Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters











Publication year range
1.
Nat Commun ; 13(1): 4221, 2022 07 21.
Article in English | MEDLINE | ID: mdl-35864091

ABSTRACT

Sonic hedgehog (Shh) is essential for limb development, and the mechanisms that govern the propagation and maintenance of its expression has been well studied; however, the mechanisms that govern the initiation of Shh expression are incomplete. Here we report that ETV2 initiates Shh expression by changing the chromatin status of the developmental limb enhancer, ZRS. Etv2 expression precedes Shh in limb buds, and Etv2 inactivation prevents the opening of limb chromatin, including the ZRS, resulting in an absence of Shh expression. Etv2 overexpression in limb buds causes nucleosomal displacement at the ZRS, ectopic Shh expression, and polydactyly. Areas of nucleosome displacement coincide with ETS binding site clusters. ETV2 also functions as a transcriptional activator of ZRS and is antagonized by ETV4/5 repressors. Known human polydactyl mutations introduce novel ETV2 binding sites in the ZRS, suggesting that ETV2 dosage regulates ZRS activation. These studies identify ETV2 as a pioneer transcription factor (TF) regulating the onset of Shh expression, having both a chromatin regulatory role and a transcriptional activation role.


Subject(s)
Hedgehog Proteins , Limb Buds , Polydactyly , Transcription Factors , Animals , Chromatin/genetics , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Limb Buds/growth & development , Mice , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Nat Biotechnol ; 38(3): 297-302, 2020 03.
Article in English | MEDLINE | ID: mdl-32094659

ABSTRACT

The scarcity of donor organs may be addressed in the future by using pigs to grow humanized organs with lower potential for immunological rejection after transplantation in humans. Previous studies have demonstrated that interspecies complementation of rodent blastocysts lacking a developmental regulatory gene can generate xenogeneic pancreas and kidney1,2. However, such organs contain host endothelium, a source of immune rejection. We used gene editing and somatic cell nuclear transfer to engineer porcine embryos deficient in ETV2, a master regulator of hematoendothelial lineages3-7. ETV2-null pig embryos lacked hematoendothelial lineages and were embryonic lethal. Blastocyst complementation with wild-type porcine blastomeres generated viable chimeric embryos whose hematoendothelial cells were entirely donor-derived. ETV2-null blastocysts were injected with human induced pluripotent stem cells (hiPSCs) or hiPSCs overexpressing the antiapoptotic factor BCL2, transferred to synchronized gilts and analyzed between embryonic day 17 and embryonic day 18. In these embryos, all endothelial cells were of human origin.


Subject(s)
Blastomeres/cytology , Embryo, Mammalian/metabolism , Endothelium/metabolism , Induced Pluripotent Stem Cells/transplantation , Transcription Factors/deficiency , Animals , Blastomeres/metabolism , Cells, Cultured , Embryonic Development , Endothelium/cytology , Gene Editing , Humans , Induced Pluripotent Stem Cells/metabolism , Nuclear Transfer Techniques , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Swine
3.
Nat Commun ; 9(1): 4237, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30315164

ABSTRACT

The mammalian heart has a limited regenerative capacity and typically progresses to heart failure following injury. Here, we defined a hedgehog (HH)-Gli1-Mycn network for cardiomyocyte proliferation and heart regeneration from amphibians to mammals. Using a genome-wide screen, we verified that HH signaling was essential for heart regeneration in the injured newt. Next, pharmacological and genetic loss- and gain-of-function of HH signaling demonstrated the essential requirement for HH signaling in the neonatal, adolescent, and adult mouse heart regeneration, and in the proliferation of hiPSC-derived cardiomyocytes. Fate-mapping and molecular biological studies revealed that HH signaling, via a HH-Gli1-Mycn network, contributed to heart regeneration by inducing proliferation of pre-existing cardiomyocytes and not by de novo cardiomyogenesis. Further, Mycn mRNA transfection experiments recapitulated the effects of HH signaling and promoted adult cardiomyocyte proliferation. These studies defined an evolutionarily conserved function of HH signaling that may serve as a platform for human regenerative therapies.


Subject(s)
Heart/physiology , Hedgehog Proteins/metabolism , N-Myc Proto-Oncogene Protein/metabolism , Regeneration/physiology , Salamandridae/metabolism , Zinc Finger Protein GLI1/metabolism , Animals , Cell Proliferation/genetics , Cell Proliferation/physiology , Hedgehog Proteins/genetics , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , N-Myc Proto-Oncogene Protein/genetics , Regeneration/genetics , Salamandridae/physiology , Signal Transduction , Zinc Finger Protein GLI1/genetics
4.
Nat Commun ; 9(1): 2749, 2018 07 16.
Article in English | MEDLINE | ID: mdl-30013097

ABSTRACT

Profiling single cell gene expression data over specified time periods are increasingly applied to the study of complex developmental processes. Here, we describe a novel prototype-based dimension reduction method to visualize high throughput temporal expression data for single cell analyses. Our software preserves the global developmental trajectories over a specified time course, and it also identifies subpopulations of cells within each time point demonstrating superior visualization performance over six commonly used methods.


Subject(s)
Algorithms , Gene Expression Regulation, Developmental , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Single-Cell Analysis/methods , Activins/pharmacology , Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation , Fibroblast Growth Factor 2/pharmacology , Gene Expression Profiling , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Multifactor Dimensionality Reduction , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology , Software
5.
BMC Bioinformatics ; 19(1): 220, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29884114

ABSTRACT

BACKGROUND: The single cell RNA sequencing (scRNA-seq) technique begin a new era by allowing the observation of gene expression at the single cell level. However, there is also a large amount of technical and biological noise. Because of the low number of RNA transcriptomes and the stochastic nature of the gene expression pattern, there is a high chance of missing nonzero entries as zero, which are called dropout events. RESULTS: We develop DrImpute to impute dropout events in scRNA-seq data. We show that DrImpute has significantly better performance on the separation of the dropout zeros from true zeros than existing imputation algorithms. We also demonstrate that DrImpute can significantly improve the performance of existing tools for clustering, visualization and lineage reconstruction of nine published scRNA-seq datasets. CONCLUSIONS: DrImpute can serve as a very useful addition to the currently existing statistical tools for single cell RNA-seq analysis. DrImpute is implemented in R and is available at https://github.com/gongx030/DrImpute .


Subject(s)
RNA/genetics , Sequence Analysis, RNA/methods , Humans
6.
PLoS One ; 12(12): e0189010, 2017.
Article in English | MEDLINE | ID: mdl-29232705

ABSTRACT

Remodeling of the primitive vasculature is necessary for the formation of a complex branched vascular architecture. However, the factors that modulate these processes are incompletely defined. Previously, we defined the role of microRNAs (miRNAs) in endothelial specification. In the present study, we further examined the Etv2-Cre mediated ablation of DicerL/L and characterized the perturbed vascular patterning in the embryo proper and yolk-sac. We mechanistically defined an important role for miR-130a, an Etv2 downstream target, in the mediation of vascular patterning and angiogenesis in vitro and in vivo. Inducible overexpression of miR-130a resulted in robust induction of vascular sprouts and angiogenesis with increased uptake of acetylated-LDL. Mechanistically, miR-130a directly regulated Jarid2 expression by binding to its 3'-UTR region. Over-expression of Jarid2 in HUVEC cells led to defective tube formation indicating its inhibitory role in angiogenesis. The knockout of miR-130a showed increased levels of Jarid2 in the ES/EB system. In addition, the levels of Jarid2 transcripts were increased in the Etv2-null embryos at E8.5. In the in vivo settings, injection of miR-130a specific morpholinos in zebrafish embryos resulted in perturbed vascular patterning with reduced levels of endothelial transcripts in the miR-130a morphants. Further, co-injection of miR-130a mimics in the miR-130a morphants rescued the vascular defects during embryogenesis. qPCR and in situ hybridization techniques demonstrated increased expression of jarid2a in the miR-130a morphants in vivo. These findings demonstrate a critical role for Etv2-miR-130a-Jarid2 in vascular patterning both in vitro and in vivo.


Subject(s)
Blood Vessels/embryology , Body Patterning/genetics , Embryonic Development , MicroRNAs/genetics , Polycomb Repressive Complex 2/genetics , Transcription Factors/genetics , Animals , Female , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Zebrafish/embryology
7.
Cardiovasc Res ; 113(11): 1294-1306, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28859300

ABSTRACT

The 'master regulatory factors' that position at the top of the genetic hierarchy of lineage determination have been a focus of intense interest, and have been investigated in various systems. Etv2/Etsrp71/ER71 is such a factor that is both necessary and sufficient for the development of haematopoietic and endothelial lineages. As such, genetic ablation of Etv2 leads to complete loss of blood and vessels, and overexpression can convert non-endothelial cells to the endothelial lineage. Understanding such master regulatory role of a lineage is not only a fundamental quest in developmental biology, but also holds immense possibilities in regenerative medicine. To harness its activity and utility for therapeutic interventions, it is essential to understand the regulatory mechanisms, molecular function, and networks that surround Etv2. In this review, we provide a comprehensive overview of Etv2 biology focused on mouse and human systems.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Hematopoietic Stem Cells/cytology , Transcription Factors/genetics , Animals , Gene Expression Regulation, Developmental/genetics , Humans , Neovascularization, Physiologic/genetics
8.
Nat Commun ; 8: 14362, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28181481

ABSTRACT

Developmental, stem cell and cancer biologists are interested in the molecular definition of cellular differentiation. Although single-cell RNA sequencing represents a transformational advance for global gene analyses, novel obstacles have emerged, including the computational management of dropout events, the reconstruction of biological pathways and the isolation of target cell populations. We develop an algorithm named dpath that applies the concept of metagene entropy and allows the ranking of cells based on their differentiation potential. We also develop self-organizing map (SOM) and random walk with restart (RWR) algorithms to separate the progenitors from the differentiated cells and reconstruct the lineage hierarchies in an unbiased manner. We test these algorithms using single cells from Etv2-EYFP transgenic mouse embryos and reveal specific molecular pathways that direct differentiation programmes involving the haemato-endothelial lineages. This software program quantitatively assesses the progenitor and committed states in single-cell RNA-seq data sets in a non-biased manner.


Subject(s)
Cell Lineage/genetics , Gene Expression Profiling , Hematopoietic Stem Cells/cytology , Mouse Embryonic Stem Cells/cytology , Single-Cell Analysis , Transcription Factors/metabolism , Algorithms , Animals , Cell Aggregation , Cell Separation , Cluster Analysis , Embryoid Bodies/cytology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Hematopoietic Stem Cells/metabolism , Male , Mice , Mouse Embryonic Stem Cells/metabolism , Reproducibility of Results , Sequence Analysis, RNA , Software , Transcriptome/genetics
9.
Nat Commun ; 7: 13101, 2016 10 07.
Article in English | MEDLINE | ID: mdl-27713415

ABSTRACT

Mechanisms of haematopoietic and cardiac patterning remain poorly understood. Here we show that the BMP and Wnt signalling pathways are integrated in an endoglin (Eng)-dependent manner in cardiac and haematopoietic lineage specification. Eng is expressed in early mesoderm and marks both haematopoietic and cardiac progenitors. In the absence of Eng, yolk sacs inappropriately express the cardiac marker, Nkx2.5. Conversely, high levels of Eng in vitro and in vivo increase haematopoiesis and inhibit cardiogenesis. Levels of Eng determine the activation of both BMP and Wnt pathways, which are integrated downstream of Eng by phosphorylation of Smad1 by Gsk3. By interrogating Eng-dependent Wnt-mediated transcriptional changes, we identify Jdp2 as a key Eng-dependent Wnt target, sufficient to establish haematopoietic fate in early mesoderm when BMP and Wnt crosstalk is disturbed. These studies provide mechanistic insight into the integration of BMP and Wnt signalling in the establishment of haematopoietic and cardiac progenitors during embryogenesis.


Subject(s)
Body Patterning/genetics , Bone Morphogenetic Protein 4/genetics , Endoglin/genetics , Hematopoiesis/genetics , Wnt3 Protein/genetics , Zebrafish Proteins/genetics , Animals , Body Patterning/physiology , Cell Line , Female , Gene Expression Regulation, Developmental , Glycogen Synthase Kinase 3/metabolism , Hematopoiesis/physiology , Homeobox Protein Nkx-2.5/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Signal Transduction/genetics , Smad1 Protein/metabolism , Wnt Signaling Pathway/genetics , Zebrafish , beta Catenin/genetics
11.
Sci Rep ; 6: 22396, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26935534

ABSTRACT

All trans retinoic acid (atRA) is one of the most potent therapeutic agents, but extensive toxicity caused by nuclear RA receptors (RARs) limits its clinical application in treating cancer. AtRA also exerts non-genomic activities for which the mechanism remains poorly understood. We determine that cellular retinoic acid binding protein 1 (Crabp1) mediates the non-genomic activity of atRA, and identify two compounds as the ligands of Crabp1 to rapidly and RAR-independently activate extracellular signal regulated kinase 1/2 (ERK1/2). Non-canonically activated ERK activates protein phosphatase 2A (PP2A) and lengthens cell cycle duration in embryonic stem cells (ESC). This is abolished in Crabp1-null ESCs. Re-expressing Crabp1 in Crabp1-negative cancer cells also sensitizes their apoptotic induction by atRA. This study reveals a physiological relevance of the non-genomic action of atRA, mediated by Crabp1, in modulating cell cycle progression and apoptosis induction, and provides a new cancer therapeutic strategy whereby compounds specifically targeting Crabp1 can modulate cell cycle and cancer cell apoptosis in a RAR-independent fashion, thereby avoiding atRA's toxicity caused by its genomic effects.


Subject(s)
Apoptosis/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Receptors, Retinoic Acid/metabolism , Tretinoin/pharmacology , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Mice , Mitogen-Activated Protein Kinase 3/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation/drug effects , Receptors, Retinoic Acid/genetics
12.
Cell Rep ; 13(5): 915-23, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26565905

ABSTRACT

MicroRNAs (miRNAs) are known to regulate critical developmental stages during embryogenesis. Here, we defined an Etv2-miR-130a cascade that regulates mesodermal specification and determination. Ablation of Dicer in the Etv2-expressing precursors resulted in altered mesodermal lineages and embryonic lethality. We identified miR-130a as a direct target of Etv2 and demonstrated its role in the segregation of bipotent hemato-endothelial progenitors toward the endothelial lineage. Gain-of-function experiments demonstrated that miR-130a promoted the endothelial program at the expense of the cardiac program without impacting the hematopoietic lineages. In contrast, CRISPR/Cas9-mediated knockout of miR-130a demonstrated a reduction of the endothelial program without affecting hematopoiesis. Mechanistically, miR-130a directly suppressed Pdgfra expression and promoted the endothelial program by blocking Pdgfra signaling. Inhibition or activation of Pdgfra signaling phenocopied the miR-130a overexpression and knockout phenotypes, respectively. In summary, we report the function of a miRNA that specifically promotes the divergence of the hemato-endothelial progenitor to the endothelial lineage.


Subject(s)
Cell Lineage , Mesoderm/cytology , MicroRNAs/genetics , Transcription Factors/genetics , Animals , Cells, Cultured , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/metabolism , Hematopoiesis , Mesoderm/metabolism , Mice , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Transcription Factors/metabolism
14.
J Biol Chem ; 290(47): 28107-28119, 2015 Nov 20.
Article in English | MEDLINE | ID: mdl-26396195

ABSTRACT

Etv2 is an essential transcriptional regulator of hematoendothelial lineages during embryogenesis. Although Etv2 downstream targets have been identified, little is known regarding the upstream transcriptional regulation of Etv2 gene expression. In this study, we established a novel methodology that utilizes the differentiating ES cell and embryoid body system to define the modules and enhancers embedded within the Etv2 promoter. Using this system, we defined an autoactivating role for Etv2 that is mediated by two adjacent Ets motifs in the proximal promoter. In addition, we defined the role of VEGF/Flk1-Calcineurin-NFAT signaling cascade in the transcriptional regulation of Etv2. Furthermore, we defined an Etv2-Flt1-Flk1 cascade that serves as a negative feedback mechanism to regulate Etv2 gene expression. To complement and extend these studies, we demonstrated that the Flt1 null embryonic phenotype was partially rescued in the Etv2 conditional knockout background. In summary, these studies define upstream and downstream networks that serve as a transcriptional rheostat to regulate Etv2 gene expression.


Subject(s)
Bone Marrow Cells/cytology , Endothelium/cytology , Gene Expression , Transcription Factors/genetics , Animals , Calcineurin/metabolism , Cell Lineage , Enhancer Elements, Genetic , Female , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/metabolism , Promoter Regions, Genetic , Signal Transduction , Transcription, Genetic , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
Genes (Basel) ; 6(2): 417-35, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-26110318

ABSTRACT

Regulatory networks that govern embryonic development have been well defined. While a common hypothesis supports the notion that the embryonic regulatory cascades are reexpressed following injury and tissue regeneration, the mechanistic regulatory pathways that mediate the regenerative response in higher organisms remain undefined. Relative to mammals, lower vertebrates, including zebrafish and newts, have a tremendous regenerative capacity to repair and regenerate a number of organs including: appendages, retina, heart, jaw and nervous system. Elucidation of the pathways that govern regeneration in these lower organisms may provide cues that will enhance the capacity for the regeneration of mammalian organs. Signaling pathways, such as the hedgehog pathway, have been shown to play critical functions during development and during regeneration in lower organisms. These signaling pathways have been shown to modulate multiple processes including cellular origin, positional identity and cellular maturation. The present review will focus on the cellular and molecular regulation of the hedgehog (HH) signaling pathway and its interaction with other signaling factors during appendage development and regeneration.

16.
Stem Cell Rev Rep ; 11(5): 710-27, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26085192

ABSTRACT

Congenital heart disease (CHD) remains a significant health problem, with a growing population of survivors with chronic disease. Despite intense efforts to understand the genetic basis of CHD in humans, the etiology of most CHD is unknown. Furthermore, new models of CHD are required to better understand the development of CHD and to explore novel therapies for this patient population. In this review, we highlight the role that human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes can serve to enhance our understanding of the development, pathophysiology and potential therapeutic targets for CHD. We highlight the use of hiPSC-derived cardiomyocytes to model gene regulatory interactions, cell-cell interactions and tissue interactions contributing to CHD. We further emphasize the importance of using hiPSC-derived cardiomyocytes as personalized research models. The use of hiPSCs presents an unprecedented opportunity to generate disease-specific cellular models, investigate the underlying molecular mechanisms of disease and uncover new therapeutic targets for CHD.


Subject(s)
Heart Diseases/pathology , Heart/physiology , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/physiology , Organogenesis/physiology , Animals , Cell Differentiation/physiology , Humans
17.
J Card Fail ; 21(9): 761-70, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25934595

ABSTRACT

Inherited cardiomyopathies, including hypertrophic cardiomyopathy, dilated cardiomyopathies, arrythmogenic right ventricular cardiomyopathy, and other inherited forms of heart failure, represent a unique set of genetically defined cardiovascular disease processes. Unraveling the molecular mechanisms of these deadly forms of human heart disease has been challenging, but recent groundbreaking scientific advances in stem cell technology have allowed for the generation of patient-specific human inducible stem cell (hiPSC)-derived cardiomyocytes (CMs). hiPSC-derived CMs retain the genetic blueprint of the patient, they can be maintained in culture, and they recapitulate the phenotypic characteristics of the disease in vitro, thus serving as a disease in a dish. This review provides an overview of in vitro modeling of inherited cardiomyopathies with the use of patient-specific hiPSC-derived CMs.


Subject(s)
Cardiomyopathies , Induced Pluripotent Stem Cells/cytology , Models, Cardiovascular , Myocytes, Cardiac/pathology , Animals , Cardiomyopathies/congenital , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Humans
18.
J Biol Chem ; 290(24): 15350-61, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25940086

ABSTRACT

We have previously isolated a muscle-specific Kelch gene, Kelch repeat and BTB domain containing protein 5 (Kbtbd5)/Kelch-like protein 40 (Klhl40). In this report, we identified DP1 as a direct interacting factor for Kbtbd5 using a yeast two-hybrid screen and in vitro binding assays. Our studies demonstrate that Kbtbd5 interacts and regulates the cytoplasmic localization of DP1. GST pulldown assays demonstrate that the dimerization domain of DP1 interacts with all three of the Kbtbd5 domains. We further show that Kbtbd5 promotes the ubiquitination and degradation of DP1, thereby inhibiting E2F1-DP1 activity. To investigate the in vivo function of Kbtbd5, we used gene disruption technology and engineered Kbtbd5 null mice. Targeted deletion of Kbtbd5 resulted in postnatal lethality. Histological studies reveal that the Kbtbd5 null mice have smaller muscle fibers, a disorganized sarcomeric structure, increased extracellular matrix, and decreased numbers of mitochondria compared with wild-type controls. RNA sequencing and quantitative PCR analyses demonstrate the up-regulation of E2F1 target apoptotic genes (Bnip3 and p53inp1) in Kbtbd5 null skeletal muscle. Consistent with these observations, the cellular apoptosis in Kbtbd5 null mice was increased. Breeding of Kbtbd5 null mouse into the E2F1 null background rescues the lethal phenotype of the Kbtbd5 null mice but not the growth defect. The expression of Bnip3 and p53inp1 in Kbtbd5 mutant skeletal muscle are also restored to control levels in the E2F1 null background. In summary, our studies demonstrate that Kbtbd5 regulates skeletal muscle myogenesis through the regulation of E2F1-DP1 activity.


Subject(s)
E2F1 Transcription Factor/physiology , Muscle Proteins/physiology , Muscle, Skeletal/growth & development , Transcription Factor DP1/physiology , Animals , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Transcription Factor DP1/genetics , Transcription Factor DP1/metabolism
19.
BMC Bioinformatics ; 16: 74, 2015 Mar 07.
Article in English | MEDLINE | ID: mdl-25887857

ABSTRACT

BACKGROUND: Decoding the temporal control of gene expression patterns is key to the understanding of the complex mechanisms that govern developmental decisions during heart development. High-throughput methods have been employed to systematically study the dynamic and coordinated nature of cardiac differentiation at the global level with multiple dimensions. Therefore, there is a pressing need to develop a systems approach to integrate these data from individual studies and infer the dynamic regulatory networks in an unbiased fashion. RESULTS: We developed a two-step strategy to integrate data from (1) temporal RNA-seq, (2) temporal histone modification ChIP-seq, (3) transcription factor (TF) ChIP-seq and (4) gene perturbation experiments to reconstruct the dynamic network during heart development. First, we trained a logistic regression model to predict the probability (LR score) of any base being bound by 543 TFs with known positional weight matrices. Second, four dimensions of data were combined using a time-varying dynamic Bayesian network model to infer the dynamic networks at four developmental stages in the mouse [mouse embryonic stem cells (ESCs), mesoderm (MES), cardiac progenitors (CP) and cardiomyocytes (CM)]. Our method not only infers the time-varying networks between different stages of heart development, but it also identifies the TF binding sites associated with promoter or enhancers of downstream genes. The LR scores of experimentally verified ESCs and heart enhancers were significantly higher than random regions (p <10(-100)), suggesting that a high LR score is a reliable indicator for functional TF binding sites. Our network inference model identified a region with an elevated LR score approximately -9400 bp upstream of the transcriptional start site of Nkx2-5, which overlapped with a previously reported enhancer region (-9435 to -8922 bp). TFs such as Tead1, Gata4, Msx2, and Tgif1 were predicted to bind to this region and participate in the regulation of Nkx2-5 gene expression. Our model also predicted the key regulatory networks for the ESC-MES, MES-CP and CP-CM transitions. CONCLUSION: We report a novel method to systematically integrate multi-dimensional -omics data and reconstruct the gene regulatory networks. This method will allow one to rapidly determine the cis-modules that regulate key genes during cardiac differentiation.


Subject(s)
Gene Regulatory Networks , Heart/embryology , Muscle Development/genetics , Animals , Bayes Theorem , Binding Sites , Cell Differentiation/genetics , Chromatin Immunoprecipitation , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Histones/metabolism , Mice , Myoblasts, Cardiac/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Promoter Regions, Genetic , Sequence Analysis, RNA , Transcription Factors/metabolism
20.
J Biol Chem ; 290(15): 9614-25, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25694434

ABSTRACT

Mesoderm posterior 1 (Mesp1) is well recognized for its role in cardiac development, although it is expressed broadly in mesodermal lineages. We have previously demonstrated important roles for Mesp1 and Ets variant 2 (Etv2) during lineage specification, but their relationship has not been defined. This study reveals that Mesp1 binds to the proximal promoter and transactivates Etv2 gene expression via the CRE motif. We also demonstrate the protein-protein interaction between Mesp1 and cAMP-responsive element binding protein 1 (Creb1) in vitro and in vivo. Utilizing transgenesis, lineage tracing, flow cytometry, and immunostaining technologies, we define the lineage relationship between Mesp1- and Etv2-expressing cell populations. We observe that the majority of Etv2-EYFP(+) cells are derived from Mesp1-Cre(+) cells in both the embryo and yolk sac. Furthermore, we observe that the conditional deletion of Etv2, using a Mesp1-Cre transgenic strategy, results in vascular and hematopoietic defects similar to those observed in the global deletion of Etv2 and that it has embryonic lethality by embryonic day 9.5. In summary, our study supports the hypothesis that Mesp1 is a direct upstream transactivator of Etv2 during embryogenesis and that Creb1 is an important cofactor of Mesp1 in the transcriptional regulation of Etv2 gene expression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Transcription Factors/genetics , Transcriptional Activation , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Cell Line , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , NIH 3T3 Cells , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL