Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Science ; 380(6646): 758-764, 2023 05 19.
Article in English | MEDLINE | ID: mdl-37200435

ABSTRACT

Zebrafish hearts can regenerate by replacing damaged tissue with new cardiomyocytes. Although the steps leading up to the proliferation of surviving cardiomyocytes have been extensively studied, little is known about the mechanisms that control proliferation and redifferentiation to a mature state. We found that the cardiac dyad, a structure that regulates calcium handling and excitation-contraction coupling, played a key role in the redifferentiation process. A component of the cardiac dyad called leucine-rich repeat-containing 10 (Lrrc10) acted as a negative regulator of proliferation, prevented cardiomegaly, and induced redifferentiation. We found that its function was conserved in mammalian cardiomyocytes. This study highlights the importance of the underlying mechanisms required for heart regeneration and their application to the generation of fully functional cardiomyocytes.


Subject(s)
Calcium , Heart , Myocytes, Cardiac , Regeneration , Sarcomeres , Zebrafish , Animals , Calcium/physiology , Cell Proliferation , Heart/physiology , Myocytes, Cardiac/physiology , Sarcomeres/physiology , Zebrafish/physiology
2.
J Cardiovasc Dev Dis ; 8(4)2021 Apr 20.
Article in English | MEDLINE | ID: mdl-33924051

ABSTRACT

Cardiomyopathies are a group of heterogeneous diseases that affect the muscles of the heart, leading to early morbidity and mortality in young and adults. Genetic forms of cardiomyopathy are caused predominantly by mutations in structural components of the cardiomyocyte sarcomeres, the contractile units of the heart, which includes cardiac Troponin T (TnT). Here, we generated mutations with CRISPR/Cas9 technology in the zebrafish tnnt2a gene, encoding cardiac TnT, at a mutational "hotspot" site to establish a zebrafish model for genetic cardiomyopathies. We found that a heterozygous tnnt2a mutation deleting Arginine at position 94 and Lysine at position 95 of TnT causes progressive cardiac structural changes resulting in heart failure. The cardiac remodeling is presented by an enlarged atrium, decreased ventricle size, increased myocardial stress as well as increased fibrosis. As early as five days post fertilization, larvae carrying the TnT RK94del mutation display diastolic dysfunction and impaired calcium dynamics related to increased Ca2+ sensitivity. In conclusion, adult zebrafish with a heterozygous TnT-RK94del mutation develop cardiomyopathy as seen in patients with TnT mutations and therefore represent a promising model to study disease mechanisms and to screen for putative therapeutic compounds.

3.
Dis Model Mech ; 11(10)2018 10 18.
Article in English | MEDLINE | ID: mdl-30355756

ABSTRACT

The zebrafish (Danio rerio) has become a popular vertebrate model organism to study organ formation and function due to its optical clarity and rapid embryonic development. The use of genetically modified zebrafish has also allowed identification of new putative therapeutic drugs. So far, most studies have relied on broad overexpression of transgenes harboring patient-derived mutations or loss-of-function mutants, which incompletely model the human disease allele in terms of expression levels or cell-type specificity of the endogenous gene of interest. Most human genetically inherited conditions are caused by alleles carrying single nucleotide changes resulting in altered gene function. Introduction of such point mutations in the zebrafish genome would be a prerequisite to recapitulate human disease but remains challenging to this day. We present an effective approach to introduce small nucleotide changes in the zebrafish genome. We generated four different knock-in lines carrying distinct human cardiovascular-disorder-causing missense mutations in their zebrafish orthologous genes by combining CRISPR/Cas9 with a short template oligonucleotide. Three of these lines carry gain-of-function mutations in genes encoding the pore-forming (Kir6.1, KCNJ8) and regulatory (SUR2, ABCC9) subunits of an ATP-sensitive potassium channel (KATP) linked to Cantú syndrome (CS). Our heterozygous zebrafish knock-in lines display significantly enlarged ventricles with enhanced cardiac output and contractile function, and distinct cerebral vasodilation, demonstrating the causality of the introduced mutations for CS. These results demonstrate that introducing patient alleles in their zebrafish orthologs promises a broad application for modeling human genetic diseases, paving the way for new therapeutic strategies using this model organism.


Subject(s)
CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Cardiovascular Diseases/genetics , Gene Editing , Nucleotides/genetics , Zebrafish/genetics , Animals , Base Sequence , Disease Models, Animal , Gene Knock-In Techniques , Genetic Testing , Heterozygote , Humans , Mutation/genetics
4.
Eur J Hum Genet ; 24(12): 1783-1791, 2016 12.
Article in English | MEDLINE | ID: mdl-27406248

ABSTRACT

Variants in the ZIC3 gene are rare, but have demonstrated their profound clinical significance in X-linked heterotaxy, affecting in particular male patients with abnormal arrangement of thoracic and visceral organs. Several reports have shown relevance of ZIC3 gene variants in both familial and sporadic cases and with a predominance of mutations detected in zinc-finger domains. No studies so far have assessed the functional consequences of ZIC3 variants in an in vivo model organism. A study population of 348 patients collected over more than 10 years with a large variety of congenital heart disease including heterotaxy was screened for variants in the ZIC3 gene. Functional effects of three variants were assessed both in vitro and in vivo in the zebrafish. We identified six novel pathogenic variants (1,7%), all in either male patients with heterotaxy (n=5) or a female patient with multiple male deaths due to heterotaxy in the family (n=1). All variants were located within the zinc-finger domains or leading to a truncation before these domains. Truncating variants showed abnormal trafficking of mutated ZIC3 proteins, whereas the missense variant showed normal trafficking. Overexpression of wild-type and mutated ZIC protein in zebrafish showed full non-functionality of the two frame-shift variants and partial activity of the missense variant compared with wild-type, further underscoring the pathogenic character of these variants. Concluding, we greatly expanded the number of causative variants in ZIC3 and delineated the functional effects of three variants using in vitro and in vivo model systems.


Subject(s)
Dextrocardia/genetics , Gene Deletion , Genetic Diseases, X-Linked/genetics , Heterotaxy Syndrome/genetics , Homeodomain Proteins/genetics , Mutation, Missense , Transcription Factors/genetics , Animals , Dextrocardia/diagnosis , Female , Fetus/pathology , Genetic Diseases, X-Linked/diagnosis , HeLa Cells , Heterotaxy Syndrome/diagnosis , Homeodomain Proteins/metabolism , Humans , Infant, Newborn , Male , Pregnancy , Protein Transport , Transcription Factors/metabolism , Zebrafish
5.
Hum Mutat ; 37(2): 194-200, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26531781

ABSTRACT

Establishing correct left-right asymmetry during embryonic development is crucial for proper asymmetric positioning of the organs. Congenital heart defects, such as dextrocardia, transposition of the arteries, and inflow or outflow tract malformations, comprise some of the most common birth defects and may be attributed to incorrect establishment of body laterality. Here, we identify new patients with dextrocardia who have mutations in CFAP53, a coiled-coil domain containing protein. To elucidate the mechanism by which CFAP53 regulates embryonic asymmetry, we used genome editing to generate cfap53 zebrafish mutants. Zebrafish cfap53 mutants have specific defects in organ laterality and randomization of asymmetric gene expression. We show that cfap53 is required for cilia rotation specifically in Kupffer's vesicle, the zebrafish laterality organ, providing a mechanism by which patients with CFAP53 mutations develop dextrocardia and heterotaxy, and confirming previous evidence that left-right asymmetry in humans is regulated through cilia-driven fluid flow in a laterality organ.


Subject(s)
Cytoskeletal Proteins/genetics , Dextrocardia/genetics , Heterotaxy Syndrome/genetics , Mutation , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Base Sequence , Body Patterning/genetics , Cilia/metabolism , Cilia/pathology , Conserved Sequence , Cytoskeletal Proteins/metabolism , DNA Mutational Analysis , Dextrocardia/metabolism , Dextrocardia/pathology , Embryo, Nonmammalian , Embryonic Development/genetics , Female , Gene Expression , Heterotaxy Syndrome/metabolism , Heterotaxy Syndrome/pathology , Humans , Lateral Line System/embryology , Lateral Line System/metabolism , Male , Molecular Sequence Data , Pedigree , Siblings , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/metabolism
6.
PLoS Genet ; 7(9): e1002289, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21980297

ABSTRACT

In vertebrates, left-right (LR) axis specification is determined by a ciliated structure in the posterior region of the embryo. Fluid flow in this ciliated structure is responsible for the induction of unilateral left-sided Nodal activity in the lateral plate mesoderm, which in turn regulates organ laterality. Bmp signalling activity has been implied in repressing Nodal expression on the right side, however its mechanism of action has been controversial. In a forward genetic screen for mutations that affect LR patterning, we identified the zebrafish linkspoot (lin) mutant, characterized by cardiac laterality and mild dorsoventral patterning defects. Mapping of the lin mutation revealed an inactivating missense mutation in the Bmp receptor 1aa (bmpr1aa) gene. Embryos with a mutation in lin/bmpr1aa and a novel mutation in its paralogue, bmpr1ab, displayed a variety of dorsoventral and LR patterning defects with increasing severity corresponding with a decrease in bmpr1a dosage. In Bmpr1a-deficient embryos we observed bilateral expression of the Nodal-related gene, spaw, coupled with reduced expression of the Nodal-antagonist lefty1 in the midline. Using genetic models to induce or repress Bmp activity in combination with Nodal inhibition or activation, we found that Bmp and Nodal regulate lefty1 expression in the midline independently of each other. Furthermore, we observed that the regulation of lefty1 by Bmp signalling is required for its observed downregulation of Nodal activity in the LPM providing a novel explanation for this phenomenon. From these results we propose a two-step model in which Bmp regulates LR patterning. Prior to the onset of nodal flow and Nodal activation, Bmp is required to induce lefty1 expression in the midline. When nodal flow has been established and Nodal activity is apparent, both Nodal and Bmp independently are required for lefty1 expression to assure unilateral Nodal activation and correct LR patterning.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Proteins/genetics , Embryonic Development/genetics , Left-Right Determination Factors/genetics , Nodal Protein/genetics , Zebrafish Proteins/metabolism , Zebrafish/growth & development , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Animals , Body Patterning/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Proteins/metabolism , Left-Right Determination Factors/metabolism , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutation, Missense , Nodal Protein/metabolism , Signal Transduction , Zebrafish/genetics , Zebrafish Proteins/genetics
7.
Eur J Hum Genet ; 19(4): 389-93, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21248739

ABSTRACT

Down's syndrome (DS), resulting from an additional copy of chromosome 21 (trisomy 21), is frequently associated with congenital heart defects (CHDs). Although the increased dosage of chromosome 21 sequences is likely to be part of the etiology of cardiac defects, only a proportion of DS patients exhibit a congenital heart defect (birth prevalence 40-60%). Through a large-candidate gene-sequencing screen in patients with atrioventricular septal defects, substitutions were identified in bone morphogenetic protein (BMP) type I receptor ALK2 and two other genes in a patient with DS and a primum-type atrial septal defect. Structural modeling of the cytoplasmic domain of the ALK2 receptor suggests that H286 is in close proximity to the nucleotide-binding site of the kinase domain. We investigated whether this p.His286Asp substitution altered ALK2 function by using both in vitro as well as in vivo assays. The p.His286Asp variant demonstrated impaired functional activity as measured by BMP-specific transcriptional response assays. Furthermore, mild dominant-interfering activity was observed in vivo compared with wild-type ALK2 as determined by RNA injection into zebrafish embryos. These data indicate that in the context of a DS background, ALK2-mediated reduction of BMP signaling may contribute to CHDs.


Subject(s)
Activin Receptors, Type I/genetics , Down Syndrome/genetics , Heart Defects, Congenital/etiology , Heart Defects, Congenital/genetics , Mutation/genetics , Activin Receptors, Type I/chemistry , Activin Receptors, Type I/metabolism , Animals , Bone Morphogenetic Protein 1/metabolism , Cattle , Down Syndrome/complications , Female , Heart Defects, Congenital/diagnosis , Heart Septal Defects, Atrial/genetics , Heart Septal Defects, Atrial/pathology , Humans , Male , Protein Conformation , Zebrafish/genetics
8.
Circulation ; 119(24): 3062-9, 2009 Jun 23.
Article in English | MEDLINE | ID: mdl-19506109

ABSTRACT

BACKGROUND: Serious congenital heart defects occur as a result of improper atrioventricular septum (AVS) development during embryogenesis. Despite extensive knowledge of the genetic control of AVS development, few genetic lesions have been identified that are responsible for AVS-associated congenital heart defects. METHODS AND RESULTS: We sequenced 32 genes known to be important in AVS development in patients with AVS defects and identified 11 novel coding single-nucleotide polymorphisms that are predicted to impair protein function. We focused on variants identified in the bone morphogenetic protein receptor, ALK2, and subjected 2 identified variants to functional analysis. The coding single-nucleotide polymorphisms R307L and L343P are heterozygous missense substitutions and were each identified in single individuals. The L343P allele had impaired functional activity as measured by in vitro kinase and bone morphogenetic protein-specific transcriptional response assays and dominant-interfering activity in vivo. In vivo analysis of zebrafish embryos injected with ALK2 L343P RNA revealed improper atrioventricular canal formation. CONCLUSIONS: These data identify the dominant-negative allele ALK2 L343P in a patient with AVS defects.


Subject(s)
Activin Receptors, Type I/metabolism , Alleles , Genes, Dominant , Heart Septal Defects/metabolism , Polymorphism, Single Nucleotide , Activin Receptors, Type I/genetics , Amino Acid Substitution , Animals , COS Cells , Cattle , Chlorocebus aethiops , Female , Heart/embryology , Heart/physiopathology , Heart Septal Defects/genetics , Heart Septal Defects/physiopathology , Humans , Male , Mutation, Missense , Myocardium/metabolism , Netherlands , Zebrafish/embryology , Zebrafish/genetics
9.
Dev Cell ; 14(2): 287-97, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18267096

ABSTRACT

We have used high-resolution 4D imaging of cardiac progenitor cells (CPCs) in zebrafish to investigate the earliest left-right asymmetric movements during cardiac morphogenesis. Differential migratory behavior within the heart field was observed, resulting in a rotation of the heart tube. The leftward displacement and rotation of the tube requires hyaluronan synthase 2 expression within the CPCs. Furthermore, by reducing or ectopically activating BMP signaling or by implantation of BMP beads we could demonstrate that BMP signaling, which is asymmetrically activated in the lateral plate mesoderm and regulated by early left-right signals, is required to direct CPC migration and cardiac rotation. Together, these results support a model in which CPCs migrate toward a BMP source during development of the linear heart tube, providing a mechanism by which the left-right axis drives asymmetric development of the vertebrate heart.


Subject(s)
Body Patterning , Cell Movement , Heart/embryology , Myocardium/cytology , Rotation , Stem Cells/cytology , Zebrafish/embryology , Animals , Body Patterning/drug effects , Bone Morphogenetic Proteins/pharmacology , Cell Lineage/drug effects , Cell Movement/drug effects , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/drug effects , Glucuronosyltransferase/metabolism , Heart/drug effects , Humans , Hyaluronan Synthases , Left-Right Determination Factors , Mesoderm/cytology , Mesoderm/drug effects , Mutation/genetics , Signal Transduction/drug effects , Smad Proteins/metabolism , Stem Cells/drug effects , Transforming Growth Factor beta/metabolism , Zebrafish Proteins/metabolism
10.
Dev Biol ; 305(2): 577-88, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17395172

ABSTRACT

Left-right (LR) asymmetry is regulated by early asymmetric signals within the embryo. Even though the role of the bone morphogenetic protein (BMP) pathway in this process has been reported extensively in various model organisms, opposing models for the mechanism by which BMP signaling operates still prevail. Here we show that in zebrafish embryos there are two distinct phases during LR patterning in which BMP signaling is required. Using transgenic lines that ectopically express either noggin3 or bmp2b, we show a requirement for BMP signaling during early segmentation to repress southpaw expression in the right lateral plate mesoderm and regulate both visceral and heart laterality. A second phase was identified during late segmentation, when BMP signaling is required in the left lateral plate mesoderm to regulate left-sided gene expression and heart laterality. Using morpholino knock down experiments, we identified Bmp4 as the ligand responsible for both phases of BMP signaling. In addition, we detected bmp4 expression in Kupffer's vesicle and show that restricted knock down of bmp4 in this structure results in LR patterning defects. The identification of these two distinct and opposing activities of BMP signaling provides new insight into how BMP signaling can regulate LR patterning.


Subject(s)
Body Patterning/physiology , Bone Morphogenetic Proteins/physiology , Zebrafish Proteins/physiology , Zebrafish/embryology , Activin Receptors, Type I/physiology , Animals , Animals, Genetically Modified , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/deficiency , Bone Morphogenetic Proteins/genetics , Functional Laterality/genetics , Heart/embryology , Signal Transduction/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...