Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Pediatr Res ; 95(3): 775-784, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37454186

ABSTRACT

BACKGROUND: The COVID-19 pandemic affected home and work routines, which may exacerbate existing academic professional disparities. Objectives were to describe the impact of the pandemic on pediatric faculty's work productivity, identify groups at risk for widening inequities, and explore mitigation strategies. METHODS: A cross-sectional study of faculty members was conducted at nine U.S. pediatric departments. Responses were analyzed by demographics, academic rank, and change in home caregiving responsibility. RESULTS: Of 5791 pediatric faculty members eligible, 1504 (26%) completed the survey. The majority were female (64%), over 40 years old (60%), and assistant professors (47%). Only 7% faculty identified as underrepresented in medicine. Overall 41% reported an increase in caregiving during the pandemic. When comparing clinical, administrative, research, and teaching activities, faculty reported worse 1-year outlook for research activities. Faculty with increased caregiving responsibilities were more likely to report concerns over delayed promotion and less likely to have a favorable outlook regarding clinical and research efforts. Participants identified preferred strategies to mitigate challenges. CONCLUSIONS: The COVID-19 pandemic negatively impacted pediatric faculty productivity with the greatest effects on those with increased caregiving responsibilities. COVID-19 was particularly disruptive to research outlook. Mitigation strategies are needed to minimize the long-term impacts on academic pediatric careers. IMPACT: The COVID-19 pandemic most negatively impacted work productivity of academic pediatric faculty with caregiving responsibilities. COVID-19 was particularly disruptive to short-term (1-year) research outlook among pediatric faculty. Faculty identified mitigation strategies to minimize the long-term impacts of the pandemic on academic pediatric career pathways.


Subject(s)
COVID-19 , Pandemics , Humans , Male , Female , Child , Adult , Cross-Sectional Studies , Faculty, Medical , Schools
2.
Arterioscler Thromb Vasc Biol ; 43(7): e231-e237, 2023 07.
Article in English | MEDLINE | ID: mdl-37128914

ABSTRACT

BACKGROUND: The goal of this study was to identify and characterize cell-cell interactions that facilitate endothelial tip cell fusion downstream of BMP (bone morphogenic protein)-mediated venous plexus formation. METHODS: High resolution and time-lapse imaging of transgenic reporter lines and loss-of-function studies were carried out to study the involvement of mesenchymal stromal cells during venous angiogenesis. RESULTS: BMP-responsive stromal cells facilitate timely and precise fusion of venous tip cells during developmental angiogenesis. CONCLUSIONS: Stromal cells are required for anastomosis of venous tip cells in the embryonic caudal hematopoietic tissue.


Subject(s)
Bone Morphogenetic Proteins , Mesenchymal Stem Cells , Animals , Cell Fusion , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Animals, Genetically Modified , Cell Communication , Stromal Cells/metabolism
4.
Nucleic Acids Res ; 49(22): e131, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34554233

ABSTRACT

CRISPR-Cas9 sgRNA libraries have transformed functional genetic screening and have enabled several innovative methods that rely on simultaneously targeting numerous genetic loci. Such libraries could be used in a vast number of biological systems and in the development of new technologies, but library generation is hindered by the cost, time, and sequence data required for sgRNA library synthesis. Here, we describe a rapid enzymatic method for generating robust, variant-matched libraries from any source of cDNA in under 3 h. This method, which we have named SLALOM, utilizes a custom sgRNA scaffold sequence and a novel method for detaching oligonucleotides from solid supports by a strand displacing polymerase. With this method, we constructed libraries targeting the E. coli genome and the transcriptome of developing zebrafish hearts, demonstrating its ability to expand the reach of CRISPR technology and facilitate methods requiring custom libraries.


Subject(s)
CRISPR-Cas Systems , Animals , CRISPR-Associated Proteins , DNA Restriction Enzymes , DNA-Directed DNA Polymerase , Escherichia coli/genetics , Fluorescent Dyes , Genetic Techniques , Genome , Green Fluorescent Proteins , Humans , Myocardium/metabolism , Oligonucleotides , RNA/biosynthesis , Transcriptome , Zebrafish
5.
Development ; 146(24)2019 12 16.
Article in English | MEDLINE | ID: mdl-31784460

ABSTRACT

Multipotent progenitor populations are necessary for generating diverse tissue types during embryogenesis. We show the RNA polymerase-associated factor 1 complex (Paf1C) is required to maintain multipotent progenitors of the neural crest (NC) lineage in zebrafish. Mutations affecting each Paf1C component result in near-identical NC phenotypes; alyron mutant embryos carrying a null mutation in paf1 were analyzed in detail. In the absence of zygotic paf1 function, definitive premigratory NC progenitors arise but fail to maintain expression of the sox10 specification gene. The mutant NC progenitors migrate aberrantly and fail to differentiate appropriately. Blood and germ cell progenitor development is affected similarly. Development of mutant NC could be rescued by additional loss of positive transcription elongation factor b (P-TEFb) activity, a key factor in promoting transcription elongation. Consistent with the interpretation that inhibiting/delaying expression of some genes is essential for maintaining progenitors, mutant embryos lacking the CDK9 kinase component of P-TEFb exhibit a surfeit of NC progenitors and their derivatives. We propose Paf1C and P-TEFb act antagonistically to regulate the timing of the expression of genes needed for NC development.


Subject(s)
Cell Lineage/genetics , Multipotent Stem Cells/physiology , Neural Crest/cytology , Neural Stem Cells/physiology , Nuclear Proteins/physiology , Positive Transcriptional Elongation Factor B/physiology , Transcription Factors/physiology , Zebrafish Proteins/physiology , Animals , Animals, Genetically Modified , Body Patterning/genetics , Cell Differentiation/genetics , Cyclin-Dependent Kinase 9/genetics , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Multipotent Stem Cells/cytology , Multiprotein Complexes/genetics , Multiprotein Complexes/physiology , Neural Crest/physiology , Neural Stem Cells/cytology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Positive Transcriptional Elongation Factor B/antagonists & inhibitors , Positive Transcriptional Elongation Factor B/metabolism , RNA Polymerase II/metabolism , Transcription Factors/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
6.
PLoS Biol ; 17(9): e3000087, 2019 09.
Article in English | MEDLINE | ID: mdl-31479440

ABSTRACT

Kabuki Syndrome patients have a spectrum of congenital disorders, including congenital heart defects, the primary determinant of mortality. Seventy percent of Kabuki Syndrome patients have mutations in the histone methyl-transferase KMT2D. However, the underlying mechanisms that drive these congenital disorders are unknown. Here, we generated and characterized zebrafish kmt2d null mutants that recapitulate the cardinal phenotypic features of Kabuki Syndrome, including microcephaly, palate defects, abnormal ear development, and cardiac defects. The cardiac phenotype consists of a previously unknown vasculogenesis defect that affects endocardium patterning and, consequently, heart ventricle lumen formation. Additionally, zebrafish kmt2d null mutants have angiogenesis defects depicted by abnormal aortic arch development, hyperactive ectopic blood vessel sprouting, and aberrant patterning of the brain vascular plexus. We demonstrate that zebrafish kmt2d null mutants have robust Notch signaling hyperactivation in endocardial and endothelial cells, including increased protein levels of the Notch transcription factor Rbpj. Our zebrafish Kabuki Syndrome model reveals a regulatory link between the Notch pathway and Kmt2d during endothelium and endocardium patterning and shows that pharmacological inhibition of Notch signaling rebalances Rbpj protein levels and rescues the cardiovascular phenotype by enhancing endothelial and endocardial cell proliferation and stabilizing endocardial patterning. Taken together, these findings demonstrate that Kmt2d regulates vasculogenesis and angiogenesis, provide evidence for interactions between Kmt2d and Notch signaling in Kabuki Syndrome, and suggest future directions for clinical research.


Subject(s)
Abnormalities, Multiple/etiology , Face/abnormalities , Hematologic Diseases/etiology , Histone-Lysine N-Methyltransferase/genetics , Neovascularization, Physiologic/genetics , Receptors, Notch/metabolism , Vestibular Diseases/etiology , Zebrafish Proteins/genetics , Abnormalities, Multiple/metabolism , Animals , Disease Models, Animal , Ear, Middle/abnormalities , Endothelial Cells/metabolism , Heart/embryology , Heart Defects, Congenital/genetics , Hematologic Diseases/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mutation , Palate/abnormalities , Phenotype , Receptors, Notch/antagonists & inhibitors , Vestibular Diseases/metabolism , Zebrafish , Zebrafish Proteins/metabolism
7.
Angiogenesis ; 23(2): 203-220, 2020 05.
Article in English | MEDLINE | ID: mdl-31828546

ABSTRACT

Hereditary hemorrhagic telangiectasia (HHT) is an autosomal-dominant vascular disorder characterized by development of high-flow arteriovenous malformations (AVMs) that can lead to stroke or high-output heart failure. HHT2 is caused by heterozygous mutations in ACVRL1, which encodes an endothelial cell bone morphogenetic protein (BMP) receptor, ALK1. BMP9 and BMP10 are established ALK1 ligands. However, the unique and overlapping roles of these ligands remain poorly understood. To define the physiologically relevant ALK1 ligand(s) required for vascular development and maintenance, we generated zebrafish harboring mutations in bmp9 and duplicate BMP10 paralogs, bmp10 and bmp10-like. bmp9 mutants survive to adulthood with no overt phenotype. In contrast, combined loss of bmp10 and bmp10-like results in embryonic lethal cranial AVMs indistinguishable from acvrl1 mutants. However, despite embryonic functional redundancy of bmp10 and bmp10-like, bmp10 encodes the only required Alk1 ligand in the juvenile-to-adult period. bmp10 mutants exhibit blood vessel abnormalities in anterior skin and liver, heart dysmorphology, and premature death, and vascular defects correlate with increased cardiac output. Together, our findings support a unique role for Bmp10 as a non-redundant Alk1 ligand required to maintain the post-embryonic vasculature and establish zebrafish bmp10 mutants as a model for AVM-associated high-output heart failure, which is an increasingly recognized complication of severe liver involvement in HHT2.


Subject(s)
Activin Receptors/metabolism , Blood Vessels/growth & development , Blood Vessels/physiology , Bone Morphogenetic Proteins/physiology , Neovascularization, Physiologic/genetics , Regeneration/genetics , Zebrafish Proteins/metabolism , Activin Receptors/genetics , Animals , Animals, Genetically Modified , Arteriovenous Malformations/genetics , Arteriovenous Malformations/metabolism , Arteriovenous Malformations/pathology , Bone Morphogenetic Proteins/genetics , Cell Differentiation/genetics , Embryo, Nonmammalian , Endothelial Cells/physiology , Gene Expression Regulation, Developmental , Signal Transduction/genetics , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology
8.
Development ; 144(19): 3487-3498, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28807900

ABSTRACT

During embryogenesis the heart forms as a linear tube that then undergoes multiple simultaneous morphogenetic events to obtain its mature shape. To understand the gene regulatory networks (GRNs) driving this phase of heart development, during which many congenital heart disease malformations likely arise, we conducted an RNA-seq timecourse in zebrafish from 30 hpf to 72 hpf and identified 5861 genes with altered expression. We clustered the genes by temporal expression pattern, identified transcription factor binding motifs enriched in each cluster, and generated a model GRN for the major gene batteries in heart morphogenesis. This approach predicted hundreds of regulatory interactions and found batteries enriched in specific cell and tissue types, indicating that the approach can be used to narrow the search for novel genetic markers and regulatory interactions. Subsequent analyses confirmed the GRN using two mutants, Tbx5 and nkx2-5, and identified sets of duplicated zebrafish genes that do not show temporal subfunctionalization. This dataset provides an essential resource for future studies on the genetic/epigenetic pathways implicated in congenital heart defects and the mechanisms of cardiac transcriptional regulation.


Subject(s)
Gene Expression Regulation, Developmental , Gene Regulatory Networks , Heart/embryology , Morphogenesis/genetics , Animals , Cluster Analysis , Gene Expression Profiling , Genes, Duplicate , Mice , Mutation/genetics , Nucleotide Motifs/genetics , Organ Specificity/genetics , Protein Binding , Sequence Analysis, RNA , Time Factors , Transcription Factors/metabolism , Zebrafish/embryology , Zebrafish/genetics
9.
Development ; 142(20): 3456-67, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26487777

ABSTRACT

Heparan sulfate proteoglycans (HSPGs) have long been implicated in a wide range of cell-cell signaling and cell-matrix interactions, both in vitro and in vivo in invertebrate models. Although many of the genes that encode HSPG core proteins and the biosynthetic enzymes that generate and modify HSPG sugar chains have not yet been analyzed by genetics in vertebrates, recent studies have shown that HSPGs do indeed mediate a wide range of functions in early vertebrate development, for example during left-right patterning and in cardiovascular and neural development. Here, we provide a comprehensive overview of the various roles of HSPGs in these systems and explore the concept of an instructive heparan sulfate sugar code for modulating vertebrate development.


Subject(s)
Carbohydrates/chemistry , Gene Expression Regulation, Developmental , Heparan Sulfate Proteoglycans/chemistry , Vertebrates/embryology , Animals , Axons/physiology , Body Patterning , Caenorhabditis elegans , Cardiovascular System/embryology , Cell Movement , Disulfides/chemistry , Glycosylphosphatidylinositols/chemistry , Heparitin Sulfate/metabolism , Humans , Ligands , Mice , Mice, Knockout , Nervous System/embryology , Neurons/metabolism , Signal Transduction , Vertebrates/physiology , Zebrafish
10.
Nat Methods ; 11(9): 966-970, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25028895

ABSTRACT

Current methods to isolate rare (1:10,000-1:100,000) bacterial artificial chromosome (BAC) recombinants require selectable markers. For seamless BAC mutagenesis, selectable markers need to be removed after isolation of recombinants through counterselection. Here we illustrate founder principle-driven enrichment (FPE), a simple method to rapidly isolate rare recombinants without using selectable markers, allowing one-step seamless BAC mutagenesis. As proof of principle, we isolated 1:100,000 seamless fluorescent protein-modified Nodal BACs and confirmed BAC functionality by generating fluorescent reporter mice. We also isolated small indel P1 phage-derived artificial chromosome (PAC) and BAC recombinants. Statistical analysis revealed that 1:100,000 recombinants can be isolated with <40 PCRs, and we developed a web-based calculator to optimize FPE.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Mutagenesis, Site-Directed/methods , Protein Engineering/methods , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Animals , Genetic Markers/genetics , Mice
11.
Dev Dyn ; 245(11): 1107-1123, 2016 11.
Article in English | MEDLINE | ID: mdl-27580238

ABSTRACT

BACKGROUND: 14-3-3ε plays an important role in the maturation of the compact ventricular myocardium by modulating the cardiomyocyte cell cycle via p27kip1 . However, additional cardiac defects are possible given the ubiquitous expression pattern of this protein. RESULTS: Germ line deletion of 14-3-3ε led to malalignment of both the outflow tract (OFT) and atrioventricular (AV) cushions, with resulting tricuspid stenosis and atresia, mitral valve abnormalities, and perimembranous ventricular septal defects (VSDs). We confirmed myocardial non-compaction and detected a spongy septum with muscular VSDs and blebbing of the epicardium. These defects were associated with abnormal patterning of p27kip1 expression in the subendocardial and possibly the epicardial cell populations. In addition to abnormal pharyngeal arch artery patterning, we found deep endocardial recesses and paucity of intramyocardial coronary vasculature as a result of defective coronary plexus remodeling. CONCLUSIONS: The malalignment of both endocardial cushions provides a new explanation for tricuspid and mitral valve defects, while myocardial non-compaction provides the basis for the abnormal coronary vasculature patterning. These abnormalities might arise from p27kip1 dysregulation and a resulting defect in epithelial-to-mesenchymal transformation. These data suggest that 14-3-3ε, in addition to left ventricular non-compaction (LVNC), might be linked to different forms of congenital heart disease (CHD). Developmental Dynamics 245:1107-1123, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
14-3-3 Proteins/metabolism , Endocardium/pathology , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Heart Ventricles/metabolism , Heart Ventricles/pathology , 14-3-3 Proteins/genetics , Animals , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Endocardium/metabolism , Gene Expression Regulation, Developmental , Mice , Myocardium/metabolism , Myocardium/pathology
12.
Genome Res ; 23(4): 687-97, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23299975

ABSTRACT

Forward genetic screens in model organisms are vital for identifying novel genes essential for developmental or disease processes. One drawback of these screens is the labor-intensive and sometimes inconclusive process of mapping the causative mutation. To leverage high-throughput techniques to improve this mapping process, we have developed a Mutation Mapping Analysis Pipeline for Pooled RNA-seq (MMAPPR) that works without parental strain information or requiring a preexisting SNP map of the organism, and adapts to differential recombination frequencies across the genome. MMAPPR accommodates the considerable amount of noise in RNA-seq data sets, calculates allelic frequency by Euclidean distance followed by Loess regression analysis, identifies the region where the mutation lies, and generates a list of putative coding region mutations in the linked genomic segment. MMAPPR can exploit RNA-seq data sets from isolated tissues or whole organisms that are used for gene expression and transcriptome analysis in novel mutants. We tested MMAPPR on two known mutant lines in zebrafish, nkx2.5 and tbx1, and used it to map two novel ENU-induced cardiovascular mutants, with mutations found in the ctr9 and cds2 genes. MMAPPR can be directly applied to other model organisms, such as Drosophila and Caenorhabditis elegans, that are amenable to both forward genetic screens and pooled RNA-seq experiments. Thus, MMAPPR is a rapid, cost-efficient, and highly automated pipeline, available to perform mutant mapping in any organism with a well-assembled genome.


Subject(s)
Chromosome Mapping , Mutation , RNA/genetics , Software , Alleles , Animals , Computational Biology/methods , Evolution, Molecular , Genes, Recessive , Internet , Polymorphism, Single Nucleotide , RNA/chemistry , Reproducibility of Results , Selection, Genetic , Sequence Analysis, RNA , Zebrafish/genetics
13.
Development ; 140(19): 4102-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24046323

ABSTRACT

Heparan sulfate proteoglycans (HSPGs) control many cellular processes and have been implicated in the regulation of left-right (LR) development by as yet unknown mechanisms. Using lineage-targeted knockdowns, we found that the transmembrane HSPG Syndecan 2 (Sdc2) regulates LR patterning through cell-autonomous functions in the zebrafish ciliated organ of asymmetry, Kupffer's vesicle (KV), including regulation of cell proliferation and adhesion, cilia length and asymmetric fluid flow. Exploring downstream pathways, we found that the cell signaling ligand Fgf2 is exclusively expressed in KV cell lineages, and is dependent on Sdc2 and the transcription factor Tbx16. Strikingly, Fgf2 controls KV morphogenesis but not KV cilia length, and KV morphogenesis in sdc2 morphants can be rescued by expression of fgf2 mRNA. Through an Fgf2-independent pathway, Sdc2 and Tbx16 also control KV ciliogenesis. Our results uncover a novel Sdc2-Tbx16-Fgf2 pathway that regulates epithelial cell morphogenesis.


Subject(s)
Cilia/metabolism , Embryo, Nonmammalian/metabolism , Epithelial Cells/cytology , Fibroblast Growth Factor 2/metabolism , Syndecan-2/metabolism , T-Box Domain Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Epithelial Cells/metabolism , Fibroblast Growth Factor 2/genetics , Immunohistochemistry , In Situ Hybridization , Syndecan-2/genetics , T-Box Domain Proteins/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
14.
Development ; 140(18): 3892-902, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23946439

ABSTRACT

As cells integrate molecular signals from their environment, cell surface receptors require modified proteoglycans for the robust activation of signaling pathways. Heparan sulfate proteoglycans (HSPGs) have long unbranched chains of repetitive disaccharide units that can be sulfated at specific positions by heparan sulfate O-sulfotransferase (OST) families. Here, we show that two members of the 3-OST family are required in distinct signaling pathways to control left-right (LR) patterning through control of Kupffer's vesicle (KV) cilia length and motility. 3-OST-5 functions in the fibroblast growth factor pathway to control cilia length via the ciliogenic transcription factors FoxJ1a and Rfx2. By contrast, a second 3-OST family member, 3-OST-6, does not regulate cilia length, but regulates cilia motility via kinesin motor molecule (Kif3b) expression and cilia arm dynein assembly. Thus, two 3-OST family members cell-autonomously control LR patterning through distinct pathways that regulate KV fluid flow. We propose that individual 3-OST isozymes create distinct modified domains or 'glycocodes' on cell surface proteoglycans, which in turn regulate the response to diverse cell signaling pathways.


Subject(s)
Cilia/enzymology , Sulfotransferases/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animal Structures/drug effects , Animal Structures/metabolism , Animals , Body Patterning/drug effects , Cilia/drug effects , Cilia/ultrastructure , Dyneins/metabolism , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/ultrastructure , Fibroblast Growth Factors/metabolism , Kinesins/metabolism , Models, Biological , Morpholinos/pharmacology , Movement/drug effects , Signal Transduction/drug effects , Transcription Factors/metabolism , Zebrafish/embryology
15.
Development ; 140(7): 1550-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23482490

ABSTRACT

Motile cilia perform crucial functions during embryonic development and throughout adult life. Development of organs containing motile cilia involves regulation of cilia formation (ciliogenesis) and formation of a luminal space (lumenogenesis) in which cilia generate fluid flows. Control of ciliogenesis and lumenogenesis is not yet fully understood, and it remains unclear whether these processes are coupled. In the zebrafish embryo, lethal giant larvae 2 (lgl2) is expressed prominently in ciliated organs. Lgl proteins are involved in establishing cell polarity and have been implicated in vesicle trafficking. Here, we identified a role for Lgl2 in development of ciliated epithelia in Kupffer's vesicle, which directs left-right asymmetry of the embryo; the otic vesicles, which give rise to the inner ear; and the pronephric ducts of the kidney. Using Kupffer's vesicle as a model ciliated organ, we found that depletion of Lgl2 disrupted lumen formation and reduced cilia number and length. Immunofluorescence and time-lapse imaging of Kupffer's vesicle morphogenesis in Lgl2-deficient embryos suggested cell adhesion defects and revealed loss of the adherens junction component E-cadherin at lateral membranes. Genetic interaction experiments indicate that Lgl2 interacts with Rab11a to regulate E-cadherin and mediate lumen formation that is uncoupled from cilia formation. These results uncover new roles and interactions for Lgl2 that are crucial for both lumenogenesis and ciliogenesis and indicate that these processes are genetically separable in zebrafish.


Subject(s)
Cilia/physiology , Kupffer Cells/physiology , Morphogenesis/genetics , Zebrafish Proteins/physiology , Zebrafish , Animals , Animals, Genetically Modified , Body Patterning/genetics , Cell Polarity/genetics , Cilia/genetics , Cilia/metabolism , Embryo, Nonmammalian , Embryonic Development/genetics , Embryonic Development/physiology , Gene Expression Regulation, Developmental , Kupffer Cells/metabolism , Larva/genetics , Larva/metabolism , Morphogenesis/physiology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
16.
PLoS Biol ; 11(12): e1001727, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24311987

ABSTRACT

The 3-O-sulfotransferase (3-OST) family catalyzes rare modifications of glycosaminoglycan chains on heparan sulfate proteoglycans, yet their biological functions are largely unknown. Knockdown of 3-OST-7 in zebrafish uncouples cardiac ventricular contraction from normal calcium cycling and electrophysiology by reducing tropomyosin4 (tpm4) expression. Normal 3-OST-7 activity prevents the expansion of BMP signaling into ventricular myocytes, and ectopic activation of BMP mimics the ventricular noncontraction phenotype seen in 3-OST-7 depleted embryos. In 3-OST-7 morphants, ventricular contraction can be rescued by overexpression of tropomyosin tpm4 but not by troponin tnnt2, indicating that tpm4 serves as a lynchpin for ventricular sarcomere organization downstream of 3-OST-7. Contraction can be rescued by expression of 3-OST-7 in endocardium, or by genetic loss of bmp4. Strikingly, BMP misregulation seen in 3-OST-7 morphants also occurs in multiple cardiac noncontraction models, including potassium voltage-gated channel gene, kcnh2, affected in Romano-Ward syndrome and long-QT syndrome, and cardiac troponin T gene, tnnt2, affected in human cardiomyopathies. Together these results reveal 3-OST-7 as a key component of a novel pathway that constrains BMP signaling from ventricular myocytes, coordinates sarcomere assembly, and promotes cardiac contractile function.


Subject(s)
Bone Morphogenetic Proteins/physiology , Myocardial Contraction/physiology , Sulfotransferases/physiology , Zebrafish Proteins/physiology , Action Potentials/physiology , Animals , Gene Knockdown Techniques , Muscle Development/physiology , Myocytes, Cardiac/physiology , Sarcomeres/physiology , Signal Transduction/physiology , Tropomyosin/physiology , Zebrafish
17.
Circ J ; 80(8): 1795-803, 2016 Jul 25.
Article in English | MEDLINE | ID: mdl-27301409

ABSTRACT

BACKGROUND: Ventricular non-compaction is characterized by a thin layer of compact ventricular myocardium and it is an important abnormality in the mouse heart. It is reminiscent of left ventricular non-compaction, a fairly common human congenital cardiomyopathy. Non-compaction in transgenic mice has been classically evaluated by measuring the thickness of the compact myocardium through histological techniques involving image analysis of 2-dimensional (D) sections. Given the 3D nature of the heart, the aim of this study was to determine whether a technique for the non-destructive, 3D assessment of the mouse embryonic compact myocardium could be developed. METHODS AND RESULTS: Micro-computed tomography (micro-CT), in combination with iodine staining, enabled the differentiation of the trabecular from the compact myocardium in wild-type mice. The 3D and digital nature of the micro-CT data allowed computation anatomical techniques to be readily applied, which were demonstrated via construction of group atlases and atlas-based descriptive statistics. Finally, micro-CT was used to identify the presence of non-compaction in mice with a deletion of the cell cycle inhibitor protein, p27(Kip1). CONCLUSIONS: Iodine staining-enhanced micro-CT with computational anatomical analysis represents a valid addition to classical histology for the delineation of compact myocardial wall thickness in the mouse embryo. Given the quantitative 3D resolution of micro-CT, these approaches might provide helpful information for the analysis of non-compaction. (Circ J 2016; 80: 1795-1803).


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/deficiency , Embryo, Mammalian , Heart Defects, Congenital , Myocardium , X-Ray Microtomography , Animals , Embryo, Mammalian/diagnostic imaging , Embryo, Mammalian/embryology , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Humans , Mice , Mice, Knockout
18.
Dev Biol ; 386(1): 123-34, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24333178

ABSTRACT

Early disruption of FGF signaling alters left-right (LR) asymmetry throughout the embryo. Here we uncover a role for FGF signaling that specifically disrupts brain asymmetry, independent of normal lateral plate mesoderm (LPM) asymmetry. When FGF signaling is inhibited during mid-somitogenesis, asymmetrically expressed LPM markers southpaw and lefty2 are not affected. However, asymmetrically expressed brain markers lefty1 and cyclops become bilateral. We show that FGF signaling controls expression of six3b and six7, two transcription factors required for repression of asymmetric lefty1 in the brain. We found that Z0-1, atypical PKC (aPKC) and ß-catenin protein distribution revealed a midline structure in the forebrain that is dependent on a balance of FGF signaling. Ectopic activation of FGF signaling leads to overexpression of six3b, loss of organized midline adherins junctions and bilateral loss of lefty1 expression. Reducing FGF signaling leads to a reduction in six3b and six7 expression, an increase in cell boundary formation in the brain midline, and bilateral expression of lefty1. Together, these results suggest a novel role for FGF signaling in the brain to control LR asymmetry, six transcription factor expressions, and a midline barrier structure.


Subject(s)
Body Patterning , Brain/embryology , Brain/physiology , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Signal Transduction , Zebrafish Proteins/metabolism , Animals , Crosses, Genetic , Eye Proteins/metabolism , Genotype , Homeodomain Proteins/metabolism , In Situ Hybridization , Intracellular Signaling Peptides and Proteins/metabolism , Left-Right Determination Factors/metabolism , Nerve Tissue Proteins/metabolism , Prosencephalon/embryology , Receptors, Fibroblast Growth Factor/metabolism , Transcription Factors , Zebrafish/embryology , beta Catenin/metabolism , Homeobox Protein SIX3
19.
Development ; 139(7): 1296-305, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22357927

ABSTRACT

O-sulfotransferases modify heparan sulfate proteoglycans (HSPGs) by catalyzing the transfer of a sulfate to a specific position on heparan sulfate glycosaminoglycan (GAG) chains. Although the roles of specific HSPG modifications have been described in cell culture and invertebrates, little is known about their functions or abilities to modulate specific cell signaling pathways in vertebrate development. Here, we report that 2-O-sulfotransferase (2-OST) is an essential component of canonical Wnt signaling in zebrafish development. 2-OST-deficient embryos have reduced GAG chain sulfation and are refractory to exogenous Wnt8 overexpression. Embryos in which maternally encoded 2-OST is knocked down have normal activation of several zygotic mesoderm, endoderm and ectoderm patterning genes, but have decreased deep cell adhesion and fail to initiate epiboly, which can be rescued by re-expression of 2-OST protein. Reduced cell adhesion and altered cell cycle regulation in 2-OST-deficient embryos are associated with decreased ß-catenin and E-cadherin protein levels at cell junctions, and these defects can be rescued by reactivation of the intracellular Wnt pathway, utilizing stabilized ß-catenin or dominant-negative Gsk3, but not by overexpression of Wnt8 ligand. Together, these results indicate that 2-OST functions within the Wnt pathway, downstream of Wnt ligand signaling and upstream of Gsk3ß and ß-catenin intracellular localization and function.


Subject(s)
Sulfotransferases/chemistry , Wnt Proteins/metabolism , Zebrafish Proteins/chemistry , Animals , Animals, Genetically Modified , Cadherins/metabolism , Cell Adhesion , Cell Cycle , Cytoskeletal Proteins/metabolism , Gene Expression Regulation, Developmental , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Heparan Sulfate Proteoglycans/chemistry , Ligands , Models, Biological , Signal Transduction , Sulfotransferases/physiology , Transcription, Genetic , Zebrafish , Zebrafish Proteins/metabolism , Zebrafish Proteins/physiology , beta Catenin/metabolism
20.
Am J Med Genet A ; 167A(12): 2975-84, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26284702

ABSTRACT

Wolff-Parkinson-White (WPW) syndrome is a common cause of supraventricular tachycardia that carries a risk of sudden cardiac death. To date, mutations in only one gene, PRKAG2, which encodes the 5'-AMP-activated protein kinase subunit γ-2, have been identified as causative for WPW. DNA samples from five members of a family with WPW were analyzed by exome sequencing. We applied recently designed prioritization strategies (VAAST/pedigree VAAST) coupled with an ontology-based algorithm (Phevor) that reduced the number of potentially damaging variants to 10: a variant in KCNE2 previously associated with Long QT syndrome was also identified. Of these 11 variants, only MYH6 p.E1885K segregated with the WPW phenotype in all affected individuals and was absent in 10 unaffected family members. This variant was predicted to be damaging by in silico methods and is not present in the 1,000 genome and NHLBI exome sequencing project databases. Screening of a replication cohort of 47 unrelated WPW patients did not identify other likely causative variants in PRKAG2 or MYH6. MYH6 variants have been identified in patients with atrial septal defects, cardiomyopathies, and sick sinus syndrome. Our data highlight the pleiotropic nature of phenotypes associated with defects in this gene.


Subject(s)
Exome , Wolff-Parkinson-White Syndrome/genetics , AMP-Activated Protein Kinases/genetics , Adult , Cardiac Myosins/genetics , Female , Genetic Loci , Humans , Male , Myosin Heavy Chains/genetics , Pedigree , Potassium Channels, Voltage-Gated/genetics , Wolff-Parkinson-White Syndrome/etiology
SELECTION OF CITATIONS
SEARCH DETAIL