) motifs. Mutation of the IκBß phosphorylation sites enhances its resistance to homeostatic proteasomal degradation. These findings contribute a novel concept in NF-κB/IKK signalling research.


Subject(s)
Biocatalysis , I-kappa B Kinase/metabolism , I-kappa B Proteins/chemistry , I-kappa B Proteins/metabolism , NF-KappaB Inhibitor alpha/chemistry , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Amino Acid Motifs , Homeostasis , Humans , Models, Molecular , Mutation , Phosphorylation/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , Signal Transduction
10.
NPJ Syst Biol Appl ; 5: 21, 2019.
Article in English | MEDLINE | ID: mdl-31275609

ABSTRACT

Epithelial-to-mesenchymal transition (EMT), a fundamental transdifferentiation process in development, produces diverse phenotypes in different physiological or pathological conditions. Many genes involved in EMT have been identified to date, but mechanisms contributing to the phenotypic diversity and those governing the coupling between the dynamics of epithelial (E) genes and that of the mesenchymal (M) genes are unclear. In this study, we employed combinatorial perturbations to mammary epithelial cells to induce a series of EMT phenotypes by manipulating two essential EMT-inducing elements, namely TGF-ß and ZEB1. By measuring transcriptional changes in more than 700 E-genes and M-genes, we discovered that the M-genes exhibit a significant diversity in their dependency to these regulatory elements and identified three groups of M-genes that are controlled by different regulatory circuits. Notably, functional differences were detected among the M-gene clusters in motility regulation and in survival of breast cancer patients. We computationally predicted and experimentally confirmed that the reciprocity and reversibility of EMT are jointly regulated by ZEB1. Our integrative analysis reveals the key roles of ZEB1 in coordinating the dynamics of a large number of genes during EMT, and it provides new insights into the mechanisms for the diversity of EMT phenotypes.


Subject(s)
Computational Biology/methods , Epithelial-Mesenchymal Transition/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Mesenchymal Stem Cells/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
11.
Sci Rep ; 9(1): 6490, 2019 04 24.
Article in English | MEDLINE | ID: mdl-31019211

ABSTRACT

Mesenchymal-to-epithelial transition (MET) is an important step in cell reprogramming from fibroblasts (a cell type frequently used for this purpose) to various epithelial cell types. However, the mechanism underlying MET induction in fibroblasts remains to be understood. The present study aimed to identify the transcription factors (TFs) that efficiently induce MET in dermal fibroblasts. OVOL2 was identified as a potent inducer of key epithelial genes, and OVOL2 cooperatively enhanced MET induced by HNF1A, TP63, and KLF4, which are known reprogramming TFs to epithelial lineages. In TP63/KLF4-induced keratinocyte-like cell-state reprogramming, OVOL2 greatly facilitated the activation of epithelial and keratinocyte-specific genes. This was accompanied by enhanced changes in chromatin accessibility across the genome. Mechanistically, motif enrichment analysis revealed that the target loci of KLF4 and TP63 become accessible upon induction of TFs, whereas the OVOL2 target loci become inaccessible. This indicates that KLF4 and TP63 positively regulate keratinocyte-associated genes whereas OVOL2 suppresses fibroblast-associated genes. The exogenous expression of OVOL2 therefore disrupts fibroblast lineage identity and facilitates fibroblast cell reprogramming into epithelial lineages cooperatively with tissue-specific reprogramming factors. Identification of OVOL2 as an MET inducer and an epithelial reprogramming enhancer in fibroblasts provides new insights into cellular reprogramming improvement for future applications.


Subject(s)
Cellular Reprogramming/genetics , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Fibroblasts/metabolism , Gene Expression , Transcription Factors/genetics , Cell Lineage/genetics , Cell Transdifferentiation/genetics , Cells, Cultured , Chromatin/genetics , Chromatin/metabolism , Dermis/cytology , Epithelial Cells/cytology , Fibroblasts/cytology , Humans , Infant, Newborn , Kruppel-Like Factor 4 , Sequence Analysis, DNA/methods , Transcription Factors/metabolism
12.
FASEB J ; 33(3): 3167-3179, 2019 03.
Article in English | MEDLINE | ID: mdl-30399323

ABSTRACT

Oleoylethanolamide (OEA), a fatty acid ethanolamide (FAE), is a lipid mediator that controls food intake and lipid metabolism. Accumulating data imply the importance of intestinal OEA in controlling satiety in addition to gastrointestinal peptide hormones. Although the biochemical pathway of FAE production has been illustrated, the enzymes responsible for the cleavage of OEA from its precursor N-acyl-phosphatidylethanolamine (NAPE) must be identified among reported candidates in the gut. In this study, we assessed the involvement of NAPE-specific phospholipase D (NAPE-PLD), which can directly release FAEs from NAPE, in intestinal OEA synthesis and lipid metabolism. Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPER-associated protein 9 (Cas9)-mediated deletion of the NAPE-PLD gene in intestinal epithelial-like Caco-2 cells reduced OEA levels, regardless of their differentiation states. Transcriptome analysis revealed that deletion of NAPE-PLD activates a transcriptional program for nutrient transportation, including lipids and lipoproteins, and inactivates cell-cycle or mitosis-related genes in Caco-2 cells. In addition, the basolateral secretion of lipoproteins was increased in NAPE-PLD-deleted cells although lipoprotein size was not affected. By contrast, cellular lipid levels were reduced in NAPE-PLD-deleted cells. Overall, these results indicate that NAPE-PLD plays important roles in OEA synthesis and fat absorption by regulating lipoprotein production in the intestinal epithelial cells.-Igarashi, M., Watanabe, K., Tsuduki, T., Kimura, I., Kubota, N. NAPE-PLD controls OEA synthesis and fat absorption by regulating lipoprotein synthesis in an in vitro model of intestinal epithelial cells.


Subject(s)
Dietary Fats/metabolism , Endocannabinoids/biosynthesis , Intestinal Mucosa/metabolism , Oleic Acids/biosynthesis , Phospholipase D/metabolism , CD36 Antigens/metabolism , Caco-2 Cells , Cell Differentiation , Gene Expression Profiling , Gene Knockout Techniques , Humans , Intestinal Absorption/genetics , Intestinal Absorption/physiology , Intestinal Mucosa/cytology , Lipid Metabolism , Lipoproteins/biosynthesis , Models, Biological , Phospholipase D/deficiency , Phospholipase D/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
14.
PLoS Comput Biol ; 11(11): e1004569, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26554584

ABSTRACT

Reversible epithelial-to-mesenchymal transition (EMT) is central to tissue development, epithelial stemness, and cancer metastasis. While many regulatory elements have been identified to induce EMT, the complex process underlying such cellular plasticity remains poorly understood. Utilizing a systems biology approach integrating modeling and experiments, we found multiple intermediate states contributing to EMT and that the robustness of the transitions is modulated by transcriptional factor Ovol2. In particular, we obtained evidence for a mutual inhibition relationship between Ovol2 and EMT inducer Zeb1, and observed that adding this regulation generates a novel four-state system consisting of two distinct intermediate phenotypes that differ in differentiation propensities and are favored in different environmental conditions. We identified epithelial cells that naturally exist in an intermediate state with bidirectional differentiation potential, and found the balance between EMT-promoting and -inhibiting factors to be critical in achieving and selecting between intermediate states. Our analysis suggests a new design principle in controlling cellular plasticity through multiple intermediate cell fates and underscores the critical involvement of Ovol2 and its associated molecular regulations.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Feedback, Physiological , Homeodomain Proteins/genetics , Transcription Factors/genetics , Animals , Cell Line , Cell Line, Tumor , Computational Biology , Homeodomain Proteins/metabolism , Humans , Mice , Transcription Factors/metabolism , Zinc Fingers
15.
PLoS Genet ; 10(7): e1004520, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25079073

ABSTRACT

Mammary gland branching morphogenesis and ductal homeostasis relies on mammary stem cell function for the maintenance of basal and luminal cell compartments. The mechanisms of transcriptional regulation of the basal cell compartment are currently unknown. We explored these mechanisms in the basal cell compartment and identified the Co-factor of LIM domains (CLIM/LDB/NLI) as a transcriptional regulator that maintains these cells. Clims act within the basal cell compartment to promote branching morphogenesis by maintaining the number and proliferative potential of basal mammary epithelial stem cells. Clim2, in a complex with LMO4, supports mammary stem cells by directly targeting the Fgfr2 promoter in basal cells to increase its expression. Strikingly, Clims also coordinate basal-specific transcriptional programs to preserve luminal cell identity. These basal-derived cues inhibit epidermis-like differentiation of the luminal cell compartment and enhance the expression of luminal cell-specific oncogenes ErbB2 and ErbB3. Consistently, basal-expressed Clims promote the initiation and progression of breast cancer in the MMTV-PyMT tumor model, and the Clim-regulated branching morphogenesis gene network is a prognostic indicator of poor breast cancer outcome in humans.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/genetics , DNA-Binding Proteins/genetics , LIM Domain Proteins/genetics , Neoplasms, Basal Cell/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Transcription Factors/genetics , Breast Neoplasms/metabolism , Carcinogenesis/genetics , Cell Differentiation/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Neoplasms, Basal Cell/metabolism , Promoter Regions, Genetic , Protein Structure, Tertiary , Receptor, ErbB-2/genetics , Stem Cells/metabolism , Stem Cells/pathology
16.
Sensors (Basel) ; 14(7): 12093-103, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-25004158

ABSTRACT

Software which corrects the dynamic error of force transducers in impact force measurements using their own output signal has been developed. The software corrects the output waveform of the transducers using the output waveform itself, estimates its uncertainty and displays the results. In the experiment, the dynamic error of three transducers of the same model are evaluated using the Levitation Mass Method (LMM), in which the impact forces applied to the transducers are accurately determined as the inertial force of the moving part of the aerostatic linear bearing. The parameters for correcting the dynamic error are determined from the results of one set of impact measurements of one transducer. Then, the validity of the obtained parameters is evaluated using the results of the other sets of measurements of all the three transducers. The uncertainties in the uncorrected force and those in the corrected force are also estimated. If manufacturers determine the correction parameters for each model using the proposed method, and provide the software with the parameters corresponding to each model, then users can obtain the waveform corrected against dynamic error and its uncertainty. The present status and the future prospects of the developed software are discussed in this paper.

17.
Proc Natl Acad Sci U S A ; 111(28): 10215-20, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24982158

ABSTRACT

Understanding the epigenetic mechanisms that control the activation of adult stem cells holds the promise of tissue and organ regeneration. Hair follicle stem cells have emerged as a prime model to study stem cell activation. Wnt/ß-catenin signaling controls multiple aspects of skin epithelial regeneration, with its excessive activity promoting the hyperactivation of hair follicle stem/progenitor cells and tumorigenesis. The contribution of chromatin factors in regulating Wnt/ß-catenin pathway function in these processes is unknown. Here, we show that chromatin effector Pygopus homolog 2 (Pygo2) produced by the epithelial cells facilitates depilation-induced hair regeneration, as well as ß-catenin-induced activation of hair follicle stem/early progenitor cells and trichofolliculoma-like skin hyperplasia. Pygo2 maximizes the expression of Wnt/ß-catenin targets, but is dispensable for ß-catenin-mediated expansion of LIM/homeobox protein Lhx2(+) cells, in the stem/early progenitor cell compartment of the hair follicle. Moreover, ß-catenin and Pygo2 converge to induce the accumulation and acetylation of tumor suppressor protein p53 upon the cell cycle entry of hair follicle early progenitor cells and in cultured keratinocytes. These findings identify Pygo2 as an important regulator of Wnt/ß-catenin function in skin epithelia and p53 activation as a prominent downstream event of ß-catenin/Pygo2 action in stem cell activation.


Subject(s)
Hair Follicle/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Stem Cells/metabolism , Wnt Signaling Pathway , Animals , Hair Follicle/pathology , Hyperplasia/genetics , Hyperplasia/metabolism , Hyperplasia/pathology , Intracellular Signaling Peptides and Proteins/genetics , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mice , Mice, Knockout , Stem Cells/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
18.
Dev Cell ; 29(1): 59-74, 2014 Apr 14.
Article in English | MEDLINE | ID: mdl-24735879

ABSTRACT

Epithelial cells possess remarkable plasticity, having the ability to become mesenchymal cells through alterations in adhesion and motility (epithelial-to-mesenchymal transition [EMT]). However, how epithelial plasticity is kept in check in epithelial cells during tissue development and regeneration remains to be fully understood. Here we show that restricting the EMT of mammary epithelial cells by transcription factor Ovol2 is required for proper morphogenesis and regeneration. Deletion of Ovol2 blocks mammary ductal morphogenesis, depletes stem and progenitor cell reservoirs, and leads epithelial cells to undergo EMT in vivo to become nonepithelial cell types. Ovol2 directly represses myriad EMT inducers, and its absence switches response to TGF-ß from growth arrest to EMT. Furthermore, forced expression of the repressor isoform of Ovol2 is able to reprogram metastatic breast cancer cells from a mesenchymal to an epithelial state. Our findings underscore the critical importance of exquisitely regulating epithelial plasticity in development and cancer.


Subject(s)
Epithelial-Mesenchymal Transition , Mammary Glands, Animal/growth & development , Morphogenesis , Regeneration , Transcription Factors/metabolism , Animals , Cellular Reprogramming , Embryonic Induction , Female , Gene Deletion , Gene Expression Regulation, Developmental , Humans , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Transcription Factors/genetics , Transforming Growth Factor beta/metabolism
19.
PLoS One ; 9(3): e92317, 2014.
Article in English | MEDLINE | ID: mdl-24651522

ABSTRACT

BACKGROUND: Deregulation of canonical Wnt/CTNNB1 (beta-catenin) pathway is one of the earliest events in the pathogenesis of colon cancer. Mutations in APC or CTNNB1 are highly frequent in colon cancer and cause aberrant stabilization of CTNNB1, which activates the transcription of Wnt target genes by binding to chromatin via the TCF/LEF transcription factors. Here we report an integrative analysis of genome-wide chromatin occupancy of CTNNB1 by chromatin immunoprecipitation coupled with high-throughput sequencing (ChIP-seq) and gene expression profiling by microarray analysis upon RNAi-mediated knockdown of CTNNB1 in colon cancer cells. RESULTS: We observed 3629 CTNNB1 binding peaks across the genome and a significant correlation between CTNNB1 binding and knockdown-induced gene expression change. Our integrative analysis led to the discovery of a direct Wnt target signature composed of 162 genes. Gene ontology analysis of this signature revealed a significant enrichment of Wnt pathway genes, suggesting multiple feedback regulations of the pathway. We provide evidence that this gene signature partially overlaps with the Lgr5+ intestinal stem cell signature, and is significantly enriched in normal intestinal stem cells as well as in clinical colorectal cancer samples. Interestingly, while the expression of the CTNNB1 target gene set does not correlate with survival, elevated expression of negative feedback regulators within the signature predicts better prognosis. CONCLUSION: Our data provide a genome-wide view of chromatin occupancy and gene regulation of Wnt/CTNNB1 signaling in colon cancer cells.


Subject(s)
Chromatin Immunoprecipitation , Colonic Neoplasms/genetics , Gene Expression Profiling , Intestines/pathology , Oligonucleotide Array Sequence Analysis , Stem Cells/metabolism , beta Catenin/metabolism , Cell Line, Tumor , Cell Survival , Chromatin/metabolism , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Gene Ontology , Humans , Kaplan-Meier Estimate , Reproducibility of Results , Software , Stem Cells/pathology , Wnt Signaling Pathway/genetics
20.
Cell Stem Cell ; 13(1): 48-61, 2013 Jul 03.
Article in English | MEDLINE | ID: mdl-23684539

ABSTRACT

Epigenetic mechanisms regulating lineage differentiation of mammary stem cells (MaSCs) remain poorly understood. Pygopus 2 (Pygo2) is a histone methylation reader and a context-dependent Wnt/ß-catenin coactivator. Here we provide evidence for Pygo2's function in suppressing luminal/alveolar differentiation of MaSC-enriched basal cells. We show that Pygo2-deficient MaSC/basal cells exhibit partial molecular resemblance to luminal cells, such as elevated Notch signaling and reduced mammary repopulating capability upon transplantation. Inhibition of Notch signaling suppresses basal-level and Pygo2-deficiency-induced luminal/alveolar differentiation of MaSC/basal cells, whereas activation of Wnt/ß-catenin signaling suppresses luminal/alveolar differentiation and Notch3 expression in a Pygo2-dependent manner. We show that Notch3 is a direct target of Pygo2 and that Pygo2 is required for ß-catenin binding and maintenance of a poised/repressed chromatin state at the Notch3 locus in MaSC/basal cells. Together, our data support a model where Pygo2-mediated chromatin regulation connects Wnt signaling and Notch signaling to restrict the luminal/alveolar differentiation competence of MaSC/basal cells.


Subject(s)
Cell Differentiation , Epithelial Cells/cytology , Intracellular Signaling Peptides and Proteins/physiology , Mammary Glands, Animal/cytology , Receptors, Notch/metabolism , Stem Cells/cytology , Wnt Proteins/metabolism , Animals , Blotting, Western , Cell Lineage , Cell Proliferation , Epithelial Cells/metabolism , Female , Flow Cytometry , Gene Expression Regulation, Developmental , Male , Mammary Glands, Animal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, Notch3 , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Stem Cells/metabolism , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...