Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
JCI Insight ; 8(17)2023 09 08.
Article in English | MEDLINE | ID: mdl-37561591

ABSTRACT

Pediatric cardiomyopathy (CM) represents a group of rare, severe disorders that affect the myocardium. To date, the etiology and mechanisms underlying pediatric CM are incompletely understood, hampering accurate diagnosis and individualized therapy development. Here, we identified biallelic variants in the highly conserved flightless-I (FLII) gene in 3 families with idiopathic, early-onset dilated CM. We demonstrated that patient-specific FLII variants, when brought into the zebrafish genome using CRISPR/Cas9 genome editing, resulted in the manifestation of key aspects of morphological and functional abnormalities of the heart, as observed in our patients. Importantly, using these genetic animal models, complemented with in-depth loss-of-function studies, we provided insights into the function of Flii during ventricular chamber morphogenesis in vivo, including myofibril organization and cardiomyocyte cell adhesion, as well as trabeculation. In addition, we identified Flii function to be important for the regulation of Notch and Hippo signaling, crucial pathways associated with cardiac morphogenesis and function. Taken together, our data provide experimental evidence for a role for FLII in the pathogenesis of pediatric CM and report biallelic variants as a genetic cause of pediatric CM.


Subject(s)
Cardiomyopathies , Microfilament Proteins , Animals , Cell Adhesion/genetics , Microfilament Proteins/genetics , Myocytes, Cardiac/metabolism , Myofibrils/metabolism , Zebrafish/genetics , Trans-Activators , Cardiomyopathies/genetics
2.
Cell Rep ; 41(8): 111705, 2022 11 22.
Article in English | MEDLINE | ID: mdl-36417881

ABSTRACT

Intrinsic and extrinsic inhibition of neuronal regeneration obstruct spinal cord (SC) repair in mammals. In contrast, adult zebrafish achieve functional recovery after complete SC transection. While studies of innate SC regeneration have focused on axon regrowth as a primary repair mechanism, how local adult neurogenesis affects functional recovery is unknown. Here, we uncover dynamic expression of zebrafish myostatin b (mstnb) in a niche of dorsal SC progenitors after injury. mstnb mutants show impaired functional recovery, normal glial and axonal bridging across the lesion, and an increase in the profiles of newborn neurons. Molecularly, neuron differentiation genes are upregulated, while the neural stem cell maintenance gene fgf1b is downregulated in mstnb mutants. Finally, we show that human fibroblast growth factor 1 (FGF1) treatment rescues the molecular and cellular phenotypes of mstnb mutants. These studies uncover unanticipated neurogenic functions for mstnb and establish the importance of local adult neurogenesis for innate SC repair.


Subject(s)
Spinal Cord Injuries , Zebrafish , Adult , Humans , Animals , Infant, Newborn , Myostatin , Neurogenesis , Spinal Cord Injuries/genetics , Recovery of Function , Fibroblast Growth Factor 1 , Mammals
3.
Sci Adv ; 8(35): eabn2082, 2022 Sep 02.
Article in English | MEDLINE | ID: mdl-36044573

ABSTRACT

Endothelial specification is a key event during embryogenesis; however, when, and how, endothelial cells separate from other lineages is poorly understood. In zebrafish, Npas4l is indispensable for endothelial specification by inducing the expression of the transcription factor genes etsrp, tal1, and lmo2. We generated a knock-in reporter in zebrafish npas4l to visualize endothelial progenitors and their derivatives in wild-type and mutant embryos. Unexpectedly, we find that in npas4l mutants, npas4l reporter-expressing cells contribute to the pronephron tubules. Single-cell transcriptomics and live imaging of the early lateral plate mesoderm in wild-type embryos indeed reveals coexpression of endothelial and pronephron markers, a finding confirmed by creERT2-based lineage tracing. Increased contribution of npas4l reporter-expressing cells to pronephron tubules is also observed in tal1 and lmo2 mutants and is reversed in npas4l mutants injected with tal1 mRNA. Together, these data reveal that Npas4l/Tal1/Lmo2 regulate the fate decision between the endothelial and pronephron lineages.

4.
Sci Adv ; 7(37): eabg6497, 2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34516874

ABSTRACT

Damage-induced fibrotic scarring limits tissue regeneration in mammals and is a leading cause of morbidity. In contrast, species like zebrafish can regenerate damaged tissues without excessive fibrosis. However, whether specific signaling pathways can both limit fibrosis and promote regeneration is unclear. Here, we show that interleukin-11 (Il-11)/Stat3 signaling has such a dual function. Zebrafish lacking Il-11 receptor function display severely compromised heart, fin, and scale regeneration. Deep phenotyping and transcriptional analysis of adult hearts and fins show that Il-11 signaling drives cellular reprogramming to orchestrate global and tissue-specific regenerative programs and broadly antagonizes hallmarks of adult mammalian scarring. Mechanistically, our data indicate that IL-11 signaling in endothelial cells antagonizes profibrotic transforming growth factor­ß signaling and endothelial-to-mesenchymal transition, limiting scarring and promoting cardiomyocyte repopulation, after injury. Overall, our findings position damage-induced Il-11/Stat3 signaling in a key role limiting fibrosis and promoting regeneration, revealing novel targets for regenerative therapies.

5.
Elife ; 102021 08 17.
Article in English | MEDLINE | ID: mdl-34403334

ABSTRACT

To investigate the role of the vasculature in pancreatic ß-cell regeneration, we crossed a zebrafish ß-cell ablation model into the avascular npas4l mutant (i.e. cloche). Surprisingly, ß-cell regeneration increased markedly in npas4l mutants owing to the ectopic differentiation of ß-cells in the mesenchyme, a phenotype not previously reported in any models. The ectopic ß-cells expressed endocrine markers of pancreatic ß-cells, and also responded to glucose with increased calcium influx. Through lineage tracing, we determined that the vast majority of these ectopic ß-cells has a mesodermal origin. Notably, ectopic ß-cells were found in npas4l mutants as well as following knockdown of the endothelial/myeloid determinant Etsrp. Together, these data indicate that under the perturbation of endothelial/myeloid specification, mesodermal cells possess a remarkable plasticity enabling them to form ß-cells, which are normally endodermal in origin. Understanding the restriction of this differentiation plasticity will help exploit an alternative source for ß-cell regeneration.


Subject(s)
Cell Differentiation , Insulin-Secreting Cells/physiology , Mesoderm/embryology , Regeneration , Zebrafish/embryology , Animals , Endothelium/physiology , Insulins/metabolism , Zebrafish/physiology
6.
Development ; 146(11)2019 06 03.
Article in English | MEDLINE | ID: mdl-31097478

ABSTRACT

The development of a vascular network is essential to nourish tissues and sustain organ function throughout life. Endothelial cells (ECs) are the building blocks of blood vessels, yet our understanding of EC specification remains incomplete. Zebrafish cloche/npas4l mutants have been used broadly as an avascular model, but little is known about the molecular mechanisms of action of the Npas4l transcription factor. Here, to identify its direct and indirect target genes, we have combined complementary genome-wide approaches, including transcriptome analyses and chromatin immunoprecipitation. The cross-analysis of these datasets indicates that Npas4l functions as a master regulator by directly inducing a group of transcription factor genes that are crucial for hematoendothelial specification, such as etv2, tal1 and lmo2 We also identified new targets of Npas4l and investigated the function of a subset of them using the CRISPR/Cas9 technology. Phenotypic characterization of tspan18b mutants reveals a novel player in developmental angiogenesis, confirming the reliability of the datasets generated. Collectively, these data represent a useful resource for future studies aimed to better understand EC fate determination and vascular development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Endothelium, Vascular/embryology , Gene Expression Regulation, Developmental , Neovascularization, Physiologic/genetics , Zebrafish Proteins/physiology , Zebrafish , Animals , Animals, Genetically Modified , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites/genetics , Cell Differentiation/genetics , Chromosome Mapping/methods , Datasets as Topic , Embryo, Nonmammalian , Endothelial Cells/physiology , Endothelium, Vascular/metabolism , Genomics/methods , LIM Domain Proteins/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1/genetics , Transcription Factors/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
7.
PLoS One ; 13(9): e0204312, 2018.
Article in English | MEDLINE | ID: mdl-30252882

ABSTRACT

Muscle proteins with ankyrin repeats (MARPs) ANKRD1 and ANKRD2 are titin-associated proteins with a putative role as transcriptional co-regulators in striated muscle, involved in the cellular response to mechanical, oxidative and metabolic stress. Since many aspects of the biology of MARPs, particularly exact mechanisms of their action, in striated muscle are still elusive, research in this field will benefit from novel animal model system. Here we investigated the MARPs found in zebrafish for protein structure, evolutionary conservation, spatiotemporal expression profiles and response to increased muscle activity. Ankrd1 and Ankrd2 show overall moderate conservation at the protein level, more pronounced in the region of ankyrin repeats, motifs indispensable for their function. The two zebrafish genes, ankrd1a and ankrd1b, counterparts of mammalian ANKRD1/Ankrd1, have different expression profiles during first seven days of development. Mild increase of ankrd1a transcript levels was detected at 72 hpf (1.74±0.24 fold increase relative to 24 hpf time point), while ankrd1b expression was markedly upregulated from 24 hpf onward and peaked at 72 hpf (92.18±36.95 fold increase relative to 24 hpf time point). Spatially, they exhibited non-overlapping expression patterns during skeletal muscle development in trunk (ankrd1a) and tail (ankrd1b) somites. Expression of ankrd2 was barely detectable. Zebrafish MARPs, expressed at a relatively low level in adult striated muscle, were found to be responsive to endurance exercise training consisting of two bouts of 3 hours of forced swimming daily, for five consecutive days. Three hours after the last exercise bout, ankrd1a expression increased in cardiac muscle (6.19±5.05 fold change), while ankrd1b and ankrd2 were upregulated in skeletal muscle (1.97±1.05 and 1.84±0.58 fold change, respectively). This study provides the foundation to establish zebrafish as a novel in vivo model for further investigation of MARPs function in striated muscle.


Subject(s)
Ankyrin Repeat , Muscle Proteins/chemistry , Muscle Proteins/metabolism , Physical Conditioning, Animal , Zebrafish/physiology , Amino Acid Sequence , Animals , Gene Expression Regulation , Humans , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Myocardium/metabolism , Phylogeny , Sequence Alignment , Stress, Physiological , Synteny , Zebrafish/genetics , Zebrafish/metabolism
8.
Development ; 145(14)2018 07 30.
Article in English | MEDLINE | ID: mdl-30061167

ABSTRACT

Cardiomyocyte proliferation is crucial for cardiac growth, patterning and regeneration; however, few studies have investigated the behavior of dividing cardiomyocytes in vivo Here, we use time-lapse imaging of beating hearts in combination with the FUCCI system to monitor the behavior of proliferating cardiomyocytes in developing zebrafish. Confirming in vitro observations, sarcomere disassembly, as well as changes in cell shape and volume, precede cardiomyocyte cytokinesis. Notably, cardiomyocytes in zebrafish embryos and young larvae mostly divide parallel to the myocardial wall in both the compact and trabecular layers, and cardiomyocyte proliferation is more frequent in the trabecular layer. While analyzing known regulators of cardiomyocyte proliferation, we observed that the Nrg/ErbB2 and TGFß signaling pathways differentially affect compact and trabecular layer cardiomyocytes, indicating that distinct mechanisms drive proliferation in these two layers. In summary, our data indicate that, in zebrafish, cardiomyocyte proliferation is essential for trabecular growth, but not initiation, and set the stage to further investigate the cellular and molecular mechanisms driving cardiomyocyte proliferation in vivo.


Subject(s)
Myocytes, Cardiac/cytology , Organogenesis , Zebrafish/growth & development , Animals , Cell Division , Cell Proliferation , Cell Shape , Cell Size , Gene Expression Regulation, Developmental , Heart/growth & development , Ligands , Myocytes, Cardiac/metabolism , Sarcomeres/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Zebrafish/genetics , Zebrafish Proteins/metabolism
9.
Elife ; 72018 05 15.
Article in English | MEDLINE | ID: mdl-29762122

ABSTRACT

The ultimate formation of a four-chambered heart allowing the separation of the pulmonary and systemic circuits was key for the evolutionary success of tetrapods. Complex processes of cell diversification and tissue morphogenesis allow the left and right cardiac compartments to become distinct but remain poorly understood. Here, we describe an unexpected laterality in the single zebrafish atrium analogous to that of the two atria in amniotes, including mammals. This laterality appears to derive from an embryonic antero-posterior asymmetry revealed by the expression of the transcription factor gene meis2b. In adult zebrafish hearts, meis2b expression is restricted to the left side of the atrium where it controls the expression of pitx2c, a regulator of left atrial identity in mammals. Altogether, our studies suggest that the multi-chambered atrium in amniotes arose from a molecular blueprint present before the evolutionary emergence of cardiac septation and provide insights into the establishment of atrial asymmetry.


Subject(s)
Heart/embryology , Organogenesis , Zebrafish/embryology , Animals , Body Patterning , Gene Expression Regulation, Developmental , Homeodomain Proteins/biosynthesis , Zebrafish Proteins/biosynthesis
11.
Nat Commun ; 8(1): 1902, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29196619

ABSTRACT

Zebrafish regenerate damaged myocardial tissue very effectively. Hence, insights into the molecular networks underlying zebrafish heart regeneration might help develop alternative strategies to restore human cardiac performance. While TGF-ß signaling has been implicated in zebrafish cardiac regeneration, the role of its individual ligands remains unclear. Here, we report the opposing expression response during zebrafish heart regeneration of two genes, mstnb and inhbaa, which encode TGF-ß family ligands. Using gain-of-function (GOF) and loss-of-function (LOF) approaches, we show that these ligands mediate inverse effects on cardiac regeneration and specifically on cardiomyocyte (CM) proliferation. Notably, we find that Inhbaa functions as a CM mitogen and that its overexpression leads to accelerated cardiac recovery and scar clearance after injury. In contrast, mstnb GOF and inhbaa LOF both lead to unresolved scarring after cardiac injury. We further show that Mstnb and Inhbaa inversely control Smad2 and Smad3 transcription factor activities through alternate Activin type 2 receptors.


Subject(s)
Activin Receptors, Type II/metabolism , Cell Proliferation , Inhibin-beta Subunits/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myostatin/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Activin Receptors, Type II/genetics , Animals , Female , Heart/growth & development , Heart/physiology , Inhibin-beta Subunits/genetics , Ligands , Male , Myostatin/genetics , Regeneration , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish Proteins/genetics
12.
Nat Commun ; 8(1): 1525, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29142194

ABSTRACT

Cells can sacrifice their individuality by fusing, but the prevalence and significance of this process are poorly understood. To approach these questions, here we generate transgenic reporter lines in zebrafish to label and specifically ablate fused cells. In addition to skeletal muscle cells, the reporters label cardiomyocytes starting at an early developmental stage. Genetic mosaics generated by cell transplantation show cardiomyocytes expressing both donor- and host-derived transgenes, confirming the occurrence of fusion in larval hearts. These fusion events are transient and do not generate multinucleated cardiomyocytes. Functionally, cardiomyocyte fusion correlates with their mitotic activity during development as well as during regeneration in adult animals. By analyzing the cell fusion-compromised jam3b mutants, we propose a role for membrane fusion in cardiomyocyte proliferation and cardiac function. Together, our findings uncover the previously unrecognized process of transient cardiomyocyte fusion and identify its potential role in cardiac development and function.


Subject(s)
Cell Fusion , Gene Expression Regulation, Developmental , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified , Cell Differentiation/genetics , Cell Proliferation/genetics , Heart/embryology , Heart/growth & development , Larva/genetics , Larva/growth & development , Larva/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Myocardium/cytology , Myocytes, Cardiac/cytology , Zebrafish , Zebrafish Proteins/metabolism
13.
Physiol Rev ; 97(3): 889-938, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28468832

ABSTRACT

The burden of cardiovascular and metabolic diseases worldwide is staggering. The emergence of systems approaches in biology promises new therapies, faster and cheaper diagnostics, and personalized medicine. However, a profound understanding of pathogenic mechanisms at the cellular and molecular levels remains a fundamental requirement for discovery and therapeutics. Animal models of human disease are cornerstones of drug discovery as they allow identification of novel pharmacological targets by linking gene function with pathogenesis. The zebrafish model has been used for decades to study development and pathophysiology. More than ever, the specific strengths of the zebrafish model make it a prime partner in an age of discovery transformed by big-data approaches to genomics and disease. Zebrafish share a largely conserved physiology and anatomy with mammals. They allow a wide range of genetic manipulations, including the latest genome engineering approaches. They can be bred and studied with remarkable speed, enabling a range of large-scale phenotypic screens. Finally, zebrafish demonstrate an impressive regenerative capacity scientists hope to unlock in humans. Here, we provide a comprehensive guide on applications of zebrafish to investigate cardiovascular and metabolic diseases. We delineate advantages and limitations of zebrafish models of human disease and summarize their most significant contributions to understanding disease progression to date.


Subject(s)
Cardiovascular Diseases/genetics , Drug Discovery/methods , Metabolic Diseases/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified , Cardiovascular Agents/pharmacology , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Metabolic Diseases/drug therapy , Metabolic Diseases/metabolism , Metabolic Diseases/physiopathology , Phenotype , Species Specificity , Zebrafish/metabolism
14.
Nature ; 535(7611): 294-8, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27411634

ABSTRACT

Vascular and haematopoietic cells organize into specialized tissues during early embryogenesis to supply essential nutrients to all organs and thus play critical roles in development and disease. At the top of the haemato-vascular specification cascade lies cloche, a gene that when mutated in zebrafish leads to the striking phenotype of loss of most endothelial and haematopoietic cells and a significant increase in cardiomyocyte numbers. Although this mutant has been analysed extensively to investigate mesoderm diversification and differentiation and continues to be broadly used as a unique avascular model, the isolation of the cloche gene has been challenging due to its telomeric location. Here we used a deletion allele of cloche to identify several new cloche candidate genes within this genomic region, and systematically genome-edited each candidate. Through this comprehensive interrogation, we succeeded in isolating the cloche gene and discovered that it encodes a PAS-domain-containing bHLH transcription factor, and that it is expressed in a highly specific spatiotemporal pattern starting during late gastrulation. Gain-of-function experiments show that it can potently induce endothelial gene expression. Epistasis experiments reveal that it functions upstream of etv2 and tal1, the earliest expressed endothelial and haematopoietic transcription factor genes identified to date. A mammalian cloche orthologue can also rescue blood vessel formation in zebrafish cloche mutants, indicating a highly conserved role in vertebrate vasculogenesis and haematopoiesis. The identification of this master regulator of endothelial and haematopoietic fate enhances our understanding of early mesoderm diversification and may lead to improved protocols for the generation of endothelial and haematopoietic cells in vivo and in vitro.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Blood Cells/cytology , Blood Cells/metabolism , Cell Differentiation/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Zebrafish Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Blood Vessels/cytology , Blood Vessels/embryology , Blood Vessels/metabolism , Conserved Sequence , Epistasis, Genetic , Gene Deletion , Helix-Loop-Helix Motifs , Hematopoiesis , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/metabolism , Mutation , Protein Structure, Tertiary , Proto-Oncogene Proteins/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1 , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
15.
Nat Commun ; 7: 11303, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27066836

ABSTRACT

Atrial fibrillation (AF), the most common arrhythmia, is a growing epidemic with substantial morbidity and economic burden. Mechanisms underlying vulnerability to AF remain poorly understood, which contributes to the current lack of highly effective therapies. Recognizing mechanistic subtypes of AF may guide an individualized approach to patient management. Here, we describe a family with a previously unreported syndrome characterized by early-onset AF (age <35 years), conduction disease and signs of a primary atrial myopathy. Phenotypic penetrance was complete in all mutation carriers, although complete disease expressivity appears to be age-dependent. We show that this syndrome is caused by a novel, heterozygous p.Glu11Lys mutation in the atrial-specific myosin light chain gene MYL4. In zebrafish, mutant MYL4 leads to disruption of sarcomeric structure, atrial enlargement and electrical abnormalities associated with human AF. These findings describe the cause of a rare subtype of AF due to a primary, atrial-specific sarcomeric defect.


Subject(s)
Atrial Fibrillation/genetics , Heart Atria/pathology , Mutation/genetics , Myosin Light Chains/genetics , Adult , Animals , Animals, Genetically Modified , Atrial Fibrillation/diagnostic imaging , Binding Sites , Electrocardiography , Female , Humans , Male , Middle Aged , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/genetics , Myofibrils/pathology , Myosin Light Chains/chemistry , Pedigree , Protein Binding , Protein Structure, Secondary , Sarcomeres/pathology , Ultrasonography , Zebrafish
16.
Dev Biol ; 412(1): 71-82, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26892463

ABSTRACT

The atrioventricular canal (AVC) connects the atrial and ventricular chambers of the heart and its formation is critical for the development of the cardiac valves, chamber septation and formation of the cardiac conduction system. Consequently, problems in AVC formation can lead to congenital defects ranging from cardiac arrhythmia to incomplete cardiac septation. While our knowledge about early heart tube formation is relatively comprehensive, much remains to be investigated about the genes that regulate AVC formation. Here we identify a new role for the basic helix-loop-helix factor Id4 in zebrafish AVC valve development and function. id4 is first expressed in the AVC endocardium and later becomes more highly expressed in the atrial chamber. TALEN induced inactivation of id4 causes retrograde blood flow at the AV canal under heat induced stress conditions, indicating defects in AV valve function. At the molecular level, we found that id4 inactivation causes misexpression of several genes important for AVC and AV valve formation including bmp4 and spp1. We further show that id4 appears to control the number of endocardial cells that contribute to the AV valves by regulating Wnt signaling in the developing AVC endocardium.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Endocardium/embryology , Inhibitor of Differentiation Proteins/physiology , Signal Transduction , Zebrafish/embryology , Animals
17.
J Vis Exp ; (94)2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25548868

ABSTRACT

Use of the zebrafish model system for studying development, regeneration, and disease is expanding toward use of adult hearts for cell dissociation and purification of RNA, DNA, and proteins. All of these applications demand the rapid recovery of significant numbers of zebrafish hearts to avoid gene regulatory, metabolic, and other changes that begin after death. Adult zebrafish hearts are also required for studying heart structure for a variety of mutants and for studying heart regeneration. However, the traditional zebrafish heart dissection is slow and difficult and requires specialized tools, making large-scale dissection of adult zebrafish hearts tedious. Traditional methods also harbor the risk of damaging the heart during the dissection. Here, we describe a method for dissection of adult zebrafish hearts that is fast, reproducible, and preserves heart architecture. Furthermore, this method does not require specialized tools, is painless for the zebrafish, can be performed on fresh or fixed specimens, and can be performed on zebrafish as young as one month old. The approach described expands the use of adult zebrafish for cardiovascular research.


Subject(s)
Cardiac Surgical Procedures/veterinary , Dissection/veterinary , Heart/anatomy & histology , High-Throughput Screening Assays/methods , Zebrafish/surgery , Animals , Cardiac Surgical Procedures/methods , Dissection/methods
18.
Circ Res ; 115(10): 845-56, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25228389

ABSTRACT

RATIONALE: Dilated cardiomyopathy is a leading cause of congestive heart failure and a debilitating complication of antineoplastic therapies. Despite disparate causes for dilated cardiomyopathy, maladaptive cardiac remodeling and decreased systolic function are common clinical consequences, begging an investigation of in vivo contractile dynamics in development and disease, one that has been impossible to date. OBJECTIVE: To image myocardial contractile filament dynamics in vivo and to assess potential causes of dilated cardiomyopathy in antineoplastic therapies targeting the epidermal growth factor receptor Erbb2. METHODS AND RESULTS: We generated a transgenic zebrafish line expressing an actin-binding green fluorescent protein in cardiomyocytes, allowing an in vivo imaging of myofilaments. Analysis of this line revealed architectural differences in myofibrils of the distinct cardiomyocyte subtypes. We used this model to investigate the effects of Erbb2 signaling on myofibrillar organization because drugs targeting ERBB2 (HER2/NEU) signaling, a mainstay of breast cancer chemotherapy, cause dilated cardiomyopathy in many patients. High-resolution in vivo imaging revealed that Erbb2 signaling regulates a switch between a dense apical network of filamentous myofibrils and the assembly of basally localized myofibrils in ventricular cardiomyocytes. CONCLUSIONS: Using this novel line, we compiled a reference for myofibrillar microarchitecture among myocardial subtypes in vivo and at different developmental stages, establishing this model as a tool to analyze in vivo cardiomyocyte contractility and remodeling for a broad range of cardiovascular questions. Furthermore, we applied this model to study Erbb2 signaling in cardiomyopathy. We show a direct link between Erbb2 activity and remodeling of myofibrils, revealing an unexpected mechanism with potentially important implications for prevention and treatment of cardiomyopathy.


Subject(s)
Actins/metabolism , Green Fluorescent Proteins/metabolism , Heart/physiology , Imaging, Three-Dimensional/methods , Myofibrils/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction/physiology , Amino Acid Sequence , Animals , Animals, Genetically Modified , Cells, Cultured , Heart/anatomy & histology , Molecular Sequence Data , Myofibrils/chemistry , Protein Binding/physiology , Receptor, ErbB-2/analysis , Receptor, ErbB-2/genetics , Time Factors , Zebrafish
19.
Development ; 141(20): 3988-93, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25231762

ABSTRACT

Heterogeneity within a population of cells of the same type is a common theme in metazoan biology. Dissecting complex developmental and physiological processes crucially relies on our ability to probe the expression profile of these cell subpopulations. Current strategies rely on cell enrichment based on sequential or simultaneous use of multiple intersecting markers starting from a heterogeneous cell suspension. The extensive tissue manipulations required to generate single-cell suspensions, as well as the complexity of the required equipment, inherently complicate these approaches. Here, we propose an alternative methodology based on a genetically encoded system in the model organism Danio rerio (zebrafish). In transgenic fish, we take advantage of the combinatorial biotin transfer system, where polysome-associated mRNAs are selectively recovered from cells expressing both a tagged ribosomal subunit, Rpl10a, and the bacterial biotin ligase BirA. We have applied this technique to skeletal muscle development and identified new genes with interesting temporal expression patterns. Through this work we have thus developed additional tools for highly specific gene expression profiling.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Developmental , RNA-Binding Proteins/physiology , Transcription, Genetic , Zebrafish Proteins/physiology , Animals , Animals, Genetically Modified , Biotinylation , Coenzyme A Ligases/chemistry , Green Fluorescent Proteins/chemistry , In Situ Hybridization , Mass Spectrometry , Muscle, Skeletal/pathology , Polyribosomes/chemistry , RNA, Messenger/metabolism , Ribosomal Proteins/physiology , Ribosomes/metabolism , Zebrafish
20.
Nat Methods ; 11(9): 919-22, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25042787

ABSTRACT

The heart's continuous motion makes it difficult to capture high-resolution images of this organ in vivo. We developed tools based on high-speed selective plane illumination microscopy (SPIM), offering pristine views into the beating zebrafish heart. We captured three-dimensional cardiac dynamics with postacquisition synchronization of multiview movie stacks, obtained static high-resolution reconstructions by briefly stopping the heart with optogenetics and resolved nonperiodic phenomena by high-speed volume scanning with a liquid lens.


Subject(s)
Cell Tracking/methods , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Microscopy, Video/methods , Myocytes, Cardiac/cytology , Zebrafish/anatomy & histology , Algorithms , Animals , Myocytes, Cardiac/physiology , Reproducibility of Results , Sensitivity and Specificity , Subtraction Technique , Zebrafish/physiology
SELECTION OF CITATIONS
SEARCH DETAIL