Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
Add more filters

Publication year range
1.
Cell ; 162(1): 33-44, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26140591

ABSTRACT

SNPs affecting disease risk often reside in non-coding genomic regions. Here, we show that SNPs are highly enriched at mouse strain-selective adipose tissue binding sites for PPARγ, a nuclear receptor for anti-diabetic drugs. Many such SNPs alter binding motifs for PPARγ or cooperating factors and functionally regulate nearby genes whose expression is strain selective and imbalanced in heterozygous F1 mice. Moreover, genetically determined binding of PPARγ accounts for mouse strain-specific transcriptional effects of TZD drugs, providing proof of concept for personalized medicine related to nuclear receptor genomic occupancy. In human fat, motif-altering SNPs cause differential PPARγ binding, provide a molecular mechanism for some expression quantitative trait loci, and are risk factors for dysmetabolic traits in genome-wide association studies. One PPARγ motif-altering SNP is associated with HDL levels and other metabolic syndrome parameters. Thus, natural genetic variation in PPARγ genomic occupancy determines individual disease risk and drug response.


Subject(s)
Hypoglycemic Agents/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Polymorphism, Single Nucleotide , Adipose Tissue , Animals , Gene Expression , Humans , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Transcription Factors/metabolism
2.
Trends Genet ; 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38906738

ABSTRACT

Cell-cell interactions orchestrate complex functions in multicellular organisms, forming a regulatory network for diverse biological processes. Their disruption leads to disease states. Recent advancements - including single-cell sequencing and spatial transcriptomics, coupled with powerful bioengineering and molecular tools - have revolutionized our understanding of how cells respond to each other. Notably, spatial transcriptomics allows us to analyze gene expression changes based on cell proximity, offering a unique window into the impact of cell-cell contact. Additionally, computational approaches are being developed to decipher how cell contact governs the symphony of cellular responses. This review explores these cutting-edge approaches, providing valuable insights into deciphering the intricate cellular changes influenced by cell-cell communication.

3.
Mol Cell ; 69(3): 398-411.e6, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29395062

ABSTRACT

The inflammatory response mediated by nuclear factor κB (NF-κB) signaling is essential for host defense against pathogens. Although the regulatory mechanism of NF-κB signaling has been well studied, the molecular basis for epigenetic regulation of the inflammatory response is poorly understood. Here we identify a new signaling axis of PKCα-LSD1-NF-κB, which is critical for activation and amplification of the inflammatory response. In response to excessive inflammatory stimuli, PKCα translocates to the nucleus and phosphorylates LSD1. LSD1 phosphorylation is required for p65 binding and facilitates p65 demethylation, leading to enhanced stability. In vivo genetic analysis using Lsd1SA/SA mice with ablation of LSD1 phosphorylation and chemical approaches in wild-type mice with inhibition of PKCα or LSD1 activity show attenuated sepsis-induced inflammatory lung injury and mortality. Together, we demonstrate that the PKCα-LSD1-NF-κB signaling cascade is crucial for epigenetic control of the inflammatory response, and targeting this signaling could be a powerful therapeutic strategy for systemic inflammatory diseases, including sepsis.


Subject(s)
Histone Demethylases/metabolism , Protein Kinase C/metabolism , Animals , Cell Nucleus/metabolism , Epigenesis, Genetic/genetics , Histone Demethylases/genetics , Inflammation/metabolism , Methylation , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphorylation , Protein Kinase C/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
4.
Proc Natl Acad Sci U S A ; 120(2): e2205371120, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36595695

ABSTRACT

Development of multicellular organisms is orchestrated by persistent cell-cell communication between neighboring partners. Direct interaction between different cell types can induce molecular signals that dictate lineage specification and cell fate decisions. Current single-cell RNA-seq technology cannot adequately analyze cell-cell contact-dependent gene expression, mainly due to the loss of spatial information. To overcome this obstacle and resolve cell-cell contact-specific gene expression during embryogenesis, we performed RNA sequencing of physically interacting cells (PIC-seq) and assessed them alongside similar single-cell transcriptomes derived from developing mouse embryos between embryonic day (E) 7.5 and E9.5. Analysis of the PIC-seq data identified gene expression signatures that were dependent on the presence of specific neighboring cell types. Our computational predictions, validated experimentally, demonstrated that neural progenitor (NP) cells upregulate Lhx5 and Nkx2-1 genes, when exclusively interacting with definitive endoderm (DE) cells. Moreover, there was a reciprocal impact on the transcriptome of DE cells, as they tend to upregulate Rax and Gsc when in contact with NP cells. Using individual cell transcriptome data, we formulated a means of computationally predicting the impact of one cell type on the transcriptome of its neighboring cell types. We have further developed a distinctive spatial-t-distributed stochastic neighboring embedding to display the pseudospatial distribution of cells in a 2-dimensional space. In summary, we describe an innovative approach to study contact-specific gene regulation during embryogenesis.


Subject(s)
Embryonic Development , Gene Expression Regulation, Developmental , Animals , Mice , Embryonic Development/genetics , Cell Differentiation/genetics , Transcriptome , Sequence Analysis, RNA , Single-Cell Analysis/methods , Gene Expression Profiling
5.
EMBO J ; 40(17): e107271, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34368984

ABSTRACT

Tumors are complex cellular and acellular environments within which cancer clones are under continuous selection pressures. Cancer cells are in a permanent mode of interaction and competition with each other as well as with the immediate microenvironment. In the course of these competitive interactions, cells share information regarding their general state of fitness, with less-fit cells being typically eliminated via apoptosis at the hands of those cells with greater cellular fitness. Competitive interactions involving exchange of cell fitness information have implications for tumor growth, metastasis, and therapy outcomes. Recent research has highlighted sophisticated pathways such as Flower, Hippo, Myc, and p53 signaling, which are employed by cancer cells and the surrounding microenvironment cells to achieve their evolutionary goals by means of cell competition mechanisms. In this review, we discuss these recent findings and explain their importance and role in evolution, growth, and treatment of cancer. We further consider potential physiological conditions, such as hypoxia and chemotherapy, that can function as selective pressures under which cell competition mechanisms may evolve differently or synergistically to confer oncogenic advantages to cancer.


Subject(s)
Cell Competition , Neoplasms/metabolism , Tumor Microenvironment , Animals , Humans , Neoplasms/pathology , Signal Transduction
6.
Development ; 149(13)2022 07 01.
Article in English | MEDLINE | ID: mdl-35708349

ABSTRACT

Pancreatic and duodenal homeobox 1 (PDX1) is crucial for pancreas organogenesis, yet the dynamic changes in PDX1 binding in human or mouse developing pancreas have not been examined. To address this knowledge gap, we performed PDX1 ChIP-seq and single-cell RNA-seq using fetal human pancreata. We integrated our datasets with published datasets and revealed the dynamics of PDX1 binding and potential cell lineage-specific PDX1-bound genes in the pancreas from fetal to adult stages. We identified a core set of developmentally conserved PDX1-bound genes that reveal the broad multifaceted role of PDX1 in pancreas development. Despite the well-known dramatic changes in PDX1 function and expression, we found that PDX1-bound genes are largely conserved from embryonic to adult stages. This points towards a dual role of PDX1 in regulating the expression of its targets at different ages, dependent on other functionally congruent or directly interacting partners. We also showed that PDX1 binding is largely conserved in mouse pancreas. Together, our study reveals PDX1 targets in the developing pancreas in vivo and provides an essential resource for future studies on pancreas development.


Subject(s)
Genes, Homeobox , Homeodomain Proteins , Animals , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Pancreas , Trans-Activators/genetics , Trans-Activators/metabolism , Transcriptome/genetics
7.
Genes Dev ; 31(7): 660-673, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28428261

ABSTRACT

The transcription factor early B-cell factor 2 (EBF2) is an essential mediator of brown adipocyte commitment and terminal differentiation. However, the mechanisms by which EBF2 regulates chromatin to activate brown fat-specific genes in adipocytes were unknown. ChIP-seq (chromatin immunoprecipitation [ChIP] followed by deep sequencing) analyses in brown adipose tissue showed that EBF2 binds and regulates the activity of lineage-specific enhancers. Mechanistically, EBF2 physically interacts with the chromatin remodeler BRG1 and the BAF chromatin remodeling complex in brown adipocytes. We identified the histone reader protein DPF3 as a brown fat-selective component of the BAF complex that was required for brown fat gene programming and mitochondrial function. Loss of DPF3 in brown adipocytes reduced chromatin accessibility at EBF2-bound enhancers and led to a decrease in basal and catecholamine-stimulated expression of brown fat-selective genes. Notably, Dpf3 is a direct transcriptional target of EBF2 in brown adipocytes, thereby establishing a regulatory module through which EBF2 activates and also recruits DPF3-anchored BAF complexes to chromatin. Together, these results reveal a novel mechanism by which EBF2 cooperates with a tissue-specific chromatin remodeling complex to activate brown fat identity genes.


Subject(s)
Adipogenesis/genetics , Adipose Tissue, Brown/cytology , Basic Helix-Loop-Helix Transcription Factors/physiology , Chromatin Assembly and Disassembly , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/genetics , Histones/metabolism , Transcription Factors/genetics , Adipose Tissue, Brown/metabolism , Animals , Cell Lineage/genetics , Cells, Cultured , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Mice, Knockout , Transcription, Genetic
8.
Mol Syst Biol ; 19(11): e11670, 2023 Nov 09.
Article in English | MEDLINE | ID: mdl-37815040

ABSTRACT

Cells have evolved their communication methods to sense their microenvironments and send biological signals. In addition to communication using ligands and receptors, cells use diverse channels including gap junctions to communicate with their immediate neighbors. Current approaches, however, cannot effectively capture the influence of various microenvironments. Here, we propose a novel approach to investigate cell neighbor-dependent gene expression (CellNeighborEX) in spatial transcriptomics (ST) data. To categorize cells based on their microenvironment, CellNeighborEX uses direct cell location or the mixture of transcriptome from multiple cells depending on ST technologies. For each cell type, CellNeighborEX identifies diverse gene sets associated with partnering cell types, providing further insight. We found that cells express different genes depending on their neighboring cell types in various tissues including mouse embryos, brain, and liver cancer. Those genes are associated with critical biological processes such as development or metastases. We further validated that gene expression is induced by neighboring partners via spatial visualization. The neighbor-dependent gene expression suggests new potential genes involved in cell-cell interactions beyond what ligand-receptor co-expression can discover.


Subject(s)
Liver Neoplasms , Transcriptome , Animals , Mice , Transcriptome/genetics , Gene Expression Profiling , Brain , Cell Communication , Tumor Microenvironment
9.
Drug Metab Dispos ; 52(8): 754-764, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38302428

ABSTRACT

The mammalian gut microbiota plays diverse and essential roles in modulating host physiology. Key mediators determining the outcome of the microbiota-host interactions are the small molecule metabolites produced by the gut microbiota. The liver is a major organ exposed to gut microbial metabolites, and it serves as the nexus for maintaining healthy interactions between the gut microbiota and the host. At the same time, the liver is the primary target of potentially harmful gut microbial metabolites. In this review, we provide an up-to-date list of gut microbial metabolites that have been identified to either increase or decrease host susceptibility to acetaminophen (APAP)-induced liver injury. The signaling pathways and molecular factors involved in the progression of APAP-induced hepatotoxicity are well-established, and we propose that the mouse model of APAP-induced hepatotoxicity serves as a model system for uncovering gut microbial metabolites with previously unknown functions. Furthermore, we envision that gut microbial metabolites identified to alter APAP-induced hepatotoxicity likely have broader implications in other liver diseases. SIGNIFICANCE STATEMENT: This review provides an overview of the role of the gut microbiota in modulating the host susceptibility to acetaminophen (APAP)-induced liver injury. It focuses on the roles of gut bacterial small molecule metabolites as mediators of the interaction between the gut microbiota and the liver. It also illustrates the utility of APAP-induced liver injury as a model to identify gut microbial metabolites with biological function.


Subject(s)
Acetaminophen , Chemical and Drug Induced Liver Injury , Gastrointestinal Microbiome , Acetaminophen/metabolism , Acetaminophen/toxicity , Acetaminophen/adverse effects , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/etiology , Animals , Humans , Liver/metabolism , Liver/drug effects , Mice , Disease Susceptibility , Analgesics, Non-Narcotic/toxicity , Analgesics, Non-Narcotic/metabolism , Analgesics, Non-Narcotic/adverse effects
10.
Nucleic Acids Res ; 50(13): 7298-7309, 2022 07 22.
Article in English | MEDLINE | ID: mdl-35801910

ABSTRACT

Autophagy, a catabolic process to remove unnecessary or dysfunctional organelles, is triggered by various signals including nutrient starvation. Depending on the types of the nutrient deficiency, diverse sensing mechanisms and signaling pathways orchestrate for transcriptional and epigenetic regulation of autophagy. However, our knowledge about nutrient type-specific transcriptional regulation during autophagy is limited. To understand nutrient type-dependent transcriptional mechanisms during autophagy, we performed single cell RNA sequencing (scRNAseq) in the mouse embryonic fibroblasts (MEFs) with or without glucose starvation (GS) as well as amino acid starvation (AAS). Trajectory analysis using scRNAseq identified sequential induction of potential transcriptional regulators for each condition. Gene regulatory rules inferred using TENET newly identified CCAAT/enhancer binding protein γ (C/EBPγ) as a regulator of autophagy in AAS, but not GS, condition, and knockdown experiment confirmed the TENET result. Cell biological and biochemical studies validated that activating transcription factor 4 (ATF4) is responsible for conferring specificity to C/EBPγ for the activation of autophagy genes under AAS, but not under GS condition. Together, our data identified C/EBPγ as a previously unidentified key regulator under AAS-induced autophagy.


Subject(s)
Amino Acids , CCAAT-Enhancer-Binding Proteins/metabolism , Transcriptome , Activating Transcription Factor 4/metabolism , Amino Acids/genetics , Amino Acids/metabolism , Animals , Autophagy/genetics , Epigenesis, Genetic , Fibroblasts/metabolism , Mice , Single-Cell Analysis
11.
Genes Dev ; 30(21): 2433-2442, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27856615

ABSTRACT

Methylated cytosines are associated with gene silencing. The ten-eleven translocation (TET) hydroxylases, which oxidize methylated cytosines to 5-hydroxymethylcytosine (5hmC), are essential for cytosine demethylation. Gene silencing and activation are critical for intestinal stem cell (ISC) maintenance and differentiation, but the potential role of TET hydroxylases in these processes has not yet been examined. Here, we generated genome-wide maps of the 5hmC mark in ISCs and their differentiated progeny. Genes with high levels of hydroxymethylation in ISCs are strongly associated with Wnt signaling and developmental processes. We found Tet1 to be the most abundantly expressed Tet gene in ISCs; therefore, we analyzed intestinal development in Tet1-deficient mice and determined that these mice are growth-retarded, exhibit partial postnatal lethality, and have significantly reduced numbers of proliferative cells in the intestinal epithelium. In addition, the Tet1-deficient intestine displays reduced organoid-forming capacity. In the Tet1-deficient crypt, decreased expression of Wnt target genes such as Axin2 and Lgr5 correlates with lower 5hmC levels at their promoters. These data demonstrate that Tet1-mediated DNA hydroxymethylation plays a critical role in the epigenetic regulation of the Wnt pathway in intestinal stem and progenitor cells and consequently in the self-renewal of the intestinal epithelium.


Subject(s)
DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Developmental/genetics , Intestines/growth & development , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Stem Cells/physiology , Animals , Cell Differentiation/genetics , Cells, Cultured , Intestines/cytology , Mice , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/genetics , Sequence Deletion , Stem Cells/cytology , Wnt Signaling Pathway/genetics
12.
Genome Res ; 30(2): 205-213, 2020 02.
Article in English | MEDLINE | ID: mdl-31992615

ABSTRACT

To process large-scale single-cell RNA-sequencing (scRNA-seq) data effectively without excessive distortion during dimension reduction, we present SHARP, an ensemble random projection-based algorithm that is scalable to clustering 10 million cells. Comprehensive benchmarking tests on 17 public scRNA-seq data sets show that SHARP outperforms existing methods in terms of speed and accuracy. Particularly, for large-size data sets (more than 40,000 cells), SHARP runs faster than other competitors while maintaining high clustering accuracy and robustness. To the best of our knowledge, SHARP is the only R-based tool that is scalable to clustering scRNA-seq data with 10 million cells.


Subject(s)
RNA-Seq , Single-Cell Analysis , Software , Transcriptome/genetics , Algorithms , Cluster Analysis , Gene Expression Profiling , Humans , RNA/classification , RNA/genetics , Sequence Analysis, RNA , Exome Sequencing
13.
PLoS Pathog ; 17(1): e1009208, 2021 01.
Article in English | MEDLINE | ID: mdl-33497421

ABSTRACT

Epstein-Barr virus (EBV) immortalizes resting B-lymphocytes through a highly orchestrated reprogramming of host chromatin structure, transcription and metabolism. Here, we use a multi-omics-based approach to investigate these underlying mechanisms. ATAC-seq analysis of cellular chromatin showed that EBV alters over a third of accessible chromatin during the infection time course, with many of these sites overlapping transcription factors such as PU.1, Interferon Regulatory Factors (IRFs), and CTCF. Integration of RNA-seq analysis identified a complex transcriptional response and associations with EBV nuclear antigens (EBNAs). Focusing on EBNA1 revealed enhancer-binding activity at gene targets involved in nucleotide metabolism, supported by metabolomic analysis which indicated that adenosine and purine metabolism are significantly altered by EBV immortalization. We further validated that adenosine deaminase (ADA) is a direct and critical target of the EBV-directed immortalization process. These findings reveal that purine metabolism and ADA may be useful therapeutic targets for EBV-driven lymphoid cancers.


Subject(s)
B-Lymphocytes/pathology , Cell Transformation, Viral , Chromatin/genetics , Epstein-Barr Virus Infections/pathology , Epstein-Barr Virus Nuclear Antigens/metabolism , Nucleotides/metabolism , Viral Proteins/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Chromatin/metabolism , Epigenesis, Genetic , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Nuclear Antigens/genetics , Herpesvirus 4, Human/physiology , Host-Pathogen Interactions , Humans , Metabolome , Transcriptome , Viral Proteins/genetics
14.
Mol Syst Biol ; 18(9): e11080, 2022 09.
Article in English | MEDLINE | ID: mdl-36065846

ABSTRACT

Characterization of tissue architecture promises to deliver insights into development, cell communication, and disease. In silico spatial domain retrieval methods have been developed for spatial transcriptomics (ST) data assuming transcriptional similarity of neighboring barcodes. However, domain retrieval approaches with this assumption cannot work in complex tissues composed of multiple cell types. This task becomes especially challenging in cellular resolution ST methods. We developed Vesalius to decipher tissue anatomy from ST data by applying image processing technology. Vesalius uniquely detected territories composed of multiple cell types and successfully recovered tissue structures in high-resolution ST data including in mouse brain, embryo, liver, and colon. Utilizing this tissue architecture, Vesalius identified tissue morphology-specific gene expression and regional specific gene expression changes for astrocytes, interneuron, oligodendrocytes, and entorhinal cells in the mouse brain.


Subject(s)
Transcriptome , Animals , Mice , Transcriptome/genetics
15.
Mol Psychiatry ; 27(6): 2751-2765, 2022 06.
Article in English | MEDLINE | ID: mdl-35444258

ABSTRACT

Autism spectrum disorders (ASDs) are common neurodevelopmental disorders characterized by deficits in social interactions and communication, restricted interests, and repetitive behaviors. Despite extensive study, the molecular targets that control ASD development remain largely unclear. Here, we report that the dormancy of quiescent neural stem cells (qNSCs) is a therapeutic target for controlling the development of ASD phenotypes driven by Shank3 deficiency. Using single-cell RNA sequencing (scRNA-seq) and transposase accessible chromatin profiling (ATAC-seq), we find that abnormal epigenetic features including H3K4me3 accumulation due to up-regulation of Kmt2a levels lead to increased dormancy of qNSCs in the absence of Shank3. This result in decreased active neurogenesis in the Shank3 deficient mouse brain. Remarkably, pharmacological and molecular inhibition of qNSC dormancy restored adult neurogenesis and ameliorated the social deficits observed in Shank3-deficient mice. Moreover, we confirmed restored human qNSC activity rescues abnormal neurogenesis and autism-like phenotypes in SHANK3-targeted human NSCs. Taken together, our results offer a novel strategy to control qNSC activity as a potential therapeutic target for the development of autism.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Neural Stem Cells , Animals , Autism Spectrum Disorder/genetics , Autistic Disorder/genetics , Disease Models, Animal , Mice , Microfilament Proteins/genetics , Mutation , Nerve Tissue Proteins/genetics
16.
Nature ; 546(7659): 544-548, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28614293

ABSTRACT

Brown adipose tissue is a thermogenic organ that dissipates chemical energy as heat to protect animals against hypothermia and to counteract metabolic disease. However, the transcriptional mechanisms that determine the thermogenic capacity of brown adipose tissue before environmental cold are unknown. Here we show that histone deacetylase 3 (HDAC3) is required to activate brown adipose tissue enhancers to ensure thermogenic aptitude. Mice with brown adipose tissue-specific genetic ablation of HDAC3 become severely hypothermic and succumb to acute cold exposure. Uncoupling protein 1 (UCP1) is nearly absent in brown adipose tissue lacking HDAC3, and there is also marked downregulation of mitochondrial oxidative phosphorylation genes resulting in diminished mitochondrial respiration. Remarkably, although HDAC3 acts canonically as a transcriptional corepressor, it functions as a coactivator of oestrogen-related receptor α (ERRα) in brown adipose tissue. HDAC3 coactivation of ERRα is mediated by deacetylation of PGC-1α and is required for the transcription of Ucp1, Ppargc1a (encoding PGC-1α), and oxidative phosphorylation genes. Importantly, HDAC3 promotes the basal transcription of these genes independently of adrenergic stimulation. Thus, HDAC3 uniquely primes Ucp1 and the thermogenic transcriptional program to maintain a critical capacity for thermogenesis in brown adipose tissue that can be rapidly engaged upon exposure to dangerously cold temperature.


Subject(s)
Adipose Tissue, Brown/metabolism , Gene Expression Regulation , Histone Deacetylases/metabolism , Thermogenesis , Animals , Cell Respiration , Cold Temperature , Enhancer Elements, Genetic/genetics , Hot Temperature , Humans , Male , Mice , Mitochondria/metabolism , Oxidative Phosphorylation , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Receptors, Estrogen/metabolism , Thermogenesis/genetics , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , ERRalpha Estrogen-Related Receptor
17.
Nucleic Acids Res ; 49(1): e1, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33170214

ABSTRACT

Accurate prediction of gene regulatory rules is important towards understanding of cellular processes. Existing computational algorithms devised for bulk transcriptomics typically require a large number of time points to infer gene regulatory networks (GRNs), are applicable for a small number of genes and fail to detect potential causal relationships effectively. Here, we propose a novel approach 'TENET' to reconstruct GRNs from single cell RNA sequencing (scRNAseq) datasets. Employing transfer entropy (TE) to measure the amount of causal relationships between genes, TENET predicts large-scale gene regulatory cascades/relationships from scRNAseq data. TENET showed better performance than other GRN reconstructors, in identifying key regulators from public datasets. Specifically from scRNAseq, TENET identified key transcriptional factors in embryonic stem cells (ESCs) and during direct cardiomyocytes reprogramming, where other predictors failed. We further demonstrate that known target genes have significantly higher TE values, and TENET predicted higher TE genes were more influenced by the perturbation of their regulator. Using TENET, we identified and validated that Nme2 is a culture condition specific stem cell factor. These results indicate that TENET is uniquely capable of identifying key regulators from scRNAseq data.


Subject(s)
Algorithms , Computational Biology/methods , Entropy , Gene Regulatory Networks , Single-Cell Analysis/methods , Transcriptome , Alkaline Phosphatase/metabolism , Animals , Cell Proliferation/genetics , Gene Expression Profiling/methods , Gene Ontology , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Sequence Analysis, RNA/methods , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Genes Dev ; 29(3): 298-307, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25644604

ABSTRACT

PR (PRD1-BF1-RIZ1 homologous) domain-containing 16 (PRDM16) drives a brown fat differentiation program, but the mechanisms by which PRDM16 activates brown fat-selective genes have been unclear. Through chromatin immunoprecipitation (ChIP) followed by deep sequencing (ChIP-seq) analyses in brown adipose tissue (BAT), we reveal that PRDM16 binding is highly enriched at a broad set of brown fat-selective genes. Importantly, we found that PRDM16 physically binds to MED1, a component of the Mediator complex, and recruits it to superenhancers at brown fat-selective genes. PRDM16 deficiency in BAT reduces MED1 binding at PRDM16 target sites and causes a fundamental change in chromatin architecture at key brown fat-selective genes. Together, these data indicate that PRDM16 controls chromatin architecture and superenhancer activity in BAT.


Subject(s)
Adipose Tissue, Brown/metabolism , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Mediator Complex Subunit 1/metabolism , Transcription Factors/metabolism , Transcription, Genetic/physiology , Animals , Chromatin/chemistry , Chromatin/genetics , Enhancer Elements, Genetic , Mice
19.
Development ; 146(18)2019 09 19.
Article in English | MEDLINE | ID: mdl-31488567

ABSTRACT

The mammalian cochlea develops from a ventral outgrowth of the otic vesicle in response to Shh signaling. Mouse embryos lacking Shh or its essential signal transduction components display cochlear agenesis; however, a detailed understanding of the transcriptional network mediating this process is unclear. Here, we describe an integrated genomic approach to identify Shh-dependent genes and associated regulatory sequences that promote cochlear duct morphogenesis. A comparative transcriptome analysis of otic vesicles from mouse mutants exhibiting loss (Smoecko ) and gain (Shh-P1) of Shh signaling reveal a set of Shh-responsive genes partitioned into four expression categories in the ventral half of the otic vesicle. This target gene classification scheme provides novel insight into several unanticipated roles for Shh, including priming the cochlear epithelium for subsequent sensory development. We also mapped regions of open chromatin in the inner ear by ATAC-seq that, in combination with Gli2 ChIP-seq, identified inner ear enhancers in the vicinity of Shh-responsive genes. These datasets are useful entry points for deciphering Shh-dependent regulatory mechanisms involved in cochlear duct morphogenesis and establishment of its constituent cell types.


Subject(s)
Cochlea/embryology , Cochlea/metabolism , Genome , Hedgehog Proteins/metabolism , Morphogenesis/genetics , Animals , Base Sequence , Embryo, Mammalian/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental , Mice, Transgenic , Reproducibility of Results
20.
Bioinformatics ; 37(20): 3509-3513, 2021 Oct 25.
Article in English | MEDLINE | ID: mdl-33974009

ABSTRACT

MOTIVATION: Trajectory inference (TI) for single cell RNA sequencing (scRNAseq) data is a powerful approach to interpret dynamic cellular processes such as cell cycle and development. Still, however, accurate inference of trajectory is challenging. Recent development of RNA velocity provides an approach to visualize cell state transition without relying on prior knowledge. RESULTS: To perform TI and group cells based on RNA velocity we developed VeTra. By applying cosine similarity and merging weakly connected components, VeTra identifies cell groups from the direction of cell transition. Besides, VeTra suggests key regulators from the inferred trajectory. VeTra is a useful tool for TI and subsequent analysis. AVAILABILITY AND IMPLEMENTATION: The Vetra is available at https://github.com/wgzgithub/VeTra. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

SELECTION OF CITATIONS
SEARCH DETAIL