Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
iScience ; 25(1): 103574, 2022 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-34988408

RESUMO

Heart disease is the leading cause of death with no method to repair damaged myocardium due to the limited proliferative capacity of adult cardiomyocytes. Curiously, mouse neonates and zebrafish can regenerate their hearts via cardiomyocyte de-differentiation and proliferation. However, a molecular mechanism of why these cardiomyocytes can re-enter cell cycle is poorly understood. Here, we identify a unique metabolic state that primes adult zebrafish and neonatal mouse ventricular cardiomyocytes to proliferate. Zebrafish and neonatal mouse hearts display elevated glutamine levels, predisposing them to amino-acid-driven activation of TOR, and that TOR activation is required for zebrafish cardiomyocyte regeneration in vivo. Through a multi-omics approach with cellular validation we identify metabolic and mitochondrial changes during the first week of regeneration. These data suggest that regeneration of zebrafish myocardium is driven by metabolic remodeling and reveals a unique metabolic regulator, TOR-primed state, in which zebrafish and mammalian cardiomyocytes are regeneration competent.

2.
Methods Mol Biol ; 2255: 77-86, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34033096

RESUMO

Three-dimensional (3D) in vitro systems closely resemble tissue microenvironments and provide predictive models for studying cytotoxic drug responses. The ability to capture the kinetic profiles of such responses in a dynamic and noninvasive way can further advance the utility of 3D cell cultures. Here, we describe the use of a luminescent lactate dehydrogenase (LDH) toxicity assay for monitoring time- and dose-dependent effects of drug treatment in 3D cancer spheroids. HCT116 spheroids formed in 96-well ultralow attachment plates were treated with increasing drug concentrations. Medium samples were collected at different timepoints, frozen, stored, and analyzed at the end of experiments using the luminescent LDH-Glo™ Assay. High assay sensitivity and low volume sampling enabled drug-induced toxicity profiling in a time- and dose-dependent manner.


Assuntos
Antineoplásicos/farmacologia , Digitonina/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais/métodos , L-Lactato Desidrogenase/metabolismo , Medições Luminescentes/métodos , Neoplasias/patologia , Esferoides Celulares/patologia , Testes de Toxicidade/métodos , Relação Dose-Resposta a Droga , Humanos , Indicadores e Reagentes/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/metabolismo , Fatores de Tempo , Células Tumorais Cultivadas
3.
Methods Mol Biol ; 2255: 187-196, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34033104

RESUMO

Comprehensive understanding of cellular responses to changes in the cellular environment or by drug treatment requires time-dependent analysis ranging from hours to several days. Here, we describe a sensitive, nonlytic live-cell assay that allows continuous or 'real-time' monitoring of cell viability, growth, and cytotoxicity over an extended period of time. We illustrate the use of the assay for small drug molecule and antibody-dependent cytotoxicity studies using cancer cells in 384-well plates. We show that the ability to measure changes in live cells over time provides instantaneous information on the biological status of the cells, information about the mode of action of the drug, and offers an added advantage of preserving the cells for multiplexing with downstream applications.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos , Apoptose , Bioensaio/métodos , Neoplasias da Mama/patologia , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Medições Luminescentes/métodos , Neoplasias da Mama/metabolismo , Feminino , Humanos , Células Tumorais Cultivadas
5.
Nat Commun ; 10(1): 4671, 2019 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-31604922

RESUMO

Mitochondrial trifunctional protein deficiency, due to mutations in hydratase subunit A (HADHA), results in sudden infant death syndrome with no cure. To reveal the disease etiology, we generated stem cell-derived cardiomyocytes from HADHA-deficient hiPSCs and accelerated their maturation via an engineered microRNA maturation cocktail that upregulated the epigenetic regulator, HOPX.  Here we report, matured HADHA mutant cardiomyocytes treated with an endogenous mixture of fatty acids manifest the disease phenotype: defective calcium dynamics and repolarization kinetics which results in a pro-arrhythmic state. Single cell RNA-seq reveals a cardiomyocyte developmental intermediate, based on metabolic gene expression. This intermediate gives rise to mature-like cardiomyocytes in control cells but, mutant cells transition to a pathological state with reduced fatty acid beta-oxidation, reduced mitochondrial proton gradient, disrupted cristae structure and defective cardiolipin remodeling. This study reveals that HADHA (tri-functional protein alpha), a monolysocardiolipin acyltransferase-like enzyme, is required for fatty acid beta-oxidation and cardiolipin remodeling, essential for functional mitochondria in human cardiomyocytes.


Assuntos
Cardiolipinas/metabolismo , Ácidos Graxos/metabolismo , Subunidade alfa da Proteína Mitocondrial Trifuncional/fisiologia , Miócitos Cardíacos/metabolismo , Cálcio/metabolismo , Linhagem Celular , Eletrofisiologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/fisiologia , Células-Tronco Embrionárias Humanas , Humanos , MicroRNAs/fisiologia , Mitocôndrias/fisiologia , Proteína Mitocondrial Trifuncional/deficiência , Subunidade alfa da Proteína Mitocondrial Trifuncional/genética , Subunidade alfa da Proteína Mitocondrial Trifuncional/metabolismo , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , Oxirredução , Técnicas de Patch-Clamp , RNA-Seq , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/fisiologia
6.
Nat Biotechnol ; 37(8): 895-906, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31375810

RESUMO

The epicardium and its derivatives provide trophic and structural support for the developing and adult heart. Here we tested the ability of human embryonic stem cell (hESC)-derived epicardium to augment the structure and function of engineered heart tissue in vitro and to improve efficacy of hESC-cardiomyocyte grafts in infarcted athymic rat hearts. Epicardial cells markedly enhanced the contractility, myofibril structure and calcium handling of human engineered heart tissues, while reducing passive stiffness compared with mesenchymal stromal cells. Transplanted epicardial cells formed persistent fibroblast grafts in infarcted hearts. Cotransplantation of hESC-derived epicardial cells and cardiomyocytes doubled graft cardiomyocyte proliferation rates in vivo, resulting in 2.6-fold greater cardiac graft size and simultaneously augmenting graft and host vascularization. Notably, cotransplantation improved systolic function compared with hearts receiving either cardiomyocytes alone, epicardial cells alone or vehicle. The ability of epicardial cells to enhance cardiac graft size and function makes them a promising adjuvant therapeutic for cardiac repair.


Assuntos
Coração/fisiologia , Células-Tronco Embrionárias Humanas , Infarto do Miocárdio/terapia , Miócitos Cardíacos , Regeneração , Animais , Embrião de Galinha , Regulação da Expressão Gênica , Humanos , Masculino , Ratos , Ratos Nus , Ratos Sprague-Dawley , Engenharia Tecidual
7.
iScience ; 2: 88-100, 2018 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-29888752

RESUMO

Cardiac development requires coordinated biphasic regulation of the WNT/ß-catenin signaling pathway. By intersecting gene expression and loss-of-function siRNA screens we identified Alpha Protein Kinase 2 (ALPK2) as a candidate negative regulator of WNT/ß-catenin signaling in cardiogenesis. In differentiating human embryonic stem cells (hESCs), ALPK2 is highly induced as hESCs transition from mesoderm to cardiac progenitors. Using antisense knockdown and CRISPR/Cas9 mutagenesis in hESCs and zebrafish, we demonstrate that ALPK2 promotes cardiac function and cardiomyocyte differentiation. Quantitative phosphoproteomics, protein expression profiling, and ß-catenin reporter assays demonstrate that loss of ALPK2 led to stabilization of ß-catenin and increased WNT signaling. Furthermore, cardiac defects attributed to ALPK2 depletion can be rescued in a dose-dependent manner by direct inhibition of WNT signaling through the small molecule XAV939. Together, these results demonstrate that ALPK2 regulates ß-catenin-dependent signaling during developmental commitment of cardiomyocytes.

8.
JACC Basic Transl Sci ; 3(6): 728-740, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30623132

RESUMO

A novel myosin heavy chain 7 mutation (E848G) identified in a familial cardiomyopathy was studied in patient-specific induced pluripotent stem cell-derived cardiomyocytes. The cardiomyopathic human induced pluripotent stem cell-derived cardiomyocytes exhibited reduced contractile function as single cells and engineered heart tissues, and genome-edited isogenic cells confirmed the pathogenic nature of the E848G mutation. Reduced contractility may result from impaired interaction between myosin heavy chain 7 and cardiac myosin binding protein C.

9.
Sensors (Basel) ; 18(1)2017 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-29283402

RESUMO

Heart disease is the leading cause of mortality in the U.S. with approximately 610,000 people dying every year. Effective therapies for many cardiac diseases are lacking, largely due to an incomplete understanding of their genetic basis and underlying molecular mechanisms. Zebrafish (Danio rerio) are an excellent model system for studying heart disease as they enable a forward genetic approach to tackle this unmet medical need. In recent years, our team has been employing electrocardiogram (ECG) as an efficient tool to study the zebrafish heart along with conventional approaches, such as immunohistochemistry, DNA and protein analyses. We have overcome various challenges in the small size and aquatic environment of zebrafish in order to obtain ECG signals with favorable signal-to-noise ratio (SNR), and high spatial and temporal resolution. In this paper, we highlight our recent efforts in zebrafish ECG acquisition with a cost-effective simplified microelectrode array (MEA) membrane providing multi-channel recording, a novel multi-chamber apparatus for simultaneous screening, and a LabVIEW program to facilitate recording and processing. We also demonstrate the use of machine learning-based programs to recognize specific ECG patterns, yielding promising results with our current limited amount of zebrafish data. Our solutions hold promise to carry out numerous studies of heart diseases, drug screening, stem cell-based therapy validation, and regenerative medicine.


Assuntos
Eletrocardiografia , Animais , Coração , Microeletrodos , Razão Sinal-Ruído , Peixe-Zebra
10.
Proc Natl Acad Sci U S A ; 113(4): 1002-7, 2016 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-26755607

RESUMO

To reveal the molecular mechanisms involved in cardiac lineage determination and differentiation, we quantified the proteome of human embryonic stem cells (hESCs), cardiac progenitor cells (CPCs), and cardiomyocytes during a time course of directed differentiation by label-free quantitative proteomics. This approach correctly identified known stage-specific markers of cardiomyocyte differentiation, including SRY-box2 (SOX2), GATA binding protein 4 (GATA4), and myosin heavy chain 6 (MYH6). This led us to determine whether our proteomic screen could reveal previously unidentified mediators of heart development. We identified Disabled 2 (DAB2) as one of the most dynamically expressed proteins in hESCs, CPCs, and cardiomyocytes. We used clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) mutagenesis in zebrafish to assess whether DAB2 plays a functional role during cardiomyocyte differentiation. We found that deletion of Dab2 in zebrafish embryos led to a significant reduction in cardiomyocyte number and increased endogenous WNT/ß-catenin signaling. Furthermore, the Dab2-deficient defects in cardiomyocyte number could be suppressed by overexpression of dickkopf 1 (DKK1), an inhibitor of WNT/ß-catenin signaling. Thus, inhibition of WNT/ß-catenin signaling by DAB2 is essential for establishing the correct number of cardiomyocytes in the developing heart. Our work demonstrates that quantifying the proteome of human stem cells can identify previously unknown developmental regulators.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Coração/embriologia , Proteômica , Proteínas Supressoras de Tumor/fisiologia , Via de Sinalização Wnt/fisiologia , beta Catenina/fisiologia , Animais , Proteínas Reguladoras de Apoptose , Diferenciação Celular , Células Cultivadas , Células-Tronco Embrionárias/citologia , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Miócitos Cardíacos/citologia , Peixe-Zebra/embriologia
11.
PLoS One ; 10(5): e0126259, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25978043

RESUMO

BACKGROUND: Maternal smoking is a risk factor for low birth weight and other adverse developmental outcomes. OBJECTIVE: We sought to determine the impact of standard tobacco cigarettes and e-cigarettes on heart development in vitro and in vivo. METHODS: Zebrafish (Danio rerio) were used to assess developmental effects in vivo and cardiac differentiation of human embryonic stem cells (hESCs) was used as a model for in vitro cardiac development. RESULTS: In zebrafish, exposure to both types of cigarettes results in broad, dose-dependent developmental defects coupled with severe heart malformation, pericardial edema and reduced heart function. Tobacco cigarettes are more toxic than e-cigarettes at comparable nicotine concentrations. During cardiac differentiation of hESCs, tobacco smoke exposure results in a delayed transition through mesoderm. Both types of cigarettes decrease expression of cardiac transcription factors in cardiac progenitor cells, suggesting a persistent delay in differentiation. In definitive human cardiomyocytes, both e-cigarette- and tobacco cigarette-treated samples showed reduced expression of sarcomeric genes such as MLC2v and MYL6. Furthermore, tobacco cigarette-treated samples had delayed onset of beating and showed low levels and aberrant localization of N-cadherin, reduced myofilament content with significantly reduced sarcomere length, and increased expression of the immature cardiac marker smooth muscle alpha-actin. CONCLUSION: These data indicate a negative effect of both tobacco cigarettes and e-cigarettes on heart development in vitro and in vivo. Tobacco cigarettes are more toxic than E-cigarettes and exhibit a broader spectrum of cardiac developmental defects.


Assuntos
Células-Tronco Embrionárias/efeitos dos fármacos , Coração/embriologia , Fumar/efeitos adversos , Animais , Relação Dose-Resposta a Droga , Desenvolvimento Embrionário/efeitos dos fármacos , Citometria de Fluxo , Imunofluorescência , Coração/efeitos dos fármacos , Coração/crescimento & desenvolvimento , Humanos , Reação em Cadeia da Polimerase , Peixe-Zebra/embriologia
12.
BMC Dev Biol ; 14: 18, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24885804

RESUMO

BACKGROUND: The outermost layer of the vertebrate heart, the epicardium, forms from a cluster of progenitor cells termed the proepicardium (PE). PE cells migrate onto the myocardium to give rise to the epicardium. Impaired epicardial development has been associated with defects in valve development, cardiomyocyte proliferation and alignment, cardiac conduction system maturation and adult heart regeneration. Zebrafish are an excellent model for studying cardiac development and regeneration; however, little is known about how the zebrafish epicardium forms. RESULTS: We report that PE migration occurs through multiple mechanisms and that the zebrafish epicardium is composed of a heterogeneous population of cells. Heterogeneity is first observed within the PE and persists through epicardium formation. Using in vivo imaging, histology and confocal microscopy, we show that PE cells migrate through a cellular bridge that forms between the pericardial mesothelium and the heart. We also observed the formation of PE aggregates on the pericardial surface, which were released into the pericardial cavity. It was previously reported that heartbeat-induced pericardiac fluid advections are necessary for PE cluster formation and subsequent epicardium development. We manipulated heartbeat genetically and pharmacologically and found that PE clusters clearly form in the absence of heartbeat. However, when heartbeat was inhibited the PE failed to migrate to the myocardium and the epicardium did not form. We isolated and cultured hearts with only a few epicardial progenitor cells and found a complete epicardial layer formed. However, pharmacologically inhibiting contraction in culture prevented epicardium formation. Furthermore, we isolated control and silent heart (sih) morpholino (MO) injected hearts prior to epicardium formation (60 hpf) and co-cultured these hearts with "donor" hearts that had an epicardium forming (108 hpf). Epicardial cells from donor hearts migrated on to control but not sih MO injected hearts. CONCLUSIONS: Epicardial cells stem from a heterogeneous population of progenitors, suggesting that the progenitors in the PE have distinct identities. PE cells attach to the heart via a cellular bridge and free-floating cell clusters. Pericardiac fluid advections are not necessary for the development of the PE cluster, however heartbeat is required for epicardium formation. Epicardium formation can occur in culture without normal hydrodynamic and hemodynamic forces, but not without contraction.


Assuntos
Movimento Celular , Modelos Biológicos , Pericárdio/citologia , Células-Tronco/citologia , Animais , Animais Geneticamente Modificados , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Frequência Cardíaca/fisiologia , Imuno-Histoquímica , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Microscopia Confocal , Organogênese , Pericárdio/embriologia , Pericárdio/fisiologia , Células-Tronco/metabolismo , Fatores de Tempo , Técnicas de Cultura de Tecidos , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Peixe-Zebra/fisiologia
13.
Int J Dev Biol ; 58(9): 693-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25896205

RESUMO

The transcription factor SOX9 is a member of the SRY-related high-mobility-group box (SOX) superfamily of genes. In mammals, Sox9 plays important roles in many developmental processes including craniofacial, skeletal and heart morphogenesis, retinal and brain development, and gonad differentiation. Human mutations in SOX9 or the SOX9 promoter result in campomelic dysplasia, a severe genetic disorder, which disrupts skeletal, craniofacial, cardiac, neural and reproductive development. Due to the duplication of the teleost fish genome, zebrafish (Danio rerio) have two Sox9 genes: sox9a and sox9b. Loss of sox9b in zebrafish results in loss of function phenotypes that are similar to those observed in humans and mice. In order to generate a transgenic sox9b:EGFP reporter line, we cloned a 2450 bp fragment of the sox9b promoter and fused it to an EGFP reporter. Consistent with reported sox9b expression and function, we observed sox9b:EGFP in the developing heart, skeletal and craniofacial structures, brain, retina, and ovaries. Our resulting transgenic line is a useful tool for identifying and studying sox9b function in development and visualizing a number of zebrafish organs and tissues in which sox9b is normally expressed.


Assuntos
Animais Geneticamente Modificados/crescimento & desenvolvimento , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/metabolismo , Fatores de Transcrição SOX9/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/crescimento & desenvolvimento , Animais , Animais Geneticamente Modificados/genética , Animais Geneticamente Modificados/metabolismo , Encéfalo/embriologia , Encéfalo/metabolismo , Anormalidades Craniofaciais/embriologia , Anormalidades Craniofaciais/metabolismo , Feminino , Proteínas de Fluorescência Verde/genética , Coração/embriologia , Coração/fisiologia , Humanos , Técnicas Imunoenzimáticas , Hibridização In Situ , Camundongos , Músculo Esquelético/embriologia , Músculo Esquelético/metabolismo , Ovário/embriologia , Ovário/metabolismo , Retina/embriologia , Retina/metabolismo , Fatores de Transcrição SOX9/genética , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
14.
Mol Pharmacol ; 84(3): 353-60, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23775563

RESUMO

Activation of the transcription factor aryl hydrocarbon receptor by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) prevents the formation of the epicardium and leads to severe heart malformations in developing zebrafish (Danio rerio). The downstream genes that cause heart malformation are not known. Because TCDD causes craniofacial malformations in zebrafish by downregulating the sox9b gene, we hypothesized that cardiotoxicity might also result from sox9b downregulation. We found that sox9b is expressed in the developing zebrafish heart ventricle and that TCDD exposure markedly reduces this expression. Furthermore, we found that manipulation of sox9b expression could phenocopy many but not all of the effects of TCDD at the heart. Loss of sox9b prevented the formation of epicardium progenitors comprising the proepicardium on the pericardial wall, and prevented the formation and migration of the epicardial layer around the heart. Zebrafish lacking sox9b showed pericardial edema, an elongated heart, and reduced blood circulation. Fish lacking sox9b failed to form valve cushions and leaflets. Sox9b is one of two mammalian Sox9 homologs, sox9b and sox9a. Knock down of sox9a expression did not cause cardiac malformations, or defects in epicardium development. We conclude that the decrease in sox9b expression in the heart caused by TCDD plays a role in many of the observed signs of cardiotoxicity. We find that while sox9b is expressed in myocardial cells, it is not normally expressed in the affected epicardial cells or progenitors. We therefore speculate that sox9b is involved in signals between the cardiomyocytes and the nascent epicardial cells.


Assuntos
Anormalidades Induzidas por Medicamentos/metabolismo , Cardiopatias Congênitas/induzido quimicamente , Pericárdio/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Fatores de Transcrição SOX9/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Anormalidades Induzidas por Medicamentos/fisiopatologia , Animais , Circulação Coronária , Regulação para Baixo , Edema/induzido quimicamente , Edema/metabolismo , Cardiopatias Congênitas/metabolismo , Cardiopatias Congênitas/fisiopatologia , Valvas Cardíacas/anormalidades , Valvas Cardíacas/efeitos dos fármacos , Valvas Cardíacas/embriologia , Valvas Cardíacas/crescimento & desenvolvimento , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/embriologia , Ventrículos do Coração/crescimento & desenvolvimento , Ventrículos do Coração/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Pericárdio/embriologia , Pericárdio/crescimento & desenvolvimento , Pericárdio/metabolismo , Peixe-Zebra
15.
Toxicol Sci ; 131(2): 558-67, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23135548

RESUMO

Embryonic exposure to the environmental contaminant and aryl hydrocarbon receptor agonist, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, dioxin), disrupts cardiac development and function in fish, birds, and mammals. In zebrafish, the temporal window of sensitivity to the cardiotoxic effects of TCDD coincides with epicardium formation. We hypothesized that this TCDD-induced heart failure results from disruption of epicardial development. To determine whether embryonic TCDD exposure inhibits epicardium and proepicardium (PE) development in zebrafish, we used histology and fluorescence immunocytochemistry to examine the epicardium formation in fish exposed to TCDD. TCDD exposure prevented epicardium formation. Using live imaging and in situ hybridization, we found that TCDD exposure blocked the formation of the PE cluster. In situ hybridization experiments showed that TCDD exposure also prevented the expression of the PE marker tcf21 at the site where the PE normally forms. TCDD also inhibited expansion of the epicardial layer across the developing heart: Exposure after PE formation was completed prevented further expansion of the epicardium. However, TCDD exposure did not affect epicardial cells already present. Because TCDD blocks epicardium formation, but is not directly toxic to the epicardium once complete, we propose that inhibition of epicardium formation can account for the window of sensitivity to TCDD cardiotoxicity in developing zebrafish. Epicardium development is crucial to heart development. Loss of this layer during development may account for most if not all of the TCDD-induced cardiotoxicity in zebrafish.


Assuntos
Pericárdio/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Animais , Pericárdio/embriologia , Peixe-Zebra/embriologia
16.
Toxicol Sci ; 132(1): 211-21, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23204111

RESUMO

Normal adult zebrafish can completely regenerate lost myocardium following partial amputation of the ventricle apex. We report that 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) significantly impairs this regeneration. Adult male zebrafish were injected with vehicle (control) or TCDD (70ng/g, ip) 1 day prior to partial amputation of the ventricle apex. Gross observation and histological analysis of the amputated heart at 21 days postamputation revealed that TCDD-exposed fish had not progressed beyond the initial clot formation stage, whereas the vehicle control fish showed substantial recovery and almost complete resolution of the formed clot. In contrast, hearts that were not surgically wounded showed no signs of TCDD toxicity. Striking features in the TCDD-exposed hearts were the absence of the normal sheath of new tissue enveloping the wound and the absence of intense cell proliferation at the site of the wound. In addition, the patterns of collagen deposition at the wound site were different between the TCDD and vehicle groups. Because the receptor for TCDD is the aryl hydrocarbon receptor ligand-activated transcriptional regulator, we examined the effects of TCDD exposure on gene expression in the ventricle using DNA microarrays. Samples were collected just prior to amputation and at 6h and 7 days postamputation. TCDD-pretreated hearts had dysregulated expression of genes involved in heart function, tissue regeneration, cell growth, and extracellular matrix. Because embryonic, but not adult, hearts are major targets for TCDD-induced cardiotoxicity, we speculate that the need for embryonic-like cells in regeneration is connected with the effects of TCDD in inhibiting the response to wounding.


Assuntos
Coração/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Regeneração/efeitos dos fármacos , Aldeído Oxirredutases/genética , Animais , Proliferação de Células/efeitos dos fármacos , Colágeno/metabolismo , Coração/fisiologia , Hibridização In Situ , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , Regulação para Cima , Peixe-Zebra
17.
J Dev Biol ; 1(2): 112-125, 2013 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-25232532

RESUMO

Zebrafish (Danio rerio) are an excellent vertebrate model for studying heart development, regeneration and cardiotoxicity. Zebrafish embryos exposed during the temporal window of epicardium development to the aryl hydrocarbon receptor (AHR) agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exhibit severe heart malformations. TCDD exposure prevents both proepicardial organ (PE) and epicardium development. Exposure later in development, after the epicardium has formed, does not produce cardiac toxicity. It is not until the adult zebrafish heart is stimulated to regenerate does TCDD again cause detrimental effects. TCDD exposure prior to ventricular resection prevents cardiac regeneration. It is likely that TCDD-induced inhibition of epicardium development and cardiac regeneration occur via a common mechanism. Here, we describe experiments that focus on the epicardium as a target and sensor of zebrafish heart toxicity.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...