Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Genes Dev ; 36(7-8): 468-482, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35450884

ABSTRACT

The nucleosome remodeling and deacetylase (NuRD) complex is one of the central chromatin remodeling complexes that mediates gene repression. NuRD is essential for numerous developmental events, including heart development. Clinical and genetic studies have provided direct evidence for the role of chromodomain helicase DNA-binding protein 4 (CHD4), the catalytic component of NuRD, in congenital heart disease (CHD), including atrial and ventricular septal defects. Furthermore, it has been demonstrated that CHD4 is essential for mammalian cardiomyocyte formation and function. A key unresolved question is how CHD4/NuRD is localized to specific cardiac target genes, as neither CHD4 nor NuRD can directly bind DNA. Here, we coupled a bioinformatics-based approach with mass spectrometry analyses to demonstrate that CHD4 interacts with the core cardiac transcription factors GATA4, NKX2-5, and TBX5 during embryonic heart development. Using transcriptomics and genome-wide occupancy data, we characterized the genomic landscape of GATA4, NKX2-5, and TBX5 repression and defined the direct cardiac gene targets of the GATA4-CHD4, NKX2-5-CHD4, and TBX5-CHD4 complexes. These data were used to identify putative cis-regulatory elements controlled by these complexes. We genetically interrogated two of these silencers in vivo: Acta1 and Myh11 We show that deletion of these silencers leads to inappropriate skeletal and smooth muscle gene misexpression, respectively, in the embryonic heart. These results delineate how CHD4/NuRD is localized to specific cardiac loci and explicates how mutations in the broadly expressed CHD4 protein lead to cardiac-specific disease states.


Subject(s)
DNA Helicases , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Animals , DNA Helicases/metabolism , Genes, Homeobox , Mammals/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Myocytes, Cardiac/metabolism , Nucleosomes , Transcription Factors/genetics
2.
Development ; 151(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38619323

ABSTRACT

Regulation of chromatin states is essential for proper temporal and spatial gene expression. Chromatin states are modulated by remodeling complexes composed of components that have enzymatic activities. CHD4 is the catalytic core of the nucleosome remodeling and deacetylase (NuRD) complex, which represses gene transcription. However, it remains to be determined how CHD4, a ubiquitous enzyme that remodels chromatin structure, functions in cardiomyocytes to maintain heart development. In particular, whether other proteins besides the NuRD components interact with CHD4 in the heart is controversial. Using quantitative proteomics, we identified that CHD4 interacts with SMYD1, a striated muscle-restricted histone methyltransferase that is essential for cardiomyocyte differentiation and cardiac morphogenesis. Comprehensive transcriptomic and chromatin accessibility studies of Smyd1 and Chd4 null embryonic mouse hearts revealed that SMYD1 and CHD4 repress a group of common genes and pathways involved in glycolysis, response to hypoxia, and angiogenesis. Our study reveals a mechanism by which CHD4 functions during heart development, and a previously uncharacterized mechanism regarding how SMYD1 represses cardiac transcription in the developing heart.


Subject(s)
DNA Helicases , DNA-Binding Proteins , Gene Expression Regulation, Developmental , Heart , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Myocytes, Cardiac , Transcription Factors , Animals , Humans , Mice , Cell Differentiation/genetics , Chromatin/metabolism , Glycolysis/genetics , Heart/embryology , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mice, Knockout , Muscle Proteins/metabolism , Muscle Proteins/genetics , Myocytes, Cardiac/metabolism , Proteomics , Transcription, Genetic
3.
Development ; 150(21)2023 11 01.
Article in English | MEDLINE | ID: mdl-37756602

ABSTRACT

T-box transcription factor 20 (Tbx20) plays a multifaceted role in cardiac morphogenesis and controls a broad gene regulatory network. However, the mechanism by which Tbx20 activates and represses target genes in a tissue-specific and temporal manner remains unclear. Studies show that Tbx20 directly interacts with the Transducin-like Enhancer of Split (TLE) family of proteins to mediate transcriptional repression. However, a function for the Tbx20-TLE transcriptional repression complex during heart development has yet to be established. We created a mouse model with a two amino acid substitution in the Tbx20 EH1 domain, thereby disrupting the Tbx20-TLE interaction. Disruption of this interaction impaired crucial morphogenic events, including cardiac looping and chamber formation. Transcriptional profiling of Tbx20EH1Mut hearts and analysis of putative direct targets revealed misexpression of the retinoic acid pathway and cardiac progenitor genes. Further, we show that altered cardiac progenitor development and function contribute to the severe cardiac defects in our model. Our studies indicate that TLE-mediated repression is a primary mechanism by which Tbx20 controls gene expression.


Subject(s)
T-Box Domain Proteins , Transducin , Animals , Mice , Transducin/genetics , Transducin/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Gene Expression Regulation, Developmental , Heart , Regulatory Sequences, Nucleic Acid
4.
Circ Res ; 133(1): 48-67, 2023 06 23.
Article in English | MEDLINE | ID: mdl-37254794

ABSTRACT

BACKGROUND: Left ventricular noncompaction (LVNC) is a prevalent cardiomyopathy associated with excessive trabeculation and thin compact myocardium. Patients with LVNC are vulnerable to cardiac dysfunction and at high risk of sudden death. Although sporadic and inherited mutations in cardiac genes are implicated in LVNC, understanding of the mechanisms responsible for human LVNC is limited. METHODS: We screened the complete exome sequence database of the Pediatrics Cardiac Genomics Consortium and identified a cohort with a de novo CHD4 (chromodomain helicase DNA-binding protein 4) proband, CHD4M202I, with congenital heart defects. We engineered a humanized mouse model of CHD4M202I (mouse CHD4M195I). Histological analysis, immunohistochemistry, flow cytometry, transmission electron microscopy, and echocardiography were used to analyze cardiac anatomy and function. Ex vivo culture, immunopurification coupled with mass spectrometry, transcriptional profiling, and chromatin immunoprecipitation were performed to deduce the mechanism of CHD4M195I-mediated ventricular wall defects. RESULTS: CHD4M195I/M195I mice developed biventricular hypertrabeculation and noncompaction and died at birth. Proliferation of cardiomyocytes was significantly increased in CHD4M195I hearts, and the excessive trabeculation was associated with accumulation of ECM (extracellular matrix) proteins and a reduction of ADAMTS1 (ADAM metallopeptidase with thrombospondin type 1 motif 1), an ECM protease. We rescued the hyperproliferation and hypertrabeculation defects in CHD4M195I hearts by administration of ADAMTS1. Mechanistically, the CHD4M195I protein showed augmented affinity to endocardial BRG1 (SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily A, member 4). This enhanced affinity resulted in the failure of derepression of Adamts1 transcription such that ADAMTS1-mediated trabeculation termination was impaired. CONCLUSIONS: Our study reveals how a single mutation in the chromatin remodeler CHD4, in mice or humans, modulates ventricular chamber maturation and that cardiac defects associated with the missense mutation CHD4M195I can be attenuated by the administration of ADAMTS1.


Subject(s)
Isolated Noncompaction of the Ventricular Myocardium , Mutation, Missense , Humans , Animals , Child , Mice , Heart Ventricles , Causality , Mutation , Myocytes, Cardiac , Chromatin , Isolated Noncompaction of the Ventricular Myocardium/genetics , ADAMTS1 Protein/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics
5.
PLoS Biol ; 17(9): e3000437, 2019 09.
Article in English | MEDLINE | ID: mdl-31490923

ABSTRACT

Heart disease is the leading cause of death in the western world. Attaining a mechanistic understanding of human heart development and homeostasis and the molecular basis of associated disease states relies on the use of animal models. Here, we present the cardiac proteomes of 4 model vertebrates with dual circulatory systems: the pig (Sus scrofa), the mouse (Mus musculus), and 2 frogs (Xenopus laevis and Xenopus tropicalis). Determination of which proteins and protein pathways are conserved and which have diverged within these species will aid in our ability to choose the appropriate models for determining protein function and to model human disease. We uncover mammalian- and amphibian-specific, as well as species-specific, enriched proteins and protein pathways. Among these, we find and validate an enrichment in cell-cycle-associated proteins within Xenopus laevis. To further investigate functional units within cardiac proteomes, we develop a computational approach to profile the abundance of protein complexes across species. Finally, we demonstrate the utility of these data sets for predicting appropriate model systems for studying given cardiac conditions by testing the role of Kielin/chordin-like protein (Kcp), a protein found as enriched in frog hearts compared to mammals. We establish that germ-line mutations in Kcp in Xenopus lead to valve defects and, ultimately, cardiac failure and death. Thus, integrating these findings with data on proteins responsible for cardiac disease should lead to the development of refined, species-specific models for protein function and disease states.


Subject(s)
Evolution, Molecular , Myocardium/metabolism , Proteome , Animals , Cell Cycle , Female , Heart/growth & development , Heart Diseases/metabolism , Humans , Mass Spectrometry , Mice , Models, Cardiovascular , Sus scrofa , Xenopus Proteins/metabolism , Xenopus laevis
6.
Proc Natl Acad Sci U S A ; 115(26): 6727-6732, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29891665

ABSTRACT

Cardiac development relies on proper cardiomyocyte differentiation, including expression and assembly of cell-type-specific actomyosin subunits into a functional cardiac sarcomere. Control of this process involves not only promoting expression of cardiac sarcomere subunits but also repressing expression of noncardiac myofibril paralogs. This level of transcriptional control requires broadly expressed multiprotein machines that modify and remodel the chromatin landscape to restrict transcription machinery access. Prominent among these is the nucleosome remodeling and deacetylase (NuRD) complex, which includes the catalytic core subunit CHD4. Here, we demonstrate that direct CHD4-mediated repression of skeletal and smooth muscle myofibril isoforms is required for normal cardiac sarcomere formation, function, and embryonic survival early in gestation. Through transcriptomic and genome-wide analyses of CHD4 localization, we identified unique CHD4 binding sites in smooth muscle myosin heavy chain, fast skeletal α-actin, and the fast skeletal troponin complex genes. We further demonstrate that in the absence of CHD4, cardiomyocytes in the developing heart form a hybrid muscle cell that contains cardiac, skeletal, and smooth muscle myofibril components. These misexpressed paralogs intercalate into the nascent cardiac sarcomere to disrupt sarcomere formation and cause impaired cardiac function in utero. These results demonstrate the genomic and physiological requirements for CHD4 in mammalian cardiac development.


Subject(s)
DNA Helicases/physiology , Gene Expression Regulation, Developmental , Heart Defects, Congenital/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/physiology , Myocytes, Cardiac/physiology , Sarcomeres/physiology , Animals , DNA Helicases/chemistry , DNA Helicases/deficiency , Female , Gene Knockdown Techniques , Genes, Lethal , Heart/diagnostic imaging , Heart/embryology , Heart Defects, Congenital/diagnostic imaging , Heart Defects, Congenital/embryology , Heart Defects, Congenital/pathology , Male , Mice , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Myofibrils/metabolism , Myofibrils/pathology , Nucleosomes/metabolism , Nucleosomes/ultrastructure , Sarcomeres/ultrastructure , Transcription, Genetic , Ultrasonography, Prenatal
7.
Proc Natl Acad Sci U S A ; 115(45): E10615-E10624, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30352852

ABSTRACT

Codevelopment of the lungs and heart underlies key evolutionary innovations in the transition to terrestrial life. Cardiac specializations that support pulmonary circulation, including the atrial septum, are generated by second heart field (SHF) cardiopulmonary progenitors (CPPs). It has been presumed that transcription factors required in the SHF for cardiac septation, e.g., Tbx5, directly drive a cardiac morphogenesis gene-regulatory network. Here, we report instead that TBX5 directly drives Wnt ligands to initiate a bidirectional signaling loop between cardiopulmonary mesoderm and the foregut endoderm for endodermal pulmonary specification and, subsequently, atrial septation. We show that Tbx5 is required for pulmonary specification in mice and amphibians but not for swim bladder development in zebrafish. TBX5 is non-cell-autonomously required for pulmonary endoderm specification by directly driving Wnt2 and Wnt2b expression in cardiopulmonary mesoderm. TBX5 ChIP-sequencing identified cis-regulatory elements at Wnt2 sufficient for endogenous Wnt2 expression domains in vivo and required for Wnt2 expression in precardiac mesoderm in vitro. Tbx5 cooperated with Shh signaling to drive Wnt2b expression for lung morphogenesis. Tbx5 haploinsufficiency in mice, a model of Holt-Oram syndrome, caused a quantitative decrement of mesodermal-to-endodermal Wnt signaling and subsequent endodermal-to-mesodermal Shh signaling required for cardiac morphogenesis. Thus, Tbx5 initiates a mesoderm-endoderm-mesoderm signaling loop in lunged vertebrates that provides a molecular basis for the coevolution of pulmonary and cardiac structures required for terrestrial life.


Subject(s)
Evolution, Molecular , Heart/embryology , Lung/embryology , T-Box Domain Proteins/genetics , Wnt2 Protein/genetics , Animals , Enhancer Elements, Genetic , Gene Expression Profiling , Mice , Mice, Mutant Strains , Signal Transduction , Transcription, Genetic , Zebrafish/embryology
8.
PLoS Genet ; 13(9): e1007011, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28945738

ABSTRACT

By the age of 40, one in five adults without symptoms of cardiovascular disease are at risk for developing congestive heart failure. Within this population, dilated cardiomyopathy (DCM) remains one of the leading causes of disease and death, with nearly half of cases genetically determined. Though genetic and high throughput sequencing-based approaches have identified sporadic and inherited mutations in a multitude of genes implicated in cardiomyopathy, how combinations of asymptomatic mutations lead to cardiac failure remains a mystery. Since a number of studies have implicated mutations of the transcription factor TBX20 in congenital heart diseases, we investigated the underlying mechanisms, using an unbiased systems-based screen to identify novel, cardiac-specific binding partners. We demonstrated that TBX20 physically and genetically interacts with the essential transcription factor CASZ1. This interaction is required for survival, as mice heterozygous for both Tbx20 and Casz1 die post-natally as a result of DCM. A Tbx20 mutation associated with human familial DCM sterically interferes with the TBX20-CASZ1 interaction and provides a physical basis for how this human mutation disrupts normal cardiac function. Finally, we employed quantitative proteomic analyses to define the molecular pathways mis-regulated upon disruption of this novel complex. Collectively, our proteomic, biochemical, genetic, and structural studies suggest that the physical interaction between TBX20 and CASZ1 is required for cardiac homeostasis, and further, that reduction or loss of this critical interaction leads to DCM. This work provides strong evidence that DCM can be inherited through a digenic mechanism.


Subject(s)
Cardiomyopathy, Dilated/genetics , DNA-Binding Proteins/genetics , Heart Failure/genetics , T-Box Domain Proteins/genetics , Transcription Factors/genetics , Animals , Cardiomyopathy, Dilated/physiopathology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Heart Failure/physiopathology , Humans , Mice , Mutation , Proteomics , T-Box Domain Proteins/chemistry , T-Box Domain Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism
9.
Trends Genet ; 32(11): 707-716, 2016 11.
Article in English | MEDLINE | ID: mdl-27717505

ABSTRACT

Congenital heart defects remain a leading cause of infant mortality in the western world, despite decades of research focusing on cardiovascular development and disease. With the recent emergence of several high-throughput technologies including RNA sequencing, chromatin-immunoprecipitation-coupled sequencing, mass-spectrometry-based proteomics analyses, and the numerous variations of these strategies, investigations into cardiac development have been transformed from candidate-based studies into whole-genome, -transcriptome, and -proteome undertakings. In this review, we discuss several reports that have emerged from our laboratory and others over the past 5 years that emphasize the versatility of large dataset-based investigations of cardiogenic transcription factors, from phenotypic validations and new gene implications to the identification of novel roles of well-studied transcriptional regulators.


Subject(s)
Cardiovascular Diseases/genetics , Cardiovascular System/growth & development , Gene Expression Regulation/genetics , Genomics , Chromatin/genetics , Genome, Human , High-Throughput Nucleotide Sequencing , Humans , Proteome/genetics , Transcriptome/genetics
10.
Development ; 143(5): 831-40, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26811386

ABSTRACT

The development of the vertebrate embryonic heart occurs by hyperplastic growth as well as the incorporation of cells from tissues outside of the initial heart field. Amongst these tissues is the epicardium, a cell structure that develops from the precursor proepicardial organ on the right side of the septum transversum caudal to the developing heart. During embryogenesis, cells of the proepicardial organ migrate, adhere and envelop the maturing heart, forming the epicardium. The cells of the epicardium then delaminate and incorporate into the heart giving rise to cardiac derivatives, including smooth muscle cells and cardiac fibroblasts. Here, we demonstrate that the LIM homeodomain protein Lhx9 is transiently expressed in Xenopus proepicardial cells and is essential for the position of the proepicardial organ on the septum transversum. Utilizing a small-molecule screen, we found that Lhx9 acts upstream of integrin-paxillin signaling and consistently demonstrate that either loss of Lhx9 or disruption of the integrin-paxillin pathway results in mis-positioning of the proepicardial organ and aberrant deposition of extracellular matrix proteins. This leads to a failure of proepicardial cell migration and adhesion to the heart, and eventual death of the embryo. Collectively, these studies establish a requirement for the Lhx9-integrin-paxillin pathway in proepicardial organ positioning and epicardial formation.


Subject(s)
Gene Expression Regulation, Developmental , Heart/embryology , Integrin alpha4/metabolism , LIM-Homeodomain Proteins/physiology , Pericardium/physiology , Transcription Factors/metabolism , Transcription Factors/physiology , Xenopus Proteins/metabolism , Xenopus Proteins/physiology , Animals , Animals, Genetically Modified , Cell Movement/physiology , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Immunohistochemistry , In Situ Hybridization , Integrins/metabolism , Mesoderm/metabolism , Paxillin/metabolism , Pericardium/embryology , Protein Structure, Tertiary , Xenopus laevis/embryology
11.
Development ; 142(11): 2037-47, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25953344

ABSTRACT

Organ growth occurs through the integration of external growth signals during the G1 phase of the cell cycle to initiate DNA replication. Although numerous growth factor signals have been shown to be required for the proliferation of cardiomyocytes, genetic studies have only identified a very limited number of transcription factors that act to regulate the entry of cardiomyocytes into S phase. Here, we report that the cardiac para-zinc-finger protein CASZ1 is expressed in murine cardiomyocytes. Genetic fate mapping with an inducible Casz1 allele demonstrates that CASZ1-expressing cells give rise to cardiomyocytes in the first and second heart fields. We show through the generation of a cardiac conditional null mutation that Casz1 is essential for the proliferation of cardiomyocytes in both heart fields and that loss of Casz1 leads to a decrease in cardiomyocyte cell number. We further report that the loss of Casz1 leads to a prolonged or arrested S phase, a decrease in DNA synthesis, an increase in phospho-RB and a concomitant decrease in the cardiac mitotic index. Taken together, these studies establish a role for CASZ1 in mammalian cardiomyocyte cell cycle progression in both the first and second heart fields.


Subject(s)
DNA-Binding Proteins/metabolism , G1 Phase , Heart/embryology , Mammals/embryology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , S Phase , Transcription Factors/metabolism , Animals , Cell Lineage , Cell Proliferation , Embryo, Mammalian/metabolism , Female , Integrases/metabolism , Male , Mice , Myocardium/cytology , Myocardium/metabolism , Myocardium/ultrastructure
12.
New Phytol ; 218(3): 1156-1166, 2018 05.
Article in English | MEDLINE | ID: mdl-28157265

ABSTRACT

The hypersensitive response (HR) is a localized programmed cell death phenomenon that occurs in response to pathogen recognition at the site of attempted invasion. Despite more than a century of research on HR, little is known about how it is so tightly regulated and how it can be contained spatially to a few cells. AtMC1 is an Arabidopsis thaliana plant metacaspase that positively regulates the HR. Here, we used an unbiased approach to identify new AtMC1 regulators. Immunoaffinity purification of AtMC1-containing complexes led us to the identification of the protease inhibitor AtSerpin1. Our data clearly showed that coimmunoprecipitation between AtMC1 and AtSerpin1 and formation of a complex between them was lost upon mutation of the AtMC1 catalytic site, and that the AtMC1 prodomain was not required for the interaction. AtSerpin1 blocked AtMC1 self-processing and inhibited AtMC1-mediated cell death. Our results constitute an in vivo example of a Serpin acting as a suicide inhibitor in plants, reminiscent of the activity of animal or viral serpins on immune/cell death regulators, including caspase-1. These results indicate a conserved function of a protease inhibitor on cell death regulators from different kingdoms with unrelated modes of action (i.e. caspases vs metacaspases).


Subject(s)
Apoptosis , Arabidopsis Proteins/metabolism , Arabidopsis/cytology , Arabidopsis/enzymology , Caspases/metabolism , Serpins/metabolism , Arabidopsis Proteins/chemistry , Biocatalysis , Caspases/chemistry , Immunoprecipitation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Domains
13.
Development ; 141(15): 3040-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24993940

ABSTRACT

The identification and characterization of the cellular and molecular pathways involved in the differentiation and morphogenesis of specific cell types of the developing heart are crucial to understanding the process of cardiac development and the pathology associated with human congenital heart disease. Here, we show that the cardiac transcription factor CASTOR (CASZ1) directly interacts with congenital heart disease 5 protein (CHD5), which is also known as tryptophan-rich basic protein (WRB), a gene located on chromosome 21 in the proposed region responsible for congenital heart disease in individuals with Down's syndrome. We demonstrate that loss of CHD5 in Xenopus leads to compromised myocardial integrity, improper deposition of basement membrane, and a resultant failure of hearts to undergo cell movements associated with cardiac formation. We further report that CHD5 is essential for CASZ1 function and that the CHD5-CASZ1 interaction is necessary for cardiac morphogenesis. Collectively, these results establish a role for CHD5 and CASZ1 in the early stages of vertebrate cardiac development.


Subject(s)
Gene Expression Regulation, Developmental , Heart/embryology , Nerve Tissue Proteins/metabolism , Transcription Factors/metabolism , Xenopus Proteins/metabolism , Animals , Basement Membrane/metabolism , Cell Adhesion , Cell Movement , Heart Defects, Congenital/metabolism , Image Processing, Computer-Assisted , Morphogenesis , Myocardium/pathology , Myocytes, Cardiac/cytology , Phenotype , Protein Binding , Two-Hybrid System Techniques , Xenopus laevis
14.
Development ; 141(4): 962-73, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24496632

ABSTRACT

The proper dissection of the molecular mechanisms governing the specification and differentiation of specific cell types requires isolation of pure cell populations from heterogeneous tissues and whole organisms. Here, we describe a method for purification of nuclei from defined cell or tissue types in vertebrate embryos using INTACT (isolation of nuclei tagged in specific cell types). This method, previously developed in plants, flies and worms, utilizes in vivo tagging of the nuclear envelope with biotin and the subsequent affinity purification of the labeled nuclei. In this study we successfully purified nuclei of cardiac and skeletal muscle from Xenopus using this strategy. We went on to demonstrate the utility of this approach by coupling the INTACT approach with liquid chromatography-tandem mass spectrometry (LC-MS/MS) proteomic methodologies to profile proteins expressed in the nuclei of developing hearts. From these studies we have identified the Xenopus orthologs of 12 human proteins encoded by genes, which when mutated in human lead to congenital heart disease. Thus, by combining these technologies we are able to identify tissue-specific proteins that are expressed and required for normal vertebrate organ development.


Subject(s)
Cell Nucleus/metabolism , Cytological Techniques/methods , Muscle, Skeletal/metabolism , Myocardium/metabolism , Proteomics/methods , Xenopus/metabolism , Animals , Biotin , Chromatography, Liquid , DNA Primers/genetics , Microscopy, Fluorescence , Muscle, Skeletal/cytology , Myocardium/cytology , Nuclear Envelope/metabolism , Plasmids/genetics , Real-Time Polymerase Chain Reaction , Streptavidin , Tandem Mass Spectrometry , Xenopus/genetics
15.
Development ; 140(11): 2409-21, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23637334

ABSTRACT

The epicardium is a mesothelial cell layer essential for vertebrate heart development and pertinent for cardiac repair post-injury in the adult. The epicardium initially forms from a dynamic precursor structure, the proepicardial organ, from which cells migrate onto the heart surface. During the initial stage of epicardial development crucial epicardial-derived cell lineages are thought to be determined. Here, we define an essential requirement for transcription factor Tcf21 during early stages of epicardial development in Xenopus, and show that depletion of Tcf21 results in a disruption in proepicardial cell specification and failure to form a mature epithelial epicardium. Using a mass spectrometry-based approach we defined Tcf21 interactions and established its association with proteins that function as transcriptional co-repressors. Furthermore, using an in vivo systems-based approach, we identified a panel of previously unreported proepicardial precursor genes that are persistently expressed in the epicardial layer upon Tcf21 depletion, thereby confirming a primary role for Tcf21 in the correct determination of the proepicardial lineage. Collectively, these studies lead us to propose that Tcf21 functions as a transcriptional repressor to regulate proepicardial cell specification and the correct formation of a mature epithelial epicardium.


Subject(s)
Gene Expression Regulation, Developmental , Pericardium/embryology , Transcription Factors/physiology , Xenopus Proteins/physiology , Xenopus laevis/embryology , Animals , Animals, Genetically Modified , Cell Lineage , Cell Movement , DNA, Complementary/metabolism , HEK293 Cells , Humans , Mass Spectrometry , Pericardium/cytology , Phosphorylation , Tandem Mass Spectrometry , Transcription, Genetic , Xenopus laevis/metabolism
16.
Bioessays ; 36(3): 251-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24323945

ABSTRACT

The establishment of a functional vascular system requires multiple complex steps throughout embryogenesis, from endothelial cell (EC) specification to vascular patterning into venous and arterial hierarchies. Following the initial assembly of ECs into a network of cord-like structures, vascular expansion and remodeling occur rapidly through morphogenetic events including vessel sprouting, fusion, and pruning. In addition, vascular morphogenesis encompasses the process of lumen formation, critical for the transformation of cords into perfusable vascular tubes. Studies in mouse, zebrafish, frog, and human endothelial cells have begun to outline the cellular and molecular requirements underlying lumen formation. Although the lumen can be generated through diverse mechanisms, the coordinated participation of multiple conserved molecules including transcription factors, small GTPases, and adhesion and polarity proteins remains a fundamental principle, leading us closer to a more thorough understanding of this complex event.


Subject(s)
Blood Vessels/cytology , Blood Vessels/growth & development , Organogenesis , Animals , Cell Polarity , Cytoskeleton/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Monomeric GTP-Binding Proteins/metabolism
17.
Development ; 139(5): 948-57, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22278918

ABSTRACT

Noonan syndrome is one of the most common causes of human congenital heart disease and is frequently associated with missense mutations in the protein phosphatase SHP-2. Interestingly, patients with acute myelogenous leukemia (AML), acute lymphoblastic leukemia (ALL), juvenile myelomonocytic leukemia (JMML) and LEOPARD syndrome frequently carry a second, somatically introduced subset of missense mutations in SHP-2. To determine the cellular and molecular mechanisms by which SHP-2 regulates heart development and, thus, understand how Noonan-associated mutations affect cardiogenesis, we introduced SHP-2 encoding the most prevalent Noonan syndrome and JMML mutations into Xenopus embryos. Resulting embryos show a direct relationship between a Noonan SHP-2 mutation and its ability to cause cardiac defects in Xenopus; embryos expressing Noonan SHP-2 mutations exhibit morphologically abnormal hearts, whereas those expressing an SHP-2 JMML-associated mutation do not. Our studies indicate that the cardiac defects associated with the introduction of the Noonan-associated SHP-2 mutations are coupled with a delay or arrest of the cardiac cell cycle in M-phase and a failure of cardiomyocyte progenitors to incorporate into the developing heart. We show that these defects are a result of an underlying malformation in the formation and polarity of cardiac actin fibers and F-actin deposition. We show that these defects can be rescued in culture and in embryos through the inhibition of the Rho-associated, coiled-coil-containing protein kinase 1 (ROCK), thus demonstrating a direct relationship between SHP-2(N308D) and ROCK activation in the developing heart.


Subject(s)
Actin Cytoskeleton/metabolism , Heart , Myocardium/metabolism , Noonan Syndrome/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Xenopus laevis/embryology , rho-Associated Kinases/metabolism , Animals , Enzyme Activation , Heart/anatomy & histology , Heart/embryology , Humans , Mutation, Missense , Myocardium/cytology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/ultrastructure , Noonan Syndrome/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Xenopus laevis/anatomy & histology , rho-Associated Kinases/genetics
18.
Methods ; 66(3): 398-409, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-23792920

ABSTRACT

Advances in sequencing technology have significantly advanced the landscape of developmental biology research. The dissection of genetic networks in model and non-model organisms has been greatly enhanced with high-throughput sequencing technologies. RNA-seq has revolutionized the ability to perform developmental biology research in organisms without a published genome sequence. Here, we describe a protocol for developmental biologists to perform RNA-seq on dissected tissue or whole embryos. We start with the isolation of RNA and generation of sequencing libraries. We further show how to interpret and analyze the large amount of sequencing data that is generated in RNA-seq. We explore the abilities to examine differential expression, gene duplication, transcript assembly, alternative splicing and SNP discovery. For the purposes of this article, we use Xenopus laevis as the model organism to discuss uses of RNA-seq in an organism without a fully annotated genome sequence.


Subject(s)
Genome , Sequence Analysis, RNA/methods , Xenopus laevis/genetics , Animals , Developmental Biology/methods , Models, Animal , Xenopus laevis/growth & development
19.
Nat Genet ; 38(2): 228-33, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16380711

ABSTRACT

Understanding the molecular mechanisms that regulate cellular proliferation and differentiation is a central theme of developmental biology. MicroRNAs (miRNAs) are a class of regulatory RNAs of approximately 22 nucleotides that post-transcriptionally regulate gene expression. Increasing evidence points to the potential role of miRNAs in various biological processes. Here we show that miRNA-1 (miR-1) and miRNA-133 (miR-133), which are clustered on the same chromosomal loci, are transcribed together in a tissue-specific manner during development. miR-1 and miR-133 have distinct roles in modulating skeletal muscle proliferation and differentiation in cultured myoblasts in vitro and in Xenopus laevis embryos in vivo. miR-1 promotes myogenesis by targeting histone deacetylase 4 (HDAC4), a transcriptional repressor of muscle gene expression. By contrast, miR-133 enhances myoblast proliferation by repressing serum response factor (SRF). Our results show that two mature miRNAs, derived from the same miRNA polycistron and transcribed together, can carry out distinct biological functions. Together, our studies suggest a molecular mechanism in which miRNAs participate in transcriptional circuits that control skeletal muscle gene expression and embryonic development.


Subject(s)
Cell Differentiation , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Animals , Cell Proliferation , Cells, Cultured , Embryo, Nonmammalian/cytology , Gene Expression , Mice , Models, Biological , Myoblasts/cytology , Myoblasts/metabolism , Myocardium/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Xenopus/embryology
20.
Dev Dyn ; 243(7): 948-56, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24633745

ABSTRACT

BACKGROUND: The zinc-finger transcription factor CASZ1 is required for differentiation of a distinct population of cardiomyocytes during development. However, expression of Casz1 mRNA is detected throughout the developing heart, suggesting the spatial regulation of CASZ1 occurs at the protein level. Relatively little is known about posttranscriptional regulation of Casz1 in the heart. RESULTS: We generated antibodies that specifically recognize CASZ1 in developing Xenopus embryos, and performed immunofluorescence analysis of CASZ1 during cardiac development. CASZ1 was detected throughout the developing myocardium. CASZ1 was restricted to terminally differentiated cardiomyocytes, and was down-regulated in cells that re-enter the cell cycle. We determined that CASZ1 expression correlated with terminal differentiation in cardiac muscle cells, skeletal muscle cells, and lymph-heart musculature. CONCLUSIONS: This study indicates that spatially distinct expression of CASZ1 protein may be due to posttranscriptional control of Casz1 mRNA during cardiac development. The results of this study provide insights into the role of Casz1 in cardiac function and in the differentiation of other cell types, including skeletal muscle and lymph heart.


Subject(s)
Muscle Development/physiology , Muscle, Skeletal/metabolism , Myocardium/metabolism , Xenopus Proteins/metabolism , Animals , Cell Cycle/genetics , Cell Cycle/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Heart , Muscle Development/genetics , Xenopus , Xenopus Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL