Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Elife ; 122023 03 27.
Article in English | MEDLINE | ID: mdl-36971347

ABSTRACT

Histone acetylation is a pivotal epigenetic modification that controls chromatin structure and regulates gene expression. It plays an essential role in modulating zygotic transcription and cell lineage specification of developing embryos. While the outcomes of many inductive signals have been described to require enzymatic activities of histone acetyltransferases and deacetylases (HDACs), the mechanisms by which HDACs confine the utilization of the zygotic genome remain to be elucidated. Here, we show that histone deacetylase 1 (Hdac1) progressively binds to the zygotic genome from mid-blastula and onward. The recruitment of Hdac1 to the genome at blastula is instructed maternally. Cis-regulatory modules (CRMs) bound by Hdac1 possess epigenetic signatures underlying distinct functions. We highlight a dual function model of Hdac1 where Hdac1 not only represses gene expression by sustaining a histone hypoacetylation state on inactive chromatin, but also maintains gene expression through participating in dynamic histone acetylation-deacetylation cycles on active chromatin. As a result, Hdac1 maintains differential histone acetylation states of bound CRMs between different germ layers and reinforces the transcriptional program underlying cell lineage identities, both in time and space. Taken together, our study reveals a comprehensive role for Hdac1 during early vertebrate embryogenesis.


Subject(s)
Histone Deacetylase 1 , Histones , Histones/metabolism , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Chromatin/metabolism , Blastocyst/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Embryonic Development/genetics , Acetylation , Histone Deacetylase 2/metabolism
2.
Cell Rep ; 38(7): 110364, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35172134

ABSTRACT

Mesendodermal specification is one of the earliest events in embryogenesis, where cells first acquire distinct identities. Cell differentiation is a highly regulated process that involves the function of numerous transcription factors (TFs) and signaling molecules, which can be described with gene regulatory networks (GRNs). Cell differentiation GRNs are difficult to build because existing mechanistic methods are low throughput, and high-throughput methods tend to be non-mechanistic. Additionally, integrating highly dimensional data composed of more than two data types is challenging. Here, we use linked self-organizing maps to combine chromatin immunoprecipitation sequencing (ChIP-seq)/ATAC-seq with temporal, spatial, and perturbation RNA sequencing (RNA-seq) data from Xenopus tropicalis mesendoderm development to build a high-resolution genome scale mechanistic GRN. We recover both known and previously unsuspected TF-DNA/TF-TF interactions validated through reporter assays. Our analysis provides insights into transcriptional regulation of early cell fate decisions and provides a general approach to building GRNs using highly dimensional multi-omic datasets.


Subject(s)
Endoderm/embryology , Gene Regulatory Networks , Genomics , Mesoderm/embryology , Xenopus/embryology , Xenopus/genetics , Animals , Chromatin/metabolism , Consensus Sequence/genetics , DNA/metabolism , Gastrulation/genetics , Gene Expression Regulation, Developmental , Protein Binding , RNA/metabolism , Transcription Factors/metabolism , Transcription, Genetic
3.
Cold Spring Harb Protoc ; 2022(3)2022 03 01.
Article in English | MEDLINE | ID: mdl-34244352

ABSTRACT

CRISPR-Cas9 mutagenesis is being widely used to create targeted loss-of-function mutations in the diploid frog Xenopus tropicalis Here we describe a simple mutagenesis protocol using microinjection of Cas9 protein or mRNA, together with synthetic guide RNAs (sgRNAs) targeting specific DNA sequences, into the early embryo. Cas9-catalyzed double-strand breaks undergo error-prone repair, resulting in production of short insertions and/or deletions. Thus, careful selection of target sites can lead to mutations that impair normal function of the protein product. CRISPR-Cas9 can be used to create either mosaic loss-of-function Xenopus embryos that display F0 generation phenotypes or mutant lines for downstream analysis. In addition to describing how to mutagenize genes using CRISPR-Cas9, we also discuss a simple method to determine the mutagenesis efficiency, some potential problems that can arise, and possible solutions to overcome them. The protocol described here should be applicable to other amphibians and, in principle, many other organisms.


Subject(s)
CRISPR-Cas Systems , Chromosomes, Human, Y , Animals , CRISPR-Cas Systems/genetics , Gene Editing/methods , Humans , Male , Mosaicism , Mutagenesis , Phenotype , Xenopus/genetics
4.
JCI Insight ; 6(17)2021 09 08.
Article in English | MEDLINE | ID: mdl-34314389

ABSTRACT

Mitochondrial biogenesis and function are controlled by anterograde regulatory pathways involving more than 1000 nuclear-encoded proteins. Transcriptional networks controlling the nuclear-encoded mitochondrial genes remain to be fully elucidated. Here, we show that histone demethylase LSD1 KO from adult mouse liver (LSD1-LKO) reduces the expression of one-third of all nuclear-encoded mitochondrial genes and decreases mitochondrial biogenesis and function. LSD1-modulated histone methylation epigenetically regulates nuclear-encoded mitochondrial genes. Furthermore, LSD1 regulates gene expression and protein methylation of nicotinamide mononucleotide adenylyltransferase 1 (NMNAT1), which controls the final step of NAD+ synthesis and limits NAD+ availability in the nucleus. Lsd1 KO reduces NAD+-dependent SIRT1 and SIRT7 deacetylase activity, leading to hyperacetylation and hypofunctioning of GABPß and PGC-1α, the major transcriptional factor/cofactor for nuclear-encoded mitochondrial genes. Despite the reduced mitochondrial function in the liver, LSD1-LKO mice are protected from diet-induced hepatic steatosis and glucose intolerance, partially due to induction of hepatokine FGF21. Thus, LSD1 orchestrates a core regulatory network involving epigenetic modifications and NAD+ synthesis to control mitochondrial function and hepatokine production.


Subject(s)
Fatty Liver/genetics , Fibroblast Growth Factors/genetics , Gene Expression Regulation , Genes, Mitochondrial/genetics , Histone Demethylases/genetics , Liver/metabolism , RNA/genetics , Animals , Cells, Cultured , Epigenesis, Genetic , Fatty Liver/metabolism , Fatty Liver/pathology , Fibroblast Growth Factors/biosynthesis , Histone Demethylases/biosynthesis , Liver/pathology , Mice , Signal Transduction
5.
Curr Top Dev Biol ; 145: 167-204, 2021.
Article in English | MEDLINE | ID: mdl-34074529

ABSTRACT

The fertilized frog egg contains all the materials needed to initiate development of a new organism, including stored RNAs and proteins deposited during oogenesis, thus the earliest stages of development do not require transcription. The onset of transcription from the zygotic genome marks the first genetic switch activating the gene regulatory network that programs embryonic development. Zygotic genome activation occurs after an initial phase of transcriptional quiescence that continues until the midblastula stage, a period called the midblastula transition, which was first identified in Xenopus. Activation of transcription is programmed by maternally supplied factors and is regulated at multiple levels. A similar switch exists in most animals and is of great interest both to developmental biologists and to those interested in understanding nuclear reprogramming. Here we review in detail our knowledge on this major switch in transcription in Xenopus and place recent discoveries in the context of a decades old problem.


Subject(s)
Genome/genetics , Xenopus laevis/embryology , Xenopus laevis/genetics , Zygote/metabolism , Animals , Oogenesis , Zygote/cytology
6.
Elife ; 92020 09 07.
Article in English | MEDLINE | ID: mdl-32894225

ABSTRACT

Lineage specification is governed by gene regulatory networks (GRNs) that integrate the activity of signaling effectors and transcription factors (TFs) on enhancers. Sox17 is a key transcriptional regulator of definitive endoderm development, and yet, its genomic targets remain largely uncharacterized. Here, using genomic approaches and epistasis experiments, we define the Sox17-governed endoderm GRN in Xenopus gastrulae. We show that Sox17 functionally interacts with the canonical Wnt pathway to specify and pattern the endoderm while repressing alternative mesectoderm fates. Sox17 and ß-catenin co-occupy hundreds of key enhancers. In some cases, Sox17 and ß-catenin synergistically activate transcription apparently independent of Tcfs, whereas on other enhancers, Sox17 represses ß-catenin/Tcf-mediated transcription to spatially restrict gene expression domains. Our findings establish Sox17 as a tissue-specific modifier of Wnt responses and point to a novel paradigm where genomic specificity of Wnt/ß-catenin transcription is determined through functional interactions between lineage-specific Sox TFs and ß-catenin/Tcf transcriptional complexes. Given the ubiquitous nature of Sox TFs and Wnt signaling, this mechanism has important implications across a diverse range of developmental and disease contexts.


Subject(s)
Endoderm/metabolism , Gene Regulatory Networks/genetics , SOXF Transcription Factors/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/metabolism , Animals , Gastrula/metabolism , SOXF Transcription Factors/genetics , Wnt Proteins/genetics , Wnt Proteins/metabolism , Xenopus , beta Catenin/genetics
7.
iScience ; 23(7): 101314, 2020 Jul 24.
Article in English | MEDLINE | ID: mdl-32650116

ABSTRACT

Although Wnt/ß-catenin signaling is generally conserved and well understood, the regulatory mechanisms controlling context-specific direct Wnt target gene expression in development and disease are still unclear. The onset of zygotic gene transcription in early embryogenesis represents an ideal, accessible experimental system to investigate context-specific direct Wnt target gene regulation. We combine transcriptomics using RNA-seq with genome-wide ß-catenin association using ChIP-seq to identify stage-specific direct Wnt target genes. We propose coherent feedforward regulation involving two distinct classes of direct maternal Wnt target genes, which differ both in expression and persistence of ß-catenin association. We discover that genomic ß-catenin association overlaps with Foxh1-associated regulatory sequences and demonstrate that direct maternal Wnt target gene expression requires Foxh1 function and Nodal/Tgfß signaling. Our results support a new paradigm for direct Wnt target gene co-regulation with context-specific mechanisms that will inform future studies of embryonic development and more widely stem cell-mediated homeostasis and human disease.

8.
Cell Rep ; 27(10): 2962-2977.e5, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31167141

ABSTRACT

Elucidation of the sequence of events underlying the dynamic interaction between transcription factors and chromatin states is essential. Maternal transcription factors function at the top of the regulatory hierarchy to specify the primary germ layers at the onset of zygotic genome activation (ZGA). We focus on the formation of endoderm progenitor cells and examine the interactions between maternal transcription factors and chromatin state changes underlying the cell specification process. Endoderm-specific factors Otx1 and Vegt together with Foxh1 orchestrate endoderm formation by coordinated binding to select regulatory regions. These interactions occur before the deposition of enhancer histone marks around the regulatory regions, and these TFs recruit RNA polymerase II, regulate enhancer activity, and establish super-enhancers associated with important endodermal genes. Therefore, maternal transcription factors Otx1, Vegt, and Foxh1 combinatorially regulate the activity of super-enhancers, which in turn activate key lineage-specifying genes during ZGA.


Subject(s)
Forkhead Transcription Factors/metabolism , Genome , Otx Transcription Factors/metabolism , T-Box Domain Proteins/metabolism , Xenopus Proteins/metabolism , Zygote/metabolism , Animals , Binding Sites , Chromatin/metabolism , Endoderm/metabolism , Enhancer Elements, Genetic , Female , Forkhead Transcription Factors/genetics , Histones/genetics , Histones/metabolism , Male , Morpholinos/metabolism , Otx Transcription Factors/antagonists & inhibitors , Otx Transcription Factors/genetics , RNA Polymerase II/metabolism , T-Box Domain Proteins/genetics , Transcriptome , Xenopus/metabolism , Xenopus Proteins/antagonists & inhibitors , Xenopus Proteins/genetics
9.
Dev Cell ; 49(4): 643-650.e3, 2019 05 20.
Article in English | MEDLINE | ID: mdl-31112700

ABSTRACT

It has recently been reported that a common side effect of translation-blocking morpholino antisense oligonucleotides is the induction of a set of innate immune response genes in Xenopus embryos and that splicing-blocking morpholinos lead to unexpected off-target mis-splicing events. Here, we present an analysis of all publicly available Xenopus RNA sequencing (RNA-seq) data in a reexamination of the effects of translation-blocking morpholinos on the innate immune response. Our analysis does not support the authors' general conclusion, which was based on a limited number of RNA-seq datasets. Moreover, the strong induction of an immune response appears to be specific to the tbxt/tbxt2 morpholinos. The more comprehensive study presented here indicates that using morpholinos for targeted gene knockdowns remains of considerable value for the rapid identification of gene function.


Subject(s)
Immunity, Innate/immunology , Morpholinos/immunology , Morpholinos/metabolism , Animals , Embryonic Development/drug effects , Gene Knockdown Techniques , Immunity, Innate/physiology , Oligonucleotides, Antisense/genetics , RNA Splicing , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcriptome/genetics , Xenopus/embryology , Xenopus/genetics , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Xenopus laevis/genetics
10.
Cold Spring Harb Protoc ; 2019(4)2019 04 01.
Article in English | MEDLINE | ID: mdl-30131367

ABSTRACT

Transcriptional regulatory elements are typically found in relatively nucleosome-free genomic regions, often referred to as "open chromatin." Deoxyribonuclease I (DNase I) can digest nucleosome-depleted DNA (presumably bound by transcription factors), but DNA in nucleosomes or higher-order chromatin fibers is less accessible to the nuclease. The DNase-seq method uses high-throughput sequencing to permit the interrogation of DNase hypersensitive sites (DHSs) across the entire genome and does not require prior knowledge of histone modifications, transcription factor binding sites, or high quality antibodies to identify potentially active regions of chromatin. Here, discontinuous iodixanol gradients are used as a gentle preparation of the nuclei from Xenopus embryos. Short DNase I digestion times are followed by size selection of digested genomic DNA, yielding DHS fragments. These DNA fragments are subjected to real-time quantitative polymerase chain reaction (qPCR) and sequencing library construction. A library generation method and pipeline for analyzing DNase-seq data are also described.


Subject(s)
Chromatin/metabolism , Deoxyribonuclease I/metabolism , Xenopus/embryology , Animals , Embryo, Nonmammalian/metabolism , Regulatory Sequences, Nucleic Acid , Transcription, Genetic
11.
J Vis Exp ; (132)2018 02 01.
Article in English | MEDLINE | ID: mdl-29443056

ABSTRACT

The creation of mutant lines by genome editing is accelerating genetic analysis in many organisms. CRISPR/Cas9 methods have been adapted for use in the African clawed frog, Xenopus, a longstanding model organism for biomedical research. Traditional breeding schemes for creating homozygous mutant lines with CRISPR/Cas9-targeted mutagenesis have several time-consuming and laborious steps. To facilitate the creation of mutant embryos, particularly to overcome the obstacles associated with knocking out genes that are essential for embryogenesis, a new method called leapfrogging was developed. This technique leverages the robustness of Xenopus embryos to "cut and paste" embryological methods. Leapfrogging utilizes the transfer of primordial germ cells (PGCs) from efficiently-mutagenized donor embryos into PGC-ablated wildtype siblings. This method allows for the efficient mutation of essential genes by creating chimeric animals with wildtype somatic cells that carry a mutant germline. When two F0 animals carrying "leapfrog transplants" (i.e., mutant germ cells) are intercrossed, they produce homozygous, or compound heterozygous, null F1 embryos, thus saving a full generation time to obtain phenotypic data. Leapfrogging also provides a new approach for analyzing maternal effect genes, which are refractory to F0 phenotypic analysis following CRISPR/Cas9 mutagenesis. This manuscript details the method of leapfrogging, with special emphasis on how to successfully perform PGC transplantation.


Subject(s)
CRISPR-Cas Systems/genetics , Germ Cells/transplantation , Animals , Cell Transplantation , Gene Targeting/methods , Mutation , Xenopus laevis
12.
Semin Cell Dev Biol ; 66: 12-24, 2017 06.
Article in English | MEDLINE | ID: mdl-28341363

ABSTRACT

Germ layer formation is among the earliest differentiation events in metazoan embryos. In triploblasts, three germ layers are formed, among which the endoderm gives rise to the epithelial lining of the gut tube and associated organs including the liver, pancreas and lungs. In frogs (Xenopus), where early germ layer formation has been studied extensively, the process of endoderm specification involves the interplay of dozens of transcription factors. Here, we review the interactions between these factors, summarized in a transcriptional gene regulatory network (GRN). We highlight regulatory connections conserved between frog, fish, mouse, and human endodermal lineages. Especially prominent is the conserved role and regulatory targets of the Nodal signaling pathway and the T-box transcription factors, Vegt and Eomes. Additionally, we highlight network topologies and motifs, and speculate on their possible roles in development.


Subject(s)
Endoderm/embryology , Gene Regulatory Networks/genetics , Transcription Factors/metabolism , Xenopus Proteins/genetics , Xenopus/genetics , Animals , Cell Differentiation
13.
Dev Cell ; 40(6): 595-607.e4, 2017 03 27.
Article in English | MEDLINE | ID: mdl-28325473

ABSTRACT

The interplay between transcription factors and chromatin dictates gene regulatory network activity. Germ layer specification is tightly coupled with zygotic gene activation and, in most metazoans, is dependent upon maternal factors. We explore the dynamic genome-wide interactions of Foxh1, a maternal transcription factor that mediates Nodal/TGF-ß signaling, with cis-regulatory modules (CRMs) during mesendodermal specification. Foxh1 marks CRMs during cleavage stages and recruits the co-repressor Tle/Groucho in the early blastula. We highlight a population of CRMs that are continuously occupied by Foxh1 and show that they are marked by H3K4me1, Ep300, and Fox/Sox/Smad motifs, suggesting interplay between these factors in gene regulation. We also propose a molecular "hand-off" between maternal Foxh1 and zygotic Foxa at these CRMs to maintain enhancer activation. Our findings suggest that Foxh1 functions at the top of a hierarchy of interactions by marking developmental genes for activation, beginning with the onset of zygotic gene expression.


Subject(s)
Endoderm/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Mesoderm/metabolism , Transcription Factors/metabolism , Xenopus Proteins/metabolism , Xenopus/embryology , Xenopus/genetics , Animals , Blastula/metabolism , Cleavage Stage, Ovum/metabolism , Co-Repressor Proteins/metabolism , Embryo, Nonmammalian/metabolism , Endoderm/embryology , Enhancer Elements, Genetic/genetics , Forkhead Transcription Factors/genetics , Genome , Histones/metabolism , Lysine/metabolism , Mesoderm/embryology , Methylation , Nodal Protein/metabolism , Protein Binding/genetics , RNA Polymerase II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Sequence Analysis, RNA , Signal Transduction/genetics , Transcription, Genetic , Xenopus/metabolism , Xenopus Proteins/genetics
14.
Dev Biol ; 426(2): 409-417, 2017 06 15.
Article in English | MEDLINE | ID: mdl-27475627

ABSTRACT

Gene regulatory networks (GRNs) involve highly combinatorial interactions between transcription factors and short sequence motifs in cis-regulatory modules of target genes to control cellular phenotypes. The GRNs specifying most cell types are largely unknown and are the subject of wide interest. A catalog of transcription factors is a valuable tool toward obtaining a deeper understanding of the role of these critical effectors in any biological setting. Here we present a comprehensive catalog of the transcription factors for the diploid frog Xenopus tropicalis. We identify 1235 genes encoding DNA-binding transcription factors, comparable to the numbers found in typical mammalian species. In detail, the repertoire of X. tropicalis transcription factor genes is nearly identical to human and mouse, with the exception of zinc finger family members, and a small number of species/lineage-specific gene duplications and losses relative to the mammalian repertoires. We applied this resource to the identification of transcription factors differentially expressed in the early gastrula stage embryo. We find transcription factor enrichment in Spemann's organizer, the ventral mesoderm, ectoderm and endoderm, and report 218 TFs that show regionalized expression patterns at this stage. Many of these have not been previously reported as expressed in the early embryo, suggesting thus far unappreciated roles for many transcription factors in the GRNs regulating early development. We expect our transcription factor catalog will facilitate myriad studies using Xenopus as a model system to understand basic biology and human disease.


Subject(s)
Gastrula/metabolism , Gene Expression Regulation, Developmental , Transcription Factors/biosynthesis , Xenopus Proteins/biosynthesis , Xenopus/metabolism , Animals , Base Sequence , Embryo, Nonmammalian/metabolism , Humans , Mice , Species Specificity , Transcription Factors/genetics , Xenopus/embryology , Xenopus/genetics , Xenopus Proteins/genetics
15.
Dev Biol ; 426(2): 401-408, 2017 06 15.
Article in English | MEDLINE | ID: mdl-27418388

ABSTRACT

Advances in RNA sequencing technologies have led to the surprising discovery that a vast number of transcripts emanate from regions of the genome that are not part of coding genes. Although some of the smaller ncRNAs such as microRNAs have well-characterized functions, the majority of long ncRNA (lncRNA) functions remain poorly understood. Understanding the significance of lncRNAs is an important challenge facing biology today. A powerful approach to uncovering the function of lncRNAs is to explore temporal and spatial expression profiling. This may be particularly useful for classes of lncRNAs that have developmentally important roles as the expression of such lncRNAs will be expected to be both spatially and temporally regulated during development. Here, we take advantage of our ultra-high frequency (temporal) sampling of Xenopus embryos to analyze gene expression trajectories of lncRNA transcripts over the first 3 days of development. We computationally identify 5689 potential single- and multi-exon lncRNAs. These lncRNAs demonstrate clear dynamic expression patterns. A subset of them displays highly correlative temporal expression profiles with respect to those of the neighboring genes. We also identified spatially localized lncRNAs in the gastrula stage embryo. These results suggest that lncRNAs have regulatory roles during early embryonic development.


Subject(s)
RNA, Long Noncoding/genetics , Xenopus/genetics , Animals , Embryo, Nonmammalian/metabolism , Exons/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Models, Genetic , RNA, Long Noncoding/biosynthesis , RNA, Long Noncoding/isolation & purification , Transcriptome , Xenopus/embryology
16.
Development ; 143(15): 2868-75, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27385011

ABSTRACT

CRISPR/Cas9 genome editing is revolutionizing genetic loss-of-function analysis but technical limitations remain that slow progress when creating mutant lines. First, in conventional genetic breeding schemes, mosaic founder animals carrying mutant alleles are outcrossed to produce F1 heterozygotes. Phenotypic analysis occurs in the F2 generation following F1 intercrosses. Thus, mutant analyses will require multi-generational studies. Second, when targeting essential genes, efficient mutagenesis of founders is often lethal, preventing the acquisition of mature animals. Reducing mutagenesis levels may improve founder survival, but results in lower, more variable rates of germline transmission. Therefore, an efficient approach to study lethal mutations would be useful. To overcome these shortfalls, we introduce 'leapfrogging', a method combining efficient CRISPR mutagenesis with transplantation of mutated primordial germ cells into a wild-type host. Tested using Xenopus tropicalis, we show that founders containing transplants transmit mutant alleles with high efficiency. F1 offspring from intercrosses between F0 animals that carry embryonic lethal alleles recapitulate loss-of-function phenotypes, circumventing an entire generation of breeding. We anticipate that leapfrogging will be transferable to other species.


Subject(s)
CRISPR-Cas Systems/physiology , Germ Cells/metabolism , Mutation/genetics , Animals , Anura , Blastula/cytology , Blastula/metabolism , CRISPR-Cas Systems/genetics , Embryo, Nonmammalian , Female , Germ Cells/cytology , Male , Mutagenesis , Transcription Activator-Like Effector Nucleases/genetics , Transcription Activator-Like Effector Nucleases/metabolism , Xenopus
17.
Cell Rep ; 14(3): 632-647, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26774488

ABSTRACT

Transcript regulation is essential for cell function, and misregulation can lead to disease. Despite technologies to survey the transcriptome, we lack a comprehensive understanding of transcript kinetics, which limits quantitative biology. This is an acute challenge in embryonic development, where rapid changes in gene expression dictate cell fate decisions. By ultra-high-frequency sampling of Xenopus embryos and absolute normalization of sequence reads, we present smooth gene expression trajectories in absolute transcript numbers. During a developmental period approximating the first 8 weeks of human gestation, transcript kinetics vary by eight orders of magnitude. Ordering genes by expression dynamics, we find that "temporal synexpression" predicts common gene function. Remarkably, a single parameter, the characteristic timescale, can classify transcript kinetics globally and distinguish genes regulating development from those involved in cellular metabolism. Overall, our analysis provides unprecedented insight into the reorganization of maternal and embryonic transcripts and redefines our ability to perform quantitative biology.


Subject(s)
RNA/metabolism , Transcriptome , Animals , Bayes Theorem , Embryo, Nonmammalian/metabolism , Expressed Sequence Tags , Gene Dosage , Kinetics , MicroRNAs/metabolism , Xenopus/growth & development , Xenopus/metabolism
18.
Development ; 141(23): 4537-47, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25359723

ABSTRACT

Nodal/TGFß signaling regulates diverse biological responses. By combining RNA-seq on Foxh1 and Nodal signaling loss-of-function embryos with ChIP-seq of Foxh1 and Smad2/3, we report a comprehensive genome-wide interaction between Foxh1 and Smad2/3 in mediating Nodal signaling during vertebrate mesendoderm development. This study significantly increases the total number of Nodal target genes regulated by Foxh1 and Smad2/3, and reinforces the notion that Foxh1-Smad2/3-mediated Nodal signaling directly coordinates the expression of a cohort of genes involved in the control of gene transcription, signaling pathway modulation and tissue morphogenesis during gastrulation. We also show that Foxh1 may function independently of Nodal signaling, in addition to its role as a transcription factor mediating Nodal signaling via Smad2/3. Finally, we propose an evolutionarily conserved interaction between Foxh1 and PouV, a mechanism observed in Pou5f1-mediated regulation of pluripotency in human embryonic stem and epiblast cells.


Subject(s)
Endoderm/embryology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental/physiology , Mesoderm/embryology , Transforming Growth Factor beta/metabolism , Xenopus Proteins/metabolism , Xenopus/embryology , Animals , Blotting, Western , Chromatin Immunoprecipitation , Computational Biology , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental/genetics , Gene Knockdown Techniques , Humans , Immunoprecipitation , Morpholinos/genetics , Nodal Protein/genetics , Nodal Protein/metabolism , Real-Time Polymerase Chain Reaction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Statistics, Nonparametric , Xenopus Proteins/genetics
19.
Methods Enzymol ; 546: 355-75, 2014.
Article in English | MEDLINE | ID: mdl-25398349

ABSTRACT

Xenopus tropicalis has been developed as a model organism for developmental biology, providing a system offering both modern genetics and classical embryology. Recently, the Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR-associated (CRISPR/Cas) system for genome modification has provided an additional tool for Xenopus researchers to achieve simple and efficient targeted mutagenesis. Here, we provide insights into experimental design and procedures permitting successful application of this technique to Xenopus researchers, and offer a general strategy for performing loss-of-function assays in F0 and subsequently F1 embryos.


Subject(s)
Gene Targeting/methods , Mutagenesis , Xenopus/embryology , Xenopus/genetics , Animals , Base Sequence , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Embryo, Nonmammalian/metabolism , Genetic Engineering/methods , Genome , Molecular Sequence Data , RNA, Guide, Kinetoplastida/genetics
20.
BMC Syst Biol ; 8: 3, 2014 Jan 08.
Article in English | MEDLINE | ID: mdl-24397936

ABSTRACT

BACKGROUND: During embryogenesis, signaling molecules produced by one cell population direct gene regulatory changes in neighboring cells and influence their developmental fates and spatial organization. One of the earliest events in the development of the vertebrate embryo is the establishment of three germ layers, consisting of the ectoderm, mesoderm and endoderm. Attempts to measure gene expression in vivo in different germ layers and cell types are typically complicated by the heterogeneity of cell types within biological samples (i.e., embryos), as the responses of individual cell types are intermingled into an aggregate observation of heterogeneous cell types. Here, we propose a novel method to elucidate gene regulatory circuits from these aggregate measurements in embryos of the frog Xenopus tropicalis using gene network inference algorithms and then test the ability of the inferred networks to predict spatial gene expression patterns. RESULTS: We use two inference models with different underlying assumptions that incorporate existing network information, an ODE model for steady-state data and a Markov model for time series data, and contrast the performance of the two models. We apply our method to both control and knockdown embryos at multiple time points to reconstruct the core mesoderm and endoderm regulatory circuits. Those inferred networks are then used in combination with known dorsal-ventral spatial expression patterns of a subset of genes to predict spatial expression patterns for other genes. Both models are able to predict spatial expression patterns for some of the core mesoderm and endoderm genes, but interestingly of different gene subsets, suggesting that neither model is sufficient to recapitulate all of the spatial patterns, yet they are complementary for the patterns that they do capture. CONCLUSION: The presented methodology of gene network inference combined with spatial pattern prediction provides an additional layer of validation to elucidate the regulatory circuits controlling the spatial-temporal dynamics in embryonic development.


Subject(s)
Computational Biology/methods , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Xenopus/embryology , Xenopus/genetics , Algorithms , Animals , Markov Chains
SELECTION OF CITATIONS
SEARCH DETAIL
...