Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Dev Cell ; 2024 May 02.
Article in English | MEDLINE | ID: mdl-38701784

ABSTRACT

The oxidative phosphorylation (OXPHOS) system is intricately organized, with respiratory complexes forming super-assembled quaternary structures whose assembly mechanisms and physiological roles remain under investigation. Cox7a2l, also known as Scaf1, facilitates complex III and complex IV (CIII-CIV) super-assembly, enhancing energetic efficiency in various species. We examined the role of Cox7a1, another Cox7a family member, in supercomplex assembly and muscle physiology. Zebrafish lacking Cox7a1 exhibited reduced CIV2 formation, metabolic alterations, and non-pathological muscle performance decline. Additionally, cox7a1-/- hearts displayed a pro-regenerative metabolic profile, impacting cardiac regenerative response. The distinct phenotypic effects of cox7a1-/- and cox7a2l-/- underscore the diverse metabolic and physiological consequences of impaired supercomplex formation, emphasizing the significance of Cox7a1 in muscle maturation within the OXPHOS system.

2.
Sci Rep ; 13(1): 17037, 2023 10 09.
Article in English | MEDLINE | ID: mdl-37813860

ABSTRACT

Drug repurposing is an important strategy in COVID-19 treatment, but many clinically approved compounds have not been extensively studied in the context of embryogenesis, thus limiting their administration during pregnancy. Here we used the zebrafish embryo model organism to test the effects of 162 marketed drugs on cardiovascular development. Among the compounds used in the clinic for COVD-19 treatment, we found that Remdesivir led to reduced body size and heart functionality at clinically relevant doses. Ritonavir and Baricitinib showed reduced heart functionality and Molnupiravir and Baricitinib showed effects on embryo activity. Sabizabulin was highly toxic at concentrations only 5 times higher than Cmax and led to a mean mortality of 20% at Cmax. Furthermore, we tested if zebrafish could be used as a model to study inflammatory response in response to spike protein treatment and found that Remdesivir, Ritonavir, Molnupiravir, Baricitinib as well as Sabizabulin counteracted the inflammatory response related gene expression upon SARS-CoV-2 spike protein treatment. Our results show that the zebrafish allows to study immune-modulating properties of COVID-19 compounds and highlights the need to rule out secondary defects of compound treatment on embryogenesis. All results are available on a user friendly web-interface https://share.streamlit.io/alernst/covasc_dataapp/main/CoVasc_DataApp.py that provides a comprehensive overview of all observed phenotypic effects and allows personalized search on specific compounds or group of compounds. Furthermore, the presented platform can be expanded for rapid detection of developmental side effects of new compounds for treatment of COVID-19 and further viral infectious diseases.


Subject(s)
Antiviral Agents , Embryonic Development , Animals , Female , Humans , Pregnancy , Antiviral Agents/pharmacology , COVID-19 , COVID-19 Drug Treatment , Embryonic Development/drug effects , Ritonavir/pharmacology , SARS-CoV-2 , Zebrafish , Embryo, Nonmammalian/drug effects
4.
Sci Rep ; 13(1): 5418, 2023 04 03.
Article in English | MEDLINE | ID: mdl-37012284

ABSTRACT

Zebrafish have the capacity to fully regenerate the heart after an injury, which lies in sharp contrast to the irreversible loss of cardiomyocytes after a myocardial infarction in humans. Transcriptomics analysis has contributed to dissect underlying signaling pathways and gene regulatory networks in the zebrafish heart regeneration process. This process has been studied in response to different types of injuries namely: ventricular resection, ventricular cryoinjury, and genetic ablation of cardiomyocytes. However, there exists no database to compare injury specific and core cardiac regeneration responses. Here, we present a meta-analysis of transcriptomic data of regenerating zebrafish hearts in response to these three injury models at 7 days post injury (7dpi). We reanalyzed 36 samples and analyzed the differentially expressed genes (DEG) followed by downstream Gene Ontology Biological Processes (GO:BP) analysis. We found that the three injury models share a common core of DEG encompassing genes involved in cell proliferation, the Wnt signaling pathway and genes that are enriched in fibroblasts. We also found injury-specific gene signatures for resection and genetic ablation, and to a lower extent the cryoinjury model. Finally, we present our data in a user-friendly web interface that displays gene expression signatures across different injury types and highlights the importance to consider injury-specific gene regulatory networks when interpreting the results related to cardiac regeneration in the zebrafish. The analysis is freely available at: https://mybinder.org/v2/gh/MercaderLabAnatomy/PUB_Botos_et_al_2022_shinyapp_binder/HEAD?urlpath=shiny/bus-dashboard/ .


Subject(s)
Myocardial Infarction , Zebrafish , Animals , Humans , Zebrafish/metabolism , Transcriptome , Heart/physiology , Myocytes, Cardiac/metabolism , Myocardial Infarction/metabolism , Regeneration/genetics , Cell Proliferation
5.
Biol Imaging ; 3: e20, 2023.
Article in English | MEDLINE | ID: mdl-38510170

ABSTRACT

In vivo fluorescence microscopy is a powerful tool to image the beating heart in its early development stages. A high acquisition frame rate is necessary to study its fast contractions, but the limited fluorescence intensity requires sensitive cameras that are often too slow. Moreover, the problem is even more complex when imaging distinct tissues in the same sample using different fluorophores. We present Paired Alternating AcQuisitions, a method to image cyclic processes in multiple channels, which requires only a single (possibly slow) camera. We generate variable temporal illumination patterns in each frame, alternating between channel-specific illuminations (fluorescence) in odd frames and a motion-encoding brightfield pattern as a common reference in even frames. Starting from the image pairs, we find the position of each reference frame in the cardiac cycle through a combination of image-based sorting and regularized curve fitting. Thanks to these estimated reference positions, we assemble multichannel videos whose frame rate is virtually increased. We characterize our method on synthetic and experimental images collected in zebrafish embryos, showing quantitative and visual improvements in the reconstructed videos over existing nongated sorting-based alternatives. Using a 15 Hz camera, we showcase a reconstructed video containing two fluorescence channels at 100 fps.

7.
Front Cell Dev Biol ; 10: 824851, 2022.
Article in English | MEDLINE | ID: mdl-35242762

ABSTRACT

Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) was first identified in December 2019 as a novel respiratory pathogen and is the causative agent of Corona Virus disease 2019 (COVID-19). Early on during this pandemic, it became apparent that SARS-CoV-2 was not only restricted to infecting the respiratory tract, but the virus was also found in other tissues, including the vasculature. Individuals with underlying pre-existing co-morbidities like diabetes and hypertension have been more prone to develop severe illness and fatal outcomes during COVID-19. In addition, critical clinical observations made in COVID-19 patients include hypercoagulation, cardiomyopathy, heart arrythmia, and endothelial dysfunction, which are indicative for an involvement of the vasculature in COVID-19 pathology. Hence, this review summarizes the impact of SARS-CoV-2 infection on the vasculature and details how the virus promotes (chronic) vascular inflammation. We provide a general overview of SARS-CoV-2, its entry determinant Angiotensin-Converting Enzyme II (ACE2) and the detection of the SARS-CoV-2 in extrapulmonary tissue. Further, we describe the relation between COVID-19 and cardiovascular diseases (CVD) and their impact on the heart and vasculature. Clinical findings on endothelial changes during COVID-19 are reviewed in detail and recent evidence from in vitro studies on the susceptibility of endothelial cells to SARS-CoV-2 infection is discussed. We conclude with current notions on the contribution of cardiovascular events to long term consequences of COVID-19, also known as "Long-COVID-syndrome". Altogether, our review provides a detailed overview of the current perspectives of COVID-19 and its influence on the vasculature.

8.
Nat Commun ; 13(1): 1677, 2022 03 30.
Article in English | MEDLINE | ID: mdl-35354817

ABSTRACT

The mesothelium lines body cavities and surrounds internal organs, widely contributing to homeostasis and regeneration. Mesothelium disruptions cause visceral anomalies and mesothelioma tumors. Nonetheless, the embryonic emergence of mesothelia remains incompletely understood. Here, we track mesothelial origins in the lateral plate mesoderm (LPM) using zebrafish. Single-cell transcriptomics uncovers a post-gastrulation gene expression signature centered on hand2 in distinct LPM progenitor cells. We map mesothelial progenitors to lateral-most, hand2-expressing LPM and confirm conservation in mouse. Time-lapse imaging of zebrafish hand2 reporter embryos captures mesothelium formation including pericardium, visceral, and parietal peritoneum. We find primordial germ cells migrate with the forming mesothelium as ventral migration boundary. Functionally, hand2 loss disrupts mesothelium formation with reduced progenitor cells and perturbed migration. In mouse and human mesothelioma, we document expression of LPM-associated transcription factors including Hand2, suggesting re-initiation of a developmental program. Our data connects mesothelium development to Hand2, expanding our understanding of mesothelial pathologies.


Subject(s)
Mesothelioma , Zebrafish , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Epithelium/metabolism , Mesothelioma/genetics , Mice , Transcription Factors/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
9.
Development ; 149(6)2022 03 15.
Article in English | MEDLINE | ID: mdl-35312773

ABSTRACT

During development, the heart grows by addition of progenitor cells to the poles of the primordial heart tube. In the zebrafish, Wilms tumor 1 transcription factor a (wt1a) and b (wt1b) genes are expressed in the pericardium, at the venous pole of the heart. From this pericardial layer, the proepicardium emerges. Proepicardial cells are subsequently transferred to the myocardial surface and form the epicardium, covering the myocardium. We found that while wt1a and wt1b expression is maintained in proepicardial cells, it is downregulated in pericardial cells that contributes cardiomyocytes to the developing heart. Sustained wt1b expression in cardiomyocytes reduced chromatin accessibility of specific genomic loci. Strikingly, a subset of wt1a- and wt1b-expressing cardiomyocytes changed their cell-adhesion properties, delaminated from the myocardium and upregulated epicardial gene expression. Thus, wt1a and wt1b act as a break for cardiomyocyte differentiation, and ectopic wt1a and wt1b expression in cardiomyocytes can lead to their transdifferentiation into epicardial-like cells.


Subject(s)
Myocytes, Cardiac , Zebrafish , Animals , Gene Expression Regulation, Developmental , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Pericardium/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , WT1 Proteins/genetics , WT1 Proteins/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
10.
Mol Neurodegener ; 16(1): 69, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34593012

ABSTRACT

BACKGROUND: Contrasting with zebrafish, retinal regeneration from Müller cells (MCs) is largely limited in mammals, where they undergo reactive gliosis that consist of a hypertrophic response and ultimately results in vision loss. Transforming growth factor ß (TGFß) is essential for wound healing, including both scar formation and regeneration. However, targeting TGFß may affect other physiological mechanisms, owing its pleiotropic nature. The regulation of various cellular activities by TGFß relies on its interaction with other pathways including Notch. Here, we explore the interplay of TGFß with Notch and how this regulates MC response to injury in zebrafish and mice. Furthermore, we aimed to characterize potential similarities between murine and human MCs during chronic reactive gliosis. METHODS: Focal damage to photoreceptors was induced with a 532 nm diode laser in TgBAC (gfap:gfap-GFP) zebrafish (ZF) and B6-Tg (Rlbp1-GFP) mice. Transcriptomics, immunofluorescence, and flow cytometry were employed for a comparative analysis of MC response to laser-induced injury between ZF and mouse. The laser-induced injury was paired with pharmacological treatments to inhibit either Notch (DAPT) or TGFß (Pirfenidone) or TGFß/Notch interplay (SIS3). To determine if the murine laser-induced injury model translates to the human system, we compared the ensuing MC response to human donors with early retinal degeneration. RESULTS: Investigations into injury-induced changes in murine MCs revealed TGFß/Notch interplay during reactive gliosis. We found that TGFß1/2 and Notch1/2 interact via Smad3 to reprogram murine MCs towards an epithelial lineage and ultimately to form a glial scar. Similar to what we observed in mice, we confirmed the epithelial phenotype of human Müller cells during gliotic response. CONCLUSION: The study indicates a pivotal role for TGFß/Notch interplay in tuning MC stemness during injury response and provides novel insights into the remodeling mechanism during retinal degenerative diseases.


Subject(s)
Ependymoglial Cells , Gliosis , Animals , Ependymoglial Cells/metabolism , Mammals/metabolism , Mice , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Zebrafish/metabolism
11.
Zebrafish ; 18(4): 252-264, 2021 08.
Article in English | MEDLINE | ID: mdl-34227897

ABSTRACT

Metals and metalloids are integral to biological processes and play key roles in physiology and metabolism. Nonetheless, overexposure to some metals or lack of others can lead to serious health consequences. In this study, eight zebrafish facilities collaborated to generate a multielement analysis of their centralized recirculating water systems. We report a first set of average concentrations for 46 elements detected in zebrafish facilities. Our results help to establish an initial baseline for trouble-shooting purposes, and in general for safe ranges of metal concentrations in recirculating water systems, supporting reproducible scientific research outcomes with zebrafish.


Subject(s)
Metalloids , Water Pollutants, Chemical , Animals , Metalloids/analysis , Metalloids/metabolism , Water , Water Pollutants, Chemical/analysis , Zebrafish/metabolism
13.
Cell Mol Neurobiol ; 41(1): 43-62, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32219603

ABSTRACT

Müller cells may have stem cell-like capability as they regenerate photoreceptor loss upon injury in some vertebrates, but not in mammals. Indeed, mammalian Müller cells undergo major cellular and molecular changes summarized as reactive gliosis. Transforming growth factor beta (TGFß) isoforms are multifunctional cytokines that play a central role, both in wound healing and in tissue repair. Here, we studied the role of TGFß isoforms and their signaling pathways in response to injury induction during tissue regeneration in zebrafish and scar formation in mouse. Our transcriptome analysis showed a different activation of canonical and non-canonical signaling pathways and how they shaped the injury response. In particular, TGFß3 promotes retinal regeneration via Smad-dependent canonical pathway upon regulation of junb gene family and mycb in zebrafish Müller cells. However, in mice, TGFß1 and TGFß2 evoke the p38MAPK signaling pathway. The activation of this non-canonical pathway leads to retinal gliosis. Thus, the regenerative versus reparative effect of the TGFß pathway observed may rely on the activation of different signaling cascades. This provides one explanation of the different injury response in zebrafish and mouse retina.


Subject(s)
Gliosis/pathology , Retinal Degeneration/pathology , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Fibrinolysis , Fibrosis , Gliosis/complications , Gliosis/diagnostic imaging , Green Fluorescent Proteins/metabolism , Kinetics , Lasers , MAP Kinase Signaling System , Mice, Transgenic , Plasminogen Activator Inhibitor 1/metabolism , Protein Isoforms/metabolism , Regeneration , Retinal Degeneration/complications , Retinal Degeneration/diagnostic imaging , Tomography, Optical Coherence , Transforming Growth Factor beta2/metabolism , Up-Regulation , Zebrafish
14.
Methods Mol Biol ; 2158: 51-62, 2021.
Article in English | MEDLINE | ID: mdl-32857365

ABSTRACT

Zebrafish have the capacity to regenerate most of its organs upon injury, including the heart. Due to its amenability for genetic manipulation, the zebrafish is an excellent model organism to study the cellular and molecular mechanisms promoting heart regeneration. Several cardiac injury models have been developed in zebrafish, including ventricular resection, genetic ablation, and ventricular cryoinjury. This chapter provides a detailed protocol of zebrafish ventricular cryoinjury and highlights factors and critical steps to be considered when performing this method.


Subject(s)
Cardiac Surgical Procedures/adverse effects , Cryosurgery/adverse effects , Disease Models, Animal , Heart Injuries/pathology , Heart/physiology , Regeneration , Ventricular Remodeling , Animals , Cell Proliferation , Heart Injuries/etiology , Heart Injuries/rehabilitation , Zebrafish
15.
Dev Dyn ; 249(12): 1455-1469, 2020 12.
Article in English | MEDLINE | ID: mdl-33103836

ABSTRACT

BACKGROUND: The epicardium is the outer mesothelial layer of the heart. It encloses the myocardium and plays key roles in heart development and regeneration. It derives from the proepicardium (PE), cell clusters that appear in the dorsal pericardium (DP) close to the atrioventricular canal and the venous pole of the heart, and are released into the pericardial cavity. PE cells are advected around the beating heart until they attach to the myocardium. Bmp and Notch signaling influence PE formation, but it is unclear how both signaling pathways interact during this process in the zebrafish. RESULTS: Here, we show that the developing PE is influenced by Notch signaling derived from the endothelium. Overexpression of the intracellular receptor of notch in the endothelium enhances bmp expression, increases the number of pSmad1/5 positive cells in the DP and PE, and enhances PE formation. On the contrary, pharmacological inhibition of Notch1 impairs PE formation. bmp2b overexpression can rescue loss of PE formation in the presence of a Notch1 inhibitor, but Notch gain-of-function could not recover PE formation in the absence of Bmp signaling. CONCLUSIONS: Endothelial Notch signaling activates bmp expression in the heart tube, which in turn induces PE cluster formation from the DP layer.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Heart/embryology , Organogenesis/physiology , Pericardium/embryology , Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Cell Differentiation/physiology , Pericardium/metabolism , Zebrafish
16.
Cell Rep ; 32(3): 107932, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32698004

ABSTRACT

Cilia and the intraflagellar transport (IFT) proteins involved in ciliogenesis are associated with congenital heart diseases (CHDs). However, the molecular links between cilia, IFT proteins, and cardiogenesis are yet to be established. Using a combination of biochemistry, genetics, and live-imaging methods, we show that IFT complex B proteins (Ift88, Ift54, and Ift20) modulate the Hippo pathway effector YAP1 in zebrafish and mouse. We demonstrate that this interaction is key to restrict the formation of the proepicardium and the myocardium. In cellulo experiments suggest that IFT88 and IFT20 interact with YAP1 in the cytoplasm and functionally modulate its activity, identifying a molecular link between cilia-related proteins and the Hippo pathway. Taken together, our results highlight a noncanonical role for IFT complex B proteins during cardiogenesis and shed light on a mechanism of action for ciliary proteins in YAP1 regulation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Flagella/metabolism , Heart/embryology , Organogenesis , Protein Serine-Threonine Kinases/metabolism , Trans-Activators/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Biological Transport , Bone Morphogenetic Proteins/metabolism , Cilia/metabolism , HEK293 Cells , HeLa Cells , Humans , Mice, Inbred C57BL , Pericardium/metabolism , Protein Binding , Signal Transduction , YAP-Signaling Proteins
17.
Histol Histopathol ; 35(9): 1035-1046, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32633330

ABSTRACT

The epicardium is the outer mesothelial layer of the heart. It covers the myocardium and plays important roles in both heart development and regeneration. It is derived from the proepicardium (PE), groups of cells that emerges at early developmental stages from the dorsal pericardial layer (DP) close to the atrio-ventricular canal and the venous pole of the heart-tube. In zebrafish, PE cells extrude apically into the pericardial cavity as a consequence of DP tissue constriction, a process that is dependent on Bmp pathway signaling. Expression of the transcription factor Wilms tumor-1, Wt1, which is a leader of important morphogenetic events such as apoptosis regulation or epithelial-mesenchymal cell transition, is also necessary during PE formation. In this study, we used the zebrafish model to compare intensity level of the wt1a reporter line epi:GFP in PE and its original tissue, the DP. We found that GFP is present at higher intensity level in the PE tissue, and differentially wt1 expression at pericardial tissues could be involved in the PE formation process. Our results reveal that bmp2b overexpression leads to enhanced GFP level both in DP and in PE tissues.


Subject(s)
Gene Expression Regulation, Developmental , Organogenesis/genetics , Pericardium/embryology , WT1 Proteins/genetics , Zebrafish Proteins/genetics , Animals , Pericardium/metabolism , WT1 Proteins/metabolism , Zebrafish , Zebrafish Proteins/metabolism
18.
Curr Opin Genet Dev ; 64: 37-43, 2020 10.
Article in English | MEDLINE | ID: mdl-32599303

ABSTRACT

In humans, myocardial infarction results in ventricular remodeling, progressing ultimately to cardiac failure, one of the leading causes of death worldwide. In contrast to the adult mammalian heart, the zebrafish model organism has a remarkable regenerative capacity, offering the possibility to research the bases of natural regeneration. Here, we summarize recent insights into the cellular and molecular mechanisms that govern cardiac regeneration in the zebrafish.


Subject(s)
Heart/physiology , Myocytes, Cardiac/cytology , Regeneration , Zebrafish/physiology , Animals , Heart/embryology , Zebrafish/embryology
19.
EMBO Rep ; 21(7): e50287, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32496654

ABSTRACT

The oxidative phosphorylation (OXPHOS) system is a dynamic system in which the respiratory complexes coexist with super-assembled quaternary structures called supercomplexes (SCs). The physiological role of SCs is still disputed. Here, we used zebrafish to study the relevance of respiratory SCs. We combined immunodetection analysis and deep data-independent proteomics to characterize these structures and found similar SCs to those described in mice, as well as novel SCs including III2  + IV2 , I + IV, and I + III2  + IV2 . To study the physiological role of SCs, we generated two null allele zebrafish lines for supercomplex assembly factor 1 (scaf1). scaf1-/- fish displayed altered OXPHOS activity due to the disrupted interaction of complexes III and IV. scaf1-/- fish were smaller in size and showed abnormal fat deposition and decreased female fertility. These physiological phenotypes were rescued by doubling the food supply, which correlated with improved bioenergetics and alterations in the metabolic gene expression program. These results reveal that SC assembly by Scaf1 modulates OXPHOS efficiency and allows the optimization of metabolic resources.


Subject(s)
Electron Transport Complex IV , Serine-Arginine Splicing Factors/metabolism , Zebrafish , Animals , Electron Transport Complex IV/metabolism , Energy Metabolism/genetics , Female , Mice , Mitochondrial Membranes/metabolism , Oxidative Phosphorylation , Zebrafish/genetics , Zebrafish/metabolism
20.
Sci Rep ; 10(1): 2896, 2020 02 19.
Article in English | MEDLINE | ID: mdl-32076073

ABSTRACT

Myocardial infarction (MI) is a leading cause of death worldwide. Reperfusion is considered as an optimal therapy following cardiac ischemia. However, the promotion of a rapid elevation of O2 levels in ischemic cells produces high amounts of reactive oxygen species (ROS) leading to myocardial tissue injury. This phenomenon is called ischemia reperfusion injury (IRI). We aimed at identifying new and effective compounds to treat MI and minimize IRI. We previously studied heart regeneration following myocardial injury in zebrafish and described each step of the regeneration process, from the day of injury until complete recovery, in terms of transcriptional responses. Here, we mined the data and performed a deep in silico analysis to identify drugs highly likely to induce cardiac regeneration. Fisetin was identified as the top candidate. We validated its effects in an in vitro model of MI/IRI in mammalian cardiac cells. Fisetin enhances viability of rat cardiomyocytes following hypoxia/starvation - reoxygenation. It inhibits apoptosis, decreases ROS generation and caspase activation and protects from DNA damage. Interestingly, fisetin also activates genes involved in cell proliferation. Fisetin is thus a highly promising candidate drug with clinical potential to protect from ischemic damage following MI and to overcome IRI.


Subject(s)
Caspases/metabolism , Cytoprotection , Flavonoids/pharmacology , Myocardium/enzymology , Myocardium/pathology , Reactive Oxygen Species/metabolism , Animals , Animals, Newborn , Cell Death/drug effects , Cell Hypoxia/drug effects , Cell Line , Cell Proliferation/drug effects , Cytoprotection/drug effects , DNA Damage , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Flavonols , Gene Expression Regulation/drug effects , Models, Biological , Myocytes, Cardiac/drug effects , Oxygen , Rats
SELECTION OF CITATIONS
SEARCH DETAIL