Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
J Gen Physiol ; 155(3)2023 03 06.
Article En | MEDLINE | ID: mdl-36633587

Cardiac muscle contraction is regulated by Ca2+-induced structural changes of the thin filaments to permit myosin cross-bridge cycling driven by ATP hydrolysis in the sarcomere. In congestive heart failure, contraction is weakened, and thus targeting the contractile proteins of the sarcomere is a promising approach to therapy. However, development of novel therapeutic interventions has been challenging due to a lack of precise discovery tools. We have developed a fluorescence lifetime-based assay using an existing site-directed probe, N,N'-dimethyl-N-(iodoacetyl)-N'-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)ethylenediamine (IANBD) attached to human cardiac troponin C (cTnC) mutant cTnCT53C, exchanged into porcine cardiac myofibrils. We hypothesized that IANBD-cTnCT53C fluorescence lifetime measurements provide insight into the activation state of the thin filament. The sensitivity and precision of detecting structural changes in cTnC due to physiological and therapeutic modulators of thick and thin filament functions were determined. The effects of Ca2+ binding to cTnC and myosin binding to the thin filament were readily detected by this assay in mock high-throughput screen tests using a fluorescence lifetime plate reader. We then evaluated known effectors of altered cTnC-Ca2+ binding, W7 and pimobendan, and myosin-binding drugs, mavacamten and omecamtiv mecarbil, used to treat cardiac diseases. Screening assays were determined to be of high quality as indicated by the Z' factor. We conclude that cTnC lifetime-based probes allow for precise evaluation of the thin filament activation in functioning myofibrils that can be used in future high-throughput screens of small-molecule modulators of function of the thin and thick filaments.


Calcium , Troponin C , Humans , Animals , Swine , Calcium/metabolism , Fluorescence , Troponin C/metabolism , Myocardium/metabolism , Myocardial Contraction/physiology
2.
Int J Mol Sci ; 22(11)2021 May 23.
Article En | MEDLINE | ID: mdl-34071043

A de novo missense variant in Rag GTPase protein C (RagCS75Y) was recently identified in a syndromic dilated cardiomyopathy (DCM) patient. However, its pathogenicity and the related therapeutic strategy remain unclear. We generated a zebrafish RragcS56Y (corresponding to human RagCS75Y) knock-in (KI) line via TALEN technology. The KI fish manifested cardiomyopathy-like phenotypes and poor survival. Overexpression of RagCS75Y via adenovirus infection also led to increased cell size and fetal gene reprogramming in neonatal rat ventricle cardiomyocytes (NRVCMs), indicating a conserved mechanism. Further characterization identified aberrant mammalian target of rapamycin complex 1 (mTORC1) and transcription factor EB (TFEB) signaling, as well as metabolic abnormalities including dysregulated autophagy. However, mTOR inhibition failed to ameliorate cardiac phenotypes in the RagCS75Y cardiomyopathy models, concomitant with a failure to promote TFEB nuclear translocation. This observation was at least partially explained by increased and mTOR-independent physical interaction between RagCS75Y and TFEB in the cytosol. Importantly, TFEB overexpression resulted in more nuclear TFEB and rescued cardiomyopathy phenotypes. These findings suggest that S75Y is a pathogenic gain-of-function mutation in RagC that leads to cardiomyopathy. A primary pathological step of RagCS75Y cardiomyopathy is defective mTOR-TFEB signaling, which can be corrected by TFEB overexpression, but not mTOR inhibition.


Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Cardiomyopathy, Dilated/genetics , Gain of Function Mutation , Monomeric GTP-Binding Proteins/genetics , Mutation, Missense , Point Mutation , TOR Serine-Threonine Kinases/antagonists & inhibitors , Active Transport, Cell Nucleus , Amino Acid Substitution , Animals , Autophagy , Base Sequence , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cardiomyopathy, Dilated/therapy , Cells, Cultured , Gene Knock-In Techniques , Gene Knockout Techniques , Heart Ventricles/cytology , Humans , Mice , Monomeric GTP-Binding Proteins/physiology , Myocytes, Cardiac/metabolism , Phenotype , Rats, Wistar , Recombinant Proteins/metabolism , Signal Transduction , Transcription Activator-Like Effector Nucleases , Zebrafish , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology
3.
Sci Adv ; 6(5): eaay2939, 2020 01.
Article En | MEDLINE | ID: mdl-32064346

To uncover the genetic basis of anthracycline-induced cardiotoxicity (AIC), we recently established a genetic suppressor screening strategy in zebrafish. Here, we report the molecular and cellular nature of GBT0419, a salutary modifier mutant that affects retinoid x receptor alpha a (rxraa). We showed that endothelial, but not myocardial or epicardial, RXRA activation confers AIC protection. We then identified isotretinoin and bexarotene, two FDA-approved RXRA agonists, which exert cardioprotective effects. The therapeutic effects of these drugs only occur when administered during early, but not late, phase of AIC or as pretreatment. Mechanistically, these spatially- and temporally-predominant benefits of RXRA activation can be ascribed to repair of damaged endothelial cell-barrier via regulating tight-junction protein Zonula occludens-1. Together, our study provides the first in vivo genetic evidence supporting RXRA as the therapeutic target for AIC, and uncovers a previously unrecognized spatiotemporally-predominant mechanism that shall inform future translational efforts.


Cardiotoxicity/drug therapy , Heart/drug effects , Retinoid X Receptor alpha/genetics , Zonula Occludens-1 Protein/genetics , Animals , Bexarotene/pharmacology , Cardiotonic Agents/pharmacology , Cardiotoxicity/etiology , Cardiotoxicity/genetics , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Heart/physiopathology , Humans , Isotretinoin/pharmacology , Myocardium/metabolism , Myocardium/pathology , Neoplasms/complications , Neoplasms/drug therapy , Pericardium/drug effects , Retinoid X Receptor alpha/agonists , Zebrafish
4.
Dis Model Mech ; 12(10)2019 10 01.
Article En | MEDLINE | ID: mdl-31492659

The adult zebrafish is an emerging vertebrate model for studying human cardiomyopathies; however, whether the simple zebrafish heart can model different subtypes of cardiomyopathies, such as dilated cardiomyopathy (DCM), remains elusive. Here, we generated and characterized an inherited DCM model in adult zebrafish and used this model to search for therapeutic strategies. We employed transcription activator-like effector nuclease (TALEN) genome editing technology to generate frame-shift mutants for the zebrafish ortholog of human BCL2-associated athanogene 3 (BAG3), an established DCM-causative gene. As in mammals, the zebrafish bag3 homozygous mutant (bag3e2/e2 ) exhibited aberrant proteostasis, as indicated by impaired autophagy flux and elevated ubiquitinated protein aggregation. Through comprehensive phenotyping analysis of the mutant, we identified phenotypic traits that resembled DCM phenotypes in mammals, including cardiac chamber enlargement, reduced ejection fraction characterized by increased end-systolic volume/body weight (ESV/BW), and reduced contractile myofibril activation kinetics. Nonbiased transcriptome analysis identified the hyperactivation of the mechanistic target of rapamycin (mTOR) signaling in bag3e2/e2 mutant hearts. Further genetic studies showed that mtorxu015/+ , an mTOR haploinsufficiency mutant, repaired abnormal proteostasis, improved cardiac function and rescued the survival of the bag3e2/e2 mutant. This study established the bag3e2/e2 mutant as a DCM model in adult zebrafish and suggested mtor as a candidate therapeutic target gene for BAG3 cardiomyopathy.


Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Cardiomyopathies/genetics , Haploinsufficiency/genetics , TOR Serine-Threonine Kinases/metabolism , Zebrafish Proteins/metabolism , Zebrafish/genetics , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/genetics , Base Sequence , Cardiomyopathies/pathology , Gene Expression Profiling , Mutation/genetics , Myocardium/metabolism , Myocardium/pathology , Phenotype , Signal Transduction , Transcription Activator-Like Effector Nucleases , Zebrafish Proteins/genetics
5.
J Mol Cell Cardiol ; 133: 199-208, 2019 08.
Article En | MEDLINE | ID: mdl-31228518

Adult zebrafish is an emerging vertebrate model for studying genetic basis of cardiomyopathies; but whether the simple fish heart can model essential features of hypertrophic cardiomyopathy (HCM) remained unknown. Here, we report a comprehensive phenotyping of a lamp2 knockout (KO) mutant. LAMP2 encodes a lysosomal protein and is a causative gene of Danon disease that is characterized by HCM and massive autophagic vacuoles accumulation in the tissues. There is no effective therapy yet to treat this most lethal cardiomyopathy in the young. First, we did find the autophagic vacuoles accumulation in cardiac tissues from lamp2 KO. Next, through employing a set of emerging phenotyping tools, we revealed heart failure phenotypes in the lamp2 KO mutants, including decreased ventricular ejection fraction, reduced physical exercise capacity, blunted ß-adrenergic contractile response, and enlarged atrium. We also noted changes of the following indices suggesting cardiac hypertrophic remodeling in lamp2 KO: a rounded heart shape, increased end-systolic ventricular volume and density of ventricular myocardium, elevated actomyosin activation kinetics together with increased maximal isometric tension at the level of cardiac myofibrils. Lastly, we assessed the function of lysosomal-localized mTOR on the lamp2-associated Danon disease. We found that haploinsufficiency of mtor was able to normalize some characteristics of the lamp2 KO, including ejection fraction, ß-adrenergic response, and the actomyosin activation kinetics. In summary, we demonstrate the feasibility of modeling the inherited HCM in the adult zebrafish, which can be used to develop potential therapies.


Glycogen Storage Disease Type IIb/metabolism , Lysosomal-Associated Membrane Protein 2/genetics , Phenotype , TOR Serine-Threonine Kinases/antagonists & inhibitors , Zebrafish/genetics , Animals , Cardiomegaly/genetics , Disease Models, Animal , Gene Knockout Techniques , Glycogen Storage Disease Type IIb/genetics , Lysosomal-Associated Membrane Protein 2/metabolism , Myocardial Contraction/genetics , Myocardium/metabolism , Myofibrils/metabolism , Receptors, Adrenergic, beta/metabolism , Stroke Volume , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Ventricular Remodeling/genetics , Zebrafish/metabolism
6.
PLoS Genet ; 14(9): e1007652, 2018 09.
Article En | MEDLINE | ID: mdl-30208061

One key problem in precision genome editing is the unpredictable plurality of sequence outcomes at the site of targeted DNA double stranded breaks (DSBs). This is due to the typical activation of the versatile Non-homologous End Joining (NHEJ) pathway. Such unpredictability limits the utility of somatic gene editing for applications including gene therapy and functional genomics. For germline editing work, the accurate reproduction of the identical alleles using NHEJ is a labor intensive process. In this study, we propose Microhomology-mediated End Joining (MMEJ) as a viable solution for improving somatic sequence homogeneity in vivo, capable of generating a single predictable allele at high rates (56% ~ 86% of the entire mutant allele pool). Using a combined dataset from zebrafish (Danio rerio) in vivo and human HeLa cell in vitro, we identified specific contextual sequence determinants surrounding genomic DSBs for robust MMEJ pathway activation. We then applied our observation to prospectively design MMEJ-inducing sgRNAs against a variety of proof-of-principle genes and demonstrated high levels of mutant allele homogeneity. MMEJ-based DNA repair at these target loci successfully generated F0 mutant zebrafish embryos and larvae that faithfully recapitulated previously reported, recessive, loss-of-function phenotypes. We also tested the generalizability of our approach in cultured human cells. Finally, we provide a novel algorithm, MENTHU (http://genesculpt.org/menthu/), for improved and facile prediction of candidate MMEJ loci. We believe that this MMEJ-centric approach will have a broader impact on genome engineering and its applications. For example, whereas somatic mosaicism hinders efficient recreation of knockout mutant allele at base pair resolution via the standard NHEJ-based approach, we demonstrate that F0 founders transmitted the identical MMEJ allele of interest at high rates. Most importantly, the ability to directly dictate the reading frame of an endogenous target will have important implications for gene therapy applications in human genetic diseases.


DNA Breaks, Double-Stranded , DNA End-Joining Repair/genetics , Gene Editing/methods , Models, Genetic , Algorithms , Alleles , Animals , Feasibility Studies , Female , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/therapy , Genetic Therapy/methods , HeLa Cells , Humans , Male , Mutagenesis, Site-Directed , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , Zebrafish
7.
Dis Model Mech ; 11(9)2018 09 10.
Article En | MEDLINE | ID: mdl-30012855

Zebrafish are increasingly used as a vertebrate model to study human cardiovascular disorders. Although heart structure and function are readily visualized in zebrafish embryos because of their optical transparency, the lack of effective tools for evaluating the hearts of older, nontransparent fish has been a major limiting factor. The recent development of high-frequency echocardiography has been an important advance for in vivo cardiac assessment, but it necessitates anesthesia and has limited ability to study acute interventions. We report the development of an alternative experimental ex vivo technique for quantifying heart size and function that resembles the Langendorff heart preparations that have been widely used in mammalian models. Dissected adult zebrafish hearts were perfused with a calcium-containing buffer, and a beat frequency was maintained with electrical stimulation. The impact of pacing frequency, flow rate and perfusate calcium concentration on ventricular performance (including end-diastolic and end-systolic volumes, ejection fraction, radial strain, and maximal velocities of shortening and relaxation) were evaluated and optimal conditions defined. We determined the effects of age on heart function in wild-type male and female zebrafish, and successfully detected hypercontractile and hypocontractile responses after adrenergic stimulation or doxorubicin treatment, respectively. Good correlations were found between indices of cardiac contractility obtained with high-frequency echocardiography and with the ex vivo technique in a subset of fish studied with both methods. The ex vivo beating heart preparation is a valuable addition to the cardiac function tool kit that will expand the use of adult zebrafish for cardiovascular research.


Aging/physiology , Heart/physiology , Perfusion/methods , Zebrafish/physiology , Animals , Cardiomyopathies/chemically induced , Cardiomyopathies/physiopathology , Doxorubicin/adverse effects , Electrocardiography , Female , Heart Ventricles/anatomy & histology , Male , Myocardial Contraction , Organ Size
8.
Prog Biophys Mol Biol ; 138: 116-125, 2018 10.
Article En | MEDLINE | ID: mdl-29884423

Hypertrophic cardiomyopathy (HCM) is usually manifested by increased myofilament Ca2+ sensitivity, excessive contractility, and impaired relaxation. In contrast, dilated cardiomyopathy (DCM) originates from insufficient sarcomere contractility and reduced cardiac pump function, subsequently resulting in heart failure. The zebrafish has emerged as a new model of human cardiomyopathy with high-throughput screening, which will facilitate the discovery of novel genetic factors and the development of new therapies. Given the small hearts of zebrafish, better phenotyping tools are needed to discern different types of cardiomyopathy, such as HCM and DCM. This article reviews the existing models of cardiomyopathy, available morphologic and functional methods, and current understanding of the different types of cardiomyopathy in adult zebrafish.


Cardiomyopathies , Disease Models, Animal , Phenotype , Zebrafish , Animals , Cardiomyopathies/pathology , Humans , Intracellular Space/metabolism
9.
Development ; 143(24): 4713-4722, 2016 12 15.
Article En | MEDLINE | ID: mdl-27836965

Titin-truncating variants (TTNtvs) are the major cause of dilated cardiomyopathy (DCM); however, allelic heterogeneity (TTNtvs in different exons) results in variable phenotypes, and remains a major hurdle for disease diagnosis and therapy. Here, we generated a panel of ttn mutants in zebrafish. Four single deletion mutants in ttn.2 or ttn.1 resulted in four phenotypes and three double ttn.2/ttn.1 mutants exhibited more severe phenotypes in somites. Protein analysis identified ttnxu071 as a near-null mutant and the other six mutants as hypomorphic alleles. Studies of ttnxu071 uncovered a function of titin in guiding the assembly of nascent myofibrils from premyofibrils. By contrast, sarcomeres were assembled in the hypomorphic ttn mutants but either became susceptible to biomechanical stresses such as contraction or degenerated during development. Further genetic studies indicated that the exon usage hypothesis, but not the toxic peptide or the Cronos hypothesis, could account for these exon-dependent effects. In conclusion, we modeled TTNtv allelic heterogeneity during development and paved the way for future studies to decipher allelic heterogeneity in adult DCM.


Connectin/genetics , Myofibrils/metabolism , Sarcomeres/metabolism , Zebrafish/growth & development , Alleles , Allelic Imbalance/genetics , Animals , Animals, Genetically Modified , Cardiomyopathy, Dilated/genetics , Connectin/metabolism , Sarcomeres/genetics , Sequence Deletion/genetics
10.
J Biol Chem ; 291(41): 21817-21828, 2016 Oct 07.
Article En | MEDLINE | ID: mdl-27557662

The cardiac troponin I (cTnI) R145W mutation is associated with restrictive cardiomyopathy (RCM). Recent evidence suggests that this mutation induces perturbed myofilament length-dependent activation (LDA) under conditions of maximal protein kinase A (PKA) stimulation. Some cardiac disease-causing mutations, however, have been associated with a blunted response to PKA-mediated phosphorylation; whether this includes LDA is unknown. Endogenous troponin was exchanged in isolated skinned human myocardium for recombinant troponin containing either cTnI R145W, PKA/PKC phosphomimetic charge mutations (S23D/S24D and T143E), or various combinations thereof. Myofilament Ca2+ sensitivity of force, tension cost, LDA, and single myofibril activation/relaxation parameters were measured. Our results show that both R145W and T143E uncouple the impact of S23D/S24D phosphomimetic on myofilament function, including LDA. Molecular dynamics simulations revealed a marked reduction in interactions between helix C of cTnC (residues 56, 59, and 63), and cTnI (residue 145) in the presence of either cTnI RCM mutation or cTnI PKC phosphomimetic. These results suggest that the RCM-associated cTnI R145W mutation induces a permanent structural state that is similar to, but more extensive than, that induced by PKC-mediated phosphorylation of cTnI Thr-143. We suggest that this structural conformational change induces an increase in myofilament Ca2+ sensitivity and, moreover, uncoupling from the impact of phosphorylation of cTnI mediated by PKA at the Ser-23/Ser-24 target sites. The R145W RCM mutation by itself, however, does not impact LDA. These perturbed biophysical and biochemical myofilament properties are likely to significantly contribute to the diastolic cardiac pump dysfunction that is seen in patients suffering from a restrictive cardiomyopathy that is associated with the cTnI R145W mutation.


Cardiomyopathy, Restrictive , Molecular Dynamics Simulation , Mutation, Missense , Sarcomeres , Troponin I , Amino Acid Substitution , Cardiomyopathy, Restrictive/genetics , Cardiomyopathy, Restrictive/metabolism , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Humans , Male , Protein Kinase C/chemistry , Protein Kinase C/genetics , Protein Kinase C/metabolism , Sarcomeres/chemistry , Sarcomeres/genetics , Sarcomeres/metabolism , Structure-Activity Relationship , Troponin I/chemistry , Troponin I/genetics , Troponin I/metabolism
11.
J Physiol ; 592(9): 1949-56, 2014 May 01.
Article En | MEDLINE | ID: mdl-24591576

The zebrafish (Danio rerio) has been used extensively in cardiovascular biology, but mainly in the study of heart development. The relative ease of its genetic manipulation may indicate the suitability of this species as a cost-effective model system for the study of cardiac contractile biology. However, whether the zebrafish heart is an appropriate model system for investigations pertaining to mammalian cardiac contractile structure-function relationships remains to be resolved. Myocytes were isolated from adult zebrafish hearts by enzymatic digestion, attached to carbon rods, and twitch force and intracellular Ca(2+) were measured. We observed the modulation of twitch force, but not of intracellular Ca(2+), by both extracellular [Ca(2+)] and sarcomere length. In permeabilized cells/myofibrils, we found robust myofilament length-dependent activation. Moreover, modulation of myofilament activation-relaxation and force redevelopment kinetics by varied Ca(2+) activation levels resembled that found previously in mammalian myofilaments. We conclude that the zebrafish is a valid model system for the study of cardiac contractile structure-function relationships.


Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Age Factors , Animals , Calcium/metabolism , Cells, Cultured , Extracellular Fluid/metabolism , Heart Ventricles/cytology , Heart Ventricles/metabolism , Zebrafish
12.
Pflugers Arch ; 465(12): 1775-84, 2013 Dec.
Article En | MEDLINE | ID: mdl-23821298

The zebrafish serves as a promising transgenic animal model that can be used to study cardiac Ca(2+) regulation. However, mechanisms of sarcoplasmic reticulum (SR) Ca(2+) handling in the zebrafish heart have not been systematically explored. We found that in zebrafish ventricular myocytes, the action potential-induced Ca(2+) transient is mainly (80 %) mediated by Ca(2+) influx via L-type Ca(2+) channels (LTCC) and only 20 % by Ca(2+) released from the SR. This small contribution of the SR to the Ca(2+) transient was not the result of depleted SR Ca(2+) load. We found that the ryanodine receptor (RyR) expression level in zebrafish myocytes was ∼72 % lower compared to rabbit myocytes. In permeabilized myocytes, increasing cytosolic [Ca(2+)] from 100 to 350 nM did not trigger SR Ca(2+) release. However, an application of a low dose of caffeine activated Ca(2+) sparks. These results show that the zebrafish cardiac RyR has low sensitivity to the mechanism of Ca(2+)-induced Ca(2+) release. Activation of protein kinase A by forskolin increased phosphorylation of the RyR in zebrafish myocardium. In half of the studied cells, an increased Ca(2+) transient by forskolin was entirely mediated by augmentation of LTCC current. In the remaining myocytes, the forskolin action was associated with an increase of both LTCC and SR Ca(2+) release. These results indicate that the mechanism of excitation-contraction coupling in zebrafish myocytes differs from the mammalian one mainly because of the small contribution of SR Ca(2+) release to the Ca(2+) transient. This difference is due to a low sensitivity of RyRs to cytosolic [Ca(2+)].


Calcium/metabolism , Excitation Contraction Coupling/physiology , Myocytes, Cardiac/physiology , Sarcoplasmic Reticulum/metabolism , Animals , Caffeine/pharmacology , Calcium Signaling/physiology , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Excitation Contraction Coupling/drug effects , Myocardium/metabolism , Rabbits , Ryanodine Receptor Calcium Release Channel/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Zebrafish
...