1
|
Lei Y, Zhou X, Yao N, Zhang H, Huang X, Du Z, Tian G, Zhang W, Cheng B, Luo Q. Nitenpyram-Induced Cardiac Toxicity in Early Developmental Stages of Zebrafish. J Appl Toxicol 2025. [PMID: 40223166 DOI: 10.1002/jat.4783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/07/2025] [Accepted: 03/28/2025] [Indexed: 04/15/2025]
Abstract
Nitenpyram is a new neonicotinoid insecticide primarily used for the control of piercing-sucking pests as well as fleas and lice on animals. It is widely used around the world, and the concentrations and detection rates in surface water and groundwater are increasing. Current research has shown that nitenpyram is toxic to honey bee, silkworms, and other organisms, but the toxicity to aquatic organisms and the mechanisms of its toxicity are still unclear. This study uses zebrafish as the research model to investigate the adverse effects of nitenpyram on the early embryonic development of zebrafish. The results show that nitenpyram induces developmental toxicity and cardiac toxicity in zebrafish. Zebrafish exhibited pericardial edema and an increased distance between the atria and ventricles. Quantitative real-time PCR (qRT-PCR) analysis revealed that the expression of heart development-related genes tbx2b, gata4, vmhc, myh6, and nkx2.5 was inhibited. Additionally, nitenpyram induced oxidative stress in zebrafish, significantly increasing the production and accumulation of ROS in the cardiac region. Acridine orange staining results revealed that nitenpyram induced apoptosis in the cardiac region of zebrafish, and qRT-PCR results showed activation of the endogenous apoptosis pathways. Therefore, it is speculated that nitenpyram-induced oxidative stress alters the expression of heart-related genes and triggers endogenous apoptosis in zebrafish cardiomyocytes, ultimately leading to abnormal cardiac development.
Collapse
Affiliation(s)
- Yan Lei
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, Jiangxi, China
| | - Xin Zhou
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, Jiangxi, China
| | - Nianming Yao
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, Jiangxi, China
| | - Hua Zhang
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, Jiangxi, China
| | - Xinyue Huang
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, Jiangxi, China
| | - Zishan Du
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, Jiangxi, China
| | - Guiyou Tian
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, Jiangxi, China
| | - Weixin Zhang
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, Jiangxi, China
| | - Bo Cheng
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, Jiangxi, China
| | - Qiang Luo
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, Jiangxi, China
| |
Collapse
|
2
|
Melnikov K, Kaiglová A, Kucharíková S. The use of the model organism Caenorhabditis elegans in the investigation of the adverse effects of electronic cigarettes. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 291:117853. [PMID: 39919590 DOI: 10.1016/j.ecoenv.2025.117853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/28/2025] [Accepted: 02/02/2025] [Indexed: 02/09/2025]
Abstract
The use of tobacco products is one of the most preventable risk factors for mortality from a variety of diseases, including cardiovascular, infectious, respiratory, and neoplastic conditions. The use of electronic cigarettes (ECIGs), also known as electronic nicotine delivery devices, has increased significantly in recent years. Nicotine, propylene glycol, and / or glycerine, water, alcohol, flavorings, and other substances are among the many chemicals found in ECIGs that are vaporized and inhaled. A review of the existing literature shows that research dedicated to ECIGs is a rapidly developing and growing field of study. The rationale for the use of ECIGs is that they represent a safer alternative to traditional tobacco products. However, vaping safety profiles are still under development, as this is a relatively recent phenomenon. Various model organisms can be employed to examine the cellular processes that may be altered by exposure to the electronic liquids utilized for vaping. For example, the translucent multicellular eukaryote Caenorhabditis elegans is widely used as a model organism to explain a broad range of biological processes, including aging, stress response, development, and many others. Due to its short lifespan and easy use, C. elegans is an ideal model organism for studying chronic exposure to drugs and environmental toxicology. This review presents a summary of the most recent findings on the impact of electronic cigarettes on the physiological health of this nematode. Preliminary observations made in C. elegans can provide insight into the consequences of exposure to fundamental cellular physiology, which can then be used for future research in humans and mammalian models.
Collapse
Affiliation(s)
- Kamila Melnikov
- Department of Laboratory Medicine, Faculty of Health Care and Social Work, Trnava University in Trnava, Univerzitné námestie 1, Trnava 918 43, Slovakia
| | - Alžbeta Kaiglová
- Department of Laboratory Medicine, Faculty of Health Care and Social Work, Trnava University in Trnava, Univerzitné námestie 1, Trnava 918 43, Slovakia
| | - Soňa Kucharíková
- Department of Laboratory Medicine, Faculty of Health Care and Social Work, Trnava University in Trnava, Univerzitné námestie 1, Trnava 918 43, Slovakia.
| |
Collapse
|
3
|
Saberigarakani A, Patel RP, Almasian M, Zhang X, Brewer J, Hassan SS, Chai J, Lee J, Fei B, Yuan J, Carroll K, Ding Y. Volumetric imaging and computation to explore contractile function in zebrafish hearts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.14.623621. [PMID: 39605398 PMCID: PMC11601419 DOI: 10.1101/2024.11.14.623621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Despite advancements in cardiovascular engineering, heart diseases remain a leading cause of mortality. The limited understanding of the underlying mechanisms of cardiac dysfunction at the cellular level restricts the development of effective screening and therapeutic methods. To address this, we have developed a framework that incorporates light field detection and individual cell tracking to capture real-time volumetric data in zebrafish hearts, which share structural and electrical similarities with the human heart and generate 120 to 180 beats per minute. Our results indicate that the in-house system achieves an acquisition speed of 200 volumes per second, with resolutions of up to 5.02 ± 0.54 µm laterally and 9.02 ± 1.11 µm axially across the entire depth, using the estimated-maximized-smoothed deconvolution method. The subsequent deep learning-based cell trackers enable further investigation of contractile dynamics, including cellular displacement and velocity, followed by volumetric tracking of specific cells of interest from end-systole to end-diastole in an interactive environment. Collectively, our strategy facilitates real-time volumetric imaging and assessment of contractile dynamics across the entire ventricle at the cellular resolution over multiple cycles, providing significant potential for exploring intercellular interactions in both health and disease.
Collapse
|
4
|
Guo K, Kalyviotis K, Pantazis P, Rowlands CJ. Hyperspectral oblique plane microscopy enables spontaneous, label-free imaging of biological dynamic processes in live animals. Proc Natl Acad Sci U S A 2024; 121:e2404232121. [PMID: 39401353 PMCID: PMC11513980 DOI: 10.1073/pnas.2404232121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 08/21/2024] [Indexed: 10/30/2024] Open
Abstract
Spontaneous Raman imaging has emerged as powerful label-free technique for investigating the molecular composition of medicines and biological specimens. Although Raman imaging can facilitate understanding of complex biological phenomena in vivo, current imaging modalities are limited in speed and sample compatibility. Here, we introduce a single-objective line-scanning light-sheet microscope, named [Formula: see text]-OPM, which records Raman images on a timescale of minutes to seconds. To demonstrate its function, we use [Formula: see text]-OPM to map and identify microplastic particles based on their Raman spectral characteristics. In live zebrafish embryos, we show that [Formula: see text]-OPM can capture wound dynamics at five-minute intervals, revealing rapid changes in cellular and extracellular matrix composition in the wounded region. Finally, we use [Formula: see text]-OPM to synchronize and average 36,800 individual frames to obtain hyperspectral videos of a zebrafish embryo's beating heart at an effective 28 frames per second, recording compositional changes throughout the cardiac cycle.
Collapse
Affiliation(s)
- Ke Guo
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, United Kingdom
| | | | - Periklis Pantazis
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, United Kingdom
| | | |
Collapse
|
5
|
Risato G, Brañas Casas R, Cason M, Bueno Marinas M, Pinci S, De Gaspari M, Visentin S, Rizzo S, Thiene G, Basso C, Pilichou K, Tiso N, Celeghin R. In Vivo Approaches to Understand Arrhythmogenic Cardiomyopathy: Perspectives on Animal Models. Cells 2024; 13:1264. [PMID: 39120296 PMCID: PMC11311808 DOI: 10.3390/cells13151264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a hereditary cardiac disorder characterized by the gradual replacement of cardiomyocytes with fibrous and adipose tissue, leading to ventricular wall thinning, chamber dilation, arrhythmias, and sudden cardiac death. Despite advances in treatment, disease management remains challenging. Animal models, particularly mice and zebrafish, have become invaluable tools for understanding AC's pathophysiology and testing potential therapies. Mice models, although useful for scientific research, cannot fully replicate the complexity of the human AC. However, they have provided valuable insights into gene involvement, signalling pathways, and disease progression. Zebrafish offer a promising alternative to mammalian models, despite the phylogenetic distance, due to their economic and genetic advantages. By combining animal models with in vitro studies, researchers can comprehensively understand AC, paving the way for more effective treatments and interventions for patients and improving their quality of life and prognosis.
Collapse
Affiliation(s)
- Giovanni Risato
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
- Department of Biology, University of Padua, I-35131 Padua, Italy;
- Department of Women’s and Children’s Health, University of Padua, I-35128 Padua, Italy;
| | | | - Marco Cason
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Maria Bueno Marinas
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Serena Pinci
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Monica De Gaspari
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Silvia Visentin
- Department of Women’s and Children’s Health, University of Padua, I-35128 Padua, Italy;
| | - Stefania Rizzo
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Gaetano Thiene
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Cristina Basso
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Kalliopi Pilichou
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Natascia Tiso
- Department of Biology, University of Padua, I-35131 Padua, Italy;
| | - Rudy Celeghin
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| |
Collapse
|
6
|
Yu X, Zhao W, Zou Q, Wang L. Amphiphilic hydroxyethyl starch-based nanoparticles carrying linoleic acid modified berberine inhibit the expression of kras v12 oncogene in zebrafish. Biomed Pharmacother 2024; 176:116798. [PMID: 38795642 DOI: 10.1016/j.biopha.2024.116798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/14/2024] [Accepted: 05/20/2024] [Indexed: 05/28/2024] Open
Abstract
Cancer is one of the most lethal diseases all over the world. Despite that many drugs have been developed for cancer therapy, they still suffer from various limitations including poor treating efficacy, toxicity to normal human cells, and the emergence of multidrug resistance. In this study, the amphiphilic LHES polymers were prepared using hydroxyethyl starch (HES) and linoleic acid as starting materials. The content and substitution degree of linoleic acid groups in LHES polymers were analyzed. The LHES polymers were used for fabricating LHES-B nanoparticles carrying a linoleic acid modified berberine derivative (L-BBR). The LHES-B nanoparticles showed high drug loading efficiency (29%) and could quickly release L-BBR under acidic pH condition (pH = 4.5). Biological investigations revealed that LHES-B nanoparticles significantly inhibited the proliferation of HepG2 cells and exhibited higher cytotoxicity than L-BBR. In a transgenic Tg(fabp10:rtTA2s-M2; TRE2:EGFP-krasv12) zebrafish model, LHES-B nanoparticles obviously inhibited the expression of krasv12 oncogene. These results indicated that LHES carriers could improve the anticancer activity of L-BBR, and the synthesized LHES-B nanoparticles showed great potential as anticancer drug.
Collapse
Affiliation(s)
- Xiaoming Yu
- Cancer Center, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, China
| | - Wenbin Zhao
- Shandong Shangyuan Environmental Protection Technology Co. Ltd., Jinan 250100, China
| | - Qinglin Zou
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China.
| | - Lizhen Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China.
| |
Collapse
|
7
|
Shorbaji A, Pushparaj PN, Bakhashab S, Al-Ghafari AB, Al-Rasheed RR, Siraj Mira L, Basabrain MA, Alsulami M, Abu Zeid IM, Naseer MI, Rasool M. Current genetic models for studying congenital heart diseases: Advantages and disadvantages. Bioinformation 2024; 20:415-429. [PMID: 39132229 PMCID: PMC11309114 DOI: 10.6026/973206300200415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 08/13/2024] Open
Abstract
Congenital heart disease (CHD) encompasses a diverse range of structural and functional anomalies that affect the heart and the major blood vessels. Epidemiological studies have documented a global increase in CHD prevalence, which can be attributed to advancements in diagnostic technologies. Extensive research has identified a plethora of CHD-related genes, providing insights into the biochemical pathways and molecular mechanisms underlying this pathological state. In this review, we discuss the advantages and challenges of various In vitro and in vivo CHD models, including primates, canines, Xenopus frogs, rabbits, chicks, mice, Drosophila, zebrafish, and induced pluripotent stem cells (iPSCs). Primates are closely related to humans but are rare and expensive. Canine models are costly but structurally comparable to humans. Xenopus frogs are advantageous because of their generation of many embryos, ease of genetic modification, and cardiac similarity. Rabbits mimic human physiology but are challenging to genetically control. Chicks are inexpensive and simple to handle; however, cardiac events can vary among humans. Mice differ physiologically, while being evolutionarily close and well-resourced. Drosophila has genes similar to those of humans but different heart structures. Zebrafish have several advantages, including high gene conservation in humans and physiological cardiac similarities but limitations in cross-reactivity with mammalian antibodies, gene duplication, and limited embryonic stem cells for reverse genetic methods. iPSCs have the potential for gene editing, but face challenges in terms of 2D structure and genomic stability. CRISPR-Cas9 allows for genetic correction but requires high technical skills and resources. These models have provided valuable knowledge regarding cardiac development, disease simulation, and the verification of genetic factors. This review highlights the distinct features of various models with respect to their biological characteristics, vulnerability to developing specific heart diseases, approaches employed to induce particular conditions, and the comparability of these species to humans. Therefore, the selection of appropriate models is based on research objectives, ultimately leading to an enhanced comprehension of disease pathology and therapy.
Collapse
Affiliation(s)
- Ayat Shorbaji
- Biochemistry Department, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Peter Natesan Pushparaj
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sherin Bakhashab
- Biochemistry Department, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ayat B Al-Ghafari
- Biochemistry Department, King Abdulaziz University, Jeddah, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rana R Al-Rasheed
- Experimental Biochemistry Unit, King Fahad research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Loubna Siraj Mira
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Abdullah Basabrain
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Majed Alsulami
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Isam M Abu Zeid
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Muhammad Imran Naseer
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mahmood Rasool
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
8
|
Paquette SE, Oduor CI, Gaulke A, Stefan S, Bronk P, Dafonseca V, Barulin N, Lee C, Carley R, Morrison AR, Choi BR, Bailey JA, Plavicki JS. Loss of developmentally derived Irf8+ macrophages promotes hyperinnervation and arrhythmia in the adult zebrafish heart. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.17.589909. [PMID: 38659956 PMCID: PMC11042273 DOI: 10.1101/2024.04.17.589909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Recent developments in cardiac macrophage biology have broadened our understanding of the critical functions of macrophages in the heart. As a result, there is further interest in understanding the independent contributions of distinct subsets of macrophage to cardiac development and function. Here, we demonstrate that genetic loss of interferon regulatory factor 8 (Irf8)-positive embryonic-derived macrophages significantly disrupts cardiac conduction, chamber function, and innervation in adult zebrafish. At 4 months post-fertilization (mpf), homozygous irf8st96/st96 mutants have significantly shortened atrial action potential duration and significant differential expression of genes involved in cardiac contraction. Functional in vivo assessments via electro- and echocardiograms at 12 mpf reveal that irf8 mutants are arrhythmogenic and exhibit diastolic dysfunction and ventricular stiffening. To identify the molecular drivers of the functional disturbances in irf8 null zebrafish, we perform single cell RNA sequencing and immunohistochemistry, which reveal increased leukocyte infiltration, epicardial activation, mesenchymal gene expression, and fibrosis. Irf8 null hearts are also hyperinnervated and have aberrant axonal patterning, a phenotype not previously assessed in the context of cardiac macrophage loss. Gene ontology analysis supports a novel role for activated epicardial-derived cells (EPDCs) in promoting neurogenesis and neuronal remodeling in vivo. Together, these data uncover significant cardiac abnormalities following embryonic macrophage loss and expand our knowledge of critical macrophage functions in heart physiology and governing homeostatic heart health.
Collapse
Affiliation(s)
- Shannon E. Paquette
- Department of Pathology & Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Cliff I. Oduor
- Department of Pathology & Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Amy Gaulke
- Department of Pathology & Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Sabina Stefan
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Peter Bronk
- Cardiovascular Research Center, Brown University Warren Alpert Medical School, Providence, RI, 02912, USA
| | - Vanny Dafonseca
- Department of Pathology & Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Nikolai Barulin
- Department of Pathology & Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Cadence Lee
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI, 02908, USA
- Ocean State Research Institute, Inc., Providence, RI, 02908, USA
| | - Rachel Carley
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI, 02908, USA
- Ocean State Research Institute, Inc., Providence, RI, 02908, USA
| | - Alan R. Morrison
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI, 02908, USA
- Ocean State Research Institute, Inc., Providence, RI, 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Bum-Rak Choi
- Cardiovascular Research Center, Brown University Warren Alpert Medical School, Providence, RI, 02912, USA
| | - Jeffrey A. Bailey
- Department of Pathology & Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Jessica S. Plavicki
- Department of Pathology & Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| |
Collapse
|
9
|
Hasan M, Zedan HT, Al-Fakhroo D, Elsayed Ibrahim H, Abiib SI, El-Sherbiny IM, Yalcin HC. In vivo testing of novel nitric oxide-releasing nanoparticles for alleviating heart failure using the zebrafish embryo model. Nitric Oxide 2024; 144:47-57. [PMID: 38307377 DOI: 10.1016/j.niox.2024.01.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 12/27/2023] [Accepted: 01/27/2024] [Indexed: 02/04/2024]
Abstract
Heart failure (HF) is a multifactorial, heterogeneous systemic disease that is considered one of the leading causes of death and morbidity worldwide. It is well-known that endothelial dysfunction (ED) plays an important role in cardiac disease etiology. A reduction in the bioavailability of nitric oxide (NO) in the bloodstream leads to vasoconstriction and ED. Many studies indicated diminishment of peripheral arteries vasodilation that is mediated by the endothelium in the of patients with chronic HF. With the advancement of nanomedicine, nanotechnology can provide adequate solutions for delivering exogenous NO with the aid of nanoparticles (NPs) to treat ED. The properties of superparamagnetic iron oxide nanoparticles (SPIONs) enable both passive and active delivery of drugs. This prompted us to investigate the efficacy of our newly-developed hydrogel nanoparticles (NO-RPs) for the delivery and sustained release of NO gas to alleviate cardiac failure and inflammation in the heart failure zebrafish model. The hydrogel NO-RPs incorporate SPIONS and NO precursor. The sustainend release of NO in the NO-RPs (4200 s), overcomes the problem of the short half life of NO in vivo which is expected to ameliorate the reduced NO bioavailabilty, and its consequences in endothelial and cardiac dysfunction. Zebrafish embryos were used as the animal model in this study to determine the effect of SPIONs-loaded NO-RPs on the cardiovascular system. Cardiac failure was induced in 24hpf embryos by exposure to aristolochic acid (AA)(0.25, 0.5 μM) for 8 h, followed by the SPIONs-loaded NO-RPs (0.25, 0.5 mg/ml) for 48 h, experimental groups included: control group which is healthy non treated zebrafish embryos, AA injured zebrafish embryos (HF) model,and NO-RP treated HF zebrafish embryos. Survival rate was assessed at 72hpf. Cardiac function was also evaluated by analyzing cardiac parameters including heartbeat, major blood vessels primordial cardinal vein and dorsal aorta (PCV &DA) diameter, blood flow velocity in PCV & DA vessels, cardiac output, and PCV & DA shear stresses. All cardiac parameters were analyzed with the aid of MicroZebraLab blood flow analysis software from Viewpoint. In addition, we studied the molecular effects of the developed NO-RPs on the mRNA expression of selected pro-inflammatory markers: IL-6, and Cox-2. Our findings demonstrated that the NO-RPs improved the survival rate in the heart failure zebrafish model and reversed heart failure by enhancing blood flow perfusion in Zebrafish embryos, significantly. In addition, RT-PCR results showed that the NO-RPs significantly reduced the expression of pro-inflammatory markers (lL-6&COX-2) in the heart failure zebrafish model. Our study confirmed that the developed SPIONs-loaded NO-RPs are effective tool to alleviate cardiac failure and inflammation in the HF zebrafish model.
Collapse
Affiliation(s)
- Maram Hasan
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Hadeel T Zedan
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar; Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Dana Al-Fakhroo
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar; Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Hend Elsayed Ibrahim
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar; Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Sumaya Ibrahim Abiib
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Ibrahim M El-Sherbiny
- Nanomedicine Research Labs, Center for Materials Science, Zewail City of Science and Technology, 6 of October City, 12578, Giza, Egypt
| | - Huseyin C Yalcin
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar; Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar.
| |
Collapse
|
10
|
Zhang X, Saberigarakani A, Almasian M, Hassan S, Nekkanti M, Ding Y. 4D Light-sheet Imaging of Zebrafish Cardiac Contraction. J Vis Exp 2024:10.3791/66263. [PMID: 38251787 PMCID: PMC10939705 DOI: 10.3791/66263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024] Open
Abstract
Zebrafish is an intriguing model organism known for its remarkable cardiac regeneration capacity. Studying the contracting heart in vivo is essential for gaining insights into structural and functional changes in response to injuries. However, obtaining high-resolution and high-speed 4-dimensional (4D, 3D spatial + 1D temporal) images of the zebrafish heart to assess cardiac architecture and contractility remains challenging. In this context, an in-house light-sheet microscope (LSM) and customized computational analysis are used to overcome these technical limitations. This strategy, involving LSM system construction, retrospective synchronization, single cell tracking, and user-directed analysis, enables one to investigate the micro-structure and contractile function across the entire heart at the single-cell resolution in the transgenic Tg(myl7:nucGFP) zebrafish larvae. Additionally, we are able to further incorporate microinjection of small molecule compounds to induce cardiac injury in a precise and controlled manner. Overall, this framework allows one to track physiological and pathophysiological changes, as well as the regional mechanics at the single-cell level during cardiac morphogenesis and regeneration.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Department of Bioengineering, The University of Texas at Dallas
| | | | - Milad Almasian
- Department of Bioengineering, The University of Texas at Dallas
| | - Sohail Hassan
- Department of Bioengineering, The University of Texas at Dallas
| | - Manasa Nekkanti
- Department of Bioengineering, The University of Texas at Dallas
| | - Yichen Ding
- Department of Bioengineering, The University of Texas at Dallas; Center for Imaging and Surgical Innovation, The University of Texas at Dallas; Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center;
| |
Collapse
|
11
|
Hussen E, Aakel N, Shaito AA, Al-Asmakh M, Abou-Saleh H, Zakaria ZZ. Zebrafish ( Danio rerio) as a Model for the Study of Developmental and Cardiovascular Toxicity of Electronic Cigarettes. Int J Mol Sci 2023; 25:194. [PMID: 38203365 PMCID: PMC10779276 DOI: 10.3390/ijms25010194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/03/2023] [Accepted: 11/08/2023] [Indexed: 01/12/2024] Open
Abstract
The increasing popularity of electronic cigarettes (e-cigarettes) as an alternative to conventional tobacco products has raised concerns regarding their potential adverse effects. The cardiovascular system undergoes intricate processes forming the heart and blood vessels during fetal development. However, the precise impact of e-cigarette smoke and aerosols on these delicate developmental processes remains elusive. Previous studies have revealed changes in gene expression patterns, disruptions in cellular signaling pathways, and increased oxidative stress resulting from e-cigarette exposure. These findings indicate the potential for e-cigarettes to cause developmental and cardiovascular harm. This comprehensive review article discusses various aspects of electronic cigarette use, emphasizing the relevance of cardiovascular studies in Zebrafish for understanding the risks to human health. It also highlights novel experimental approaches and technologies while addressing their inherent challenges and limitations.
Collapse
Affiliation(s)
- Eman Hussen
- Biological Science Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Nada Aakel
- Biomedical Sciences Department, College of Health Sciences, Qatar University, Doha P.O. Box 2713, Qatar; (N.A.); (M.A.-A.); (H.A.-S.)
| | - Abdullah A. Shaito
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Maha Al-Asmakh
- Biomedical Sciences Department, College of Health Sciences, Qatar University, Doha P.O. Box 2713, Qatar; (N.A.); (M.A.-A.); (H.A.-S.)
| | - Haissam Abou-Saleh
- Biomedical Sciences Department, College of Health Sciences, Qatar University, Doha P.O. Box 2713, Qatar; (N.A.); (M.A.-A.); (H.A.-S.)
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Zain Z. Zakaria
- Medical and Health Sciences Office, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
12
|
Subramanian S. Zebrafish as a model organism - can a fish mimic human? J Basic Clin Physiol Pharmacol 2023; 34:559-575. [PMID: 34662932 DOI: 10.1515/jbcpp-2021-0113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/20/2021] [Indexed: 02/06/2023]
Abstract
From pre-historic era, all scientific discoveries have evolved around a concept - THINK BIG but for a change zebrafish as a model organism in research had managed to halt the entire medical community and made us realize that it's time to think small. From a barely imagined being in research few years ago to around 4,000 publications in just last year, zebrafish has definitely come a long way. Through these tiny fish, scientists have managed to find genes that caused human diseases and have also developed various specific models to know more about the pathology behind such diseases. This review will focus on zebrafish as a model organism from the time it was introduced to the most novel targets with particular emphasis on central nervous system (CNS) as it is rapidly evolving branch in zebrafish research these days. This review will try to shed light on the early stages of zebrafish as a model organism and will try to cover the journey of it developing as a successful model organism to map many diseases like diabetes, Alzheimer's and autism describing the rationale for using this specific model and briefly the techniques under each category and finally will summarize the pros and cons of the model with its expected future directions.
Collapse
|
13
|
Šimková A, Civáňová Křížová K, Voříšková K, Vetešník L, Bystrý V, Demko M. Transcriptome Profile Analyses of Head Kidney in Roach ( Rutilus rutilus), Common Bream ( Abramis brama) and Their Hybrids: Does Infection by Monogenean Parasites in Freshwater Fish Reveal Differences in Fish Vigour among Parental Species and Their Hybrids? BIOLOGY 2023; 12:1199. [PMID: 37759598 PMCID: PMC10525477 DOI: 10.3390/biology12091199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023]
Abstract
Hybrid generations usually face either a heterosis advantage or a breakdown, that can be expressed by the level of parasite infection in hybrid hosts. Hybrids are less infected by parasites than parental species (especially F1 generations) or more infected than parental species (especially post-F1 generations). We performed the experiment with blood-feeding gill parasite Paradiplozoon homoion (Monogenea) infecting leuciscid species, Abramis brama and Rutilus rutilus, their F1 generation and two backcross generations. Backcross generations tended to be more parasitized than parental lines and the F1 generation. The number of differentially expressed genes (DEGs) was lower in F1 hybrids and higher in backcross hybrids when compared to each of the parental lines. The main groups of DEGs were shared among lines; however, A. brama and R. rutilus differed in some of the top gene ontology (GO) terms. DEG analyses revealed the role of heme binding and erythrocyte differentiation after infection by blood-feeding P. homoion. Two backcross generations shared some of the top GO terms, representing mostly downregulated genes associated with P. homoion infection. KEGG analysis revealed the importance of disease-associated pathways; the majority of them were shared by two backcross generations. Our study revealed the most pronounced DEGs associated with blood-feeding monogeneans in backcross hybrids, potentially (but not exclusively) explainable by hybrid breakdown. The lower DEGs reported in F1 hybrids being less parasitized than backcross hybrids is in line with the hybrid advantage.
Collapse
Affiliation(s)
- Andrea Šimková
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic; (K.C.K.); (K.V.)
| | - Kristína Civáňová Křížová
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic; (K.C.K.); (K.V.)
| | - Kristýna Voříšková
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic; (K.C.K.); (K.V.)
| | - Lukáš Vetešník
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic; (K.C.K.); (K.V.)
- Institute of Vertebrate Biology of the Czech Academy of Sciences, Květná 8, 603 65 Brno, Czech Republic; (L.V.)
| | - Vojtěch Bystrý
- Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic; (V.B.); (M.D.)
| | - Martin Demko
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic; (K.C.K.); (K.V.)
- Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic; (V.B.); (M.D.)
| |
Collapse
|
14
|
Giardoglou P, Deloukas P, Dedoussis G, Beis D. Cfdp1 Is Essential for Cardiac Development and Function. Cells 2023; 12:1994. [PMID: 37566073 PMCID: PMC10417793 DOI: 10.3390/cells12151994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/25/2023] [Accepted: 08/01/2023] [Indexed: 08/12/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the prevalent cause of mortality worldwide. A combination of environmental and genetic effectors modulates the risk of developing them. Thus, it is vital to identify candidate genes and elucidate their role in the manifestation of the disease. Large-scale human studies have revealed the implication of Craniofacial Development Protein 1 (CFDP1) in Coronary Artery Disease (CAD). CFDP1 belongs to the evolutionary conserved Bucentaur (BCNT) family, and to date, its function and mechanism of action in Cardiovascular Development are still unclear. We utilized zebrafish to investigate the role of cfdp1 in the developing heart due to the high genomic homology, similarity in heart physiology, and ease of experimental manipulations. We showed that cfdp1 was expressed during development, and we tested two morpholinos and generated a cfdp1 mutant line. The cfdp1-/- embryos developed arrhythmic hearts and exhibited defective cardiac performance, which led to a lethal phenotype. Findings from both knockdown and knockout experiments showed that abrogation of cfdp1 leads to downregulation of Wnt signaling in embryonic hearts during valve development but without affecting Notch activation in this process. The cfdp1 zebrafish mutant line provides a valuable tool for unveiling the novel mechanism of regulating cardiac physiology and function. cfdp1 is essential for cardiac development, a previously unreported phenotype most likely due to early lethality in mice. The detected phenotype of bradycardia and arrhythmias is an observation with potential clinical relevance for humans carrying heterozygous CFDP1 mutations and their risk of developing CAD.
Collapse
Affiliation(s)
- Panagiota Giardoglou
- Zebrafish Disease Model Laboratory, Center for Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece;
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, 17676 Athens, Greece;
| | - Panos Deloukas
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London Medical School, Queen Mary University of London, London E1 4NS, UK;
| | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, 17676 Athens, Greece;
| | - Dimitris Beis
- Zebrafish Disease Model Laboratory, Center for Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece;
- Laboratory of Biological Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| |
Collapse
|
15
|
Zhang X, Almasian M, Hassan SS, Jotheesh R, Kadam VA, Polk AR, Saberigarakani A, Rahat A, Yuan J, Lee J, Carroll K, Ding Y. 4D Light-sheet imaging and interactive analysis of cardiac contractility in zebrafish larvae. APL Bioeng 2023; 7:026112. [PMID: 37351330 PMCID: PMC10283270 DOI: 10.1063/5.0153214] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/05/2023] [Indexed: 06/24/2023] Open
Abstract
Despite ongoing efforts in cardiovascular research, the acquisition of high-resolution and high-speed images for the purpose of assessing cardiac contraction remains challenging. Light-sheet fluorescence microscopy (LSFM) offers superior spatiotemporal resolution and minimal photodamage, providing an indispensable opportunity for the in vivo study of cardiac micro-structure and contractile function in zebrafish larvae. To track the myocardial architecture and contractility, we have developed an imaging strategy ranging from LSFM system construction, retrospective synchronization, single cell tracking, to user-directed virtual reality (VR) analysis. Our system enables the four-dimensional (4D) investigation of individual cardiomyocytes across the entire atrium and ventricle during multiple cardiac cycles in a zebrafish larva at the cellular resolution. To enhance the throughput of our model reconstruction and assessment, we have developed a parallel computing-assisted algorithm for 4D synchronization, resulting in a nearly tenfold enhancement of reconstruction efficiency. The machine learning-based nuclei segmentation and VR-based interaction further allow us to quantify cellular dynamics in the myocardium from end-systole to end-diastole. Collectively, our strategy facilitates noninvasive cardiac imaging and user-directed data interpretation with improved efficiency and accuracy, holding great promise to characterize functional changes and regional mechanics at the single cell level during cardiac development and regeneration.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Milad Almasian
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Sohail S. Hassan
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Rosemary Jotheesh
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Vinay A. Kadam
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Austin R. Polk
- Department of Computer Science, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Alireza Saberigarakani
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Aayan Rahat
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Jie Yuan
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Juhyun Lee
- Department of Bioengineering, The University of Texas at Arlington, Arlington, Texas 76019, USA
| | - Kelli Carroll
- Department of Biology, Austin College, Sherman, Texas 75090, USA
| | - Yichen Ding
- Author to whom correspondence should be addressed:. Tel.: 972–883-7217
| |
Collapse
|
16
|
Wang L, Mou L, Guan S, Wang C, Sik A, Stoika R, Liu K, Jin M. Isoliquiritigenin induces neurodevelopmental-toxicity and anxiety-like behavior in zebrafish larvae. Comp Biochem Physiol C Toxicol Pharmacol 2023; 266:109555. [PMID: 36717046 DOI: 10.1016/j.cbpc.2023.109555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/14/2023] [Accepted: 01/22/2023] [Indexed: 01/28/2023]
Abstract
Isoliquiritigenin, a flavonoid compound, exhibits a variety of pharmacological properties, including anti-inflammatory, anti-oxidative, anti-microbial, anti-viral, and anti-tumor effects. In the past few years, the consumption of isoliquiritigenin-containing dietary supplements has increased due to their health benefits. Although the neuroprotective effects of isoliquiritigenin have been well-investigated, these studies were performed in cells and adult animals. The potential effects of isoliquiritigenin on the development, especially the neurodevelopment, of certain populations, such as zebrafish larvae, have not been investigated. In this study, zebrafish larvae were employed as a model to investigate the effects of isoliquiritigenin on development and neurodevelopment. Zebrafish embryos treated with high concentrations of isoliquiritigenin (10 and 15 μM) exhibited high rates of mortality, hatching, and malformation, indicating that isoliquiritigenin can affect zebrafish development. In addition, isoliquiritigenin impeded the development of central nervous system regions and the length of dopaminergic neurons located in midbrains and thalami of transgenic zebrafish larvae. The locomotor ability of zebrafish larvae exposed to high concentrations of isoliquiritigenin was negatively affected. The total distance and the average velocity significantly decreased, and anxiety-related behaviors were observed under light-dark challenge. Furthermore, the levels of gap43, tuba1b, mbp, hcrt, vmat2, and pomc, which mediate neurodevelopment, neurotoxicity, and anxiety were significantly decreased in zebrafish larvae exposed to isoliquiritigenin. These results indicate that isoliquiritigenin can disrupt the development of dopaminergic neurons and the function of the central nervous system in zebrafish, causing anxiety-like symptoms.
Collapse
Affiliation(s)
- Lizhen Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China
| | - Lei Mou
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China
| | - Shibing Guan
- Department of Hand and Foot Surgery, Provincial Hospital Affiliated to Shandong First Medical University, 9677 Jingshi Road, Ji'nan 250098, Shandong Province, People's Republic of China
| | - Chuansen Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China
| | - Attila Sik
- Institute of Transdisciplinary Discoveries, Medical School, University of Pecs, Pecs H-7624, Hungary; Institute of Clinical Sciences, Medical School, University of Birmingham, Birmingham B15 2TT, United Kingdom; Institute of Physiology, Medical School, University of Pecs, Pecs H-7624, Hungary
| | - Rostyslav Stoika
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology, National Academy of Sciences of Ukraine, Lviv, Ukraine
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China
| | - Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China.
| |
Collapse
|
17
|
Novel amphiphilic hydroxyethyl starch-based nanoparticles loading camptothecin exhibit high anticancer activity in HepG2 cells and zebrafish. Colloids Surf B Biointerfaces 2023; 224:113215. [PMID: 36841205 DOI: 10.1016/j.colsurfb.2023.113215] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/01/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023]
Abstract
Camptothecin is a naturally occurred anticancer drug but exhibits limitations including poor aqueous solubility, low bioavailability, and high level of adverse drug reactions on normal organs. To overcome these problems, this paper developed a novel amphiphilic Lau-Leu-HES carrier using hydroxyethyl starch, lauric acid, and L-leucine as starting materials. The carrier was successfully applied to prepare Lau-Leu-HES nanoparticles loading camptothecin. The drug loading efficiency and encapsulation efficiency of the nanoparticles were calculated to be 29.04% and 81.85%, respectively. The nanoparticles exhibited high zeta potential (-15.51 mV) and small hydrodynamic diameter (105.4 nm). Camptothecin in nanoparticles could be rapidly released under acidic condition (pH = 4.5), thereby indicating the high sensitivity under cancer microenvironments. Anticancer investigation revealed that the nanoparticles could inhibit the proliferation of HepG2 cells in vitro. Compared with commercial available drug doxorubicin, the nanoparticles could significantly inhibit the expression of krasv12 oncogene in transgenic Tg (EGFP-krasV12) zebrafish. These results indicate that the camptothecin-loaded Lau-Leu-HES nanoparticles are expected to be a potential candidate for cancer therapy.
Collapse
|
18
|
Teranikar T, Nguyen P, Lee J. Biomechanics of cardiac development in zebrafish model. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2023. [DOI: 10.1016/j.cobme.2023.100459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
19
|
Adhish M, Manjubala I. Effectiveness of zebrafish models in understanding human diseases-A review of models. Heliyon 2023; 9:e14557. [PMID: 36950605 PMCID: PMC10025926 DOI: 10.1016/j.heliyon.2023.e14557] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 03/01/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Understanding the detailed mechanism behind every human disease, disorder, defect, and deficiency is a daunting task concerning the clinical diagnostic tools for patients. Hence, a closely resembling living or simulated model is of paramount interest for the development and testing of a probable novel drug for rectifying the conditions pertaining to the various ailments. The animal model that can be easily genetically manipulated to suit the study of the therapeutic motive is an indispensable asset and within the last few decades, the zebrafish models have proven their effectiveness by becoming such potent human disease models with their use being extended to various avenues of research to understand the underlying mechanisms of the diseases. As zebrafish are explored as model animals in understanding the molecular basis and genetics of many diseases owing to the 70% genetic homology between the human and zebrafish genes; new and fascinating facts about the diseases are being surfaced, establishing it as a very powerful tool for upcoming research. These prospective research areas can be explored in the near future using zebrafish as a model. In this review, the effectiveness of the zebrafish as an animal model against several human diseases such as osteoporosis, atrial fibrillation, Noonan syndrome, leukemia, autism spectrum disorders, etc. has been discussed.
Collapse
Affiliation(s)
- Mazumder Adhish
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, 632 014, India
| | - I. Manjubala
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, 632 014, India
| |
Collapse
|
20
|
Başımoğlu Koca Y, Koca S, Öztel Z, Balcan E. Determination of histopathological effects and myoglobin, periostin gene-protein expression levels in Danio rerio muscle tissue after acaricide yoksorrun-5EC (hexythiazox) application. Drug Chem Toxicol 2023; 46:50-58. [PMID: 34879781 DOI: 10.1080/01480545.2021.2007945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Although pesticides are essential agrochemicals to annihilate harmful organisms in agriculture, their uncontrolled use has become an important threat to environmental health. Exposure to pesticides can affect many biological systems including immune system, endocrine system, and nervous system. However, the potential side effects of pesticides to skeletal muscle system remain unclear. Present study has focused on the evaluation of this issue by using an acaricide, yoksorrun-5EC (hexythiazox), in an aquatic model organism, Danio rerio. The histological analyses revealed that increased concentrations of the acaricide cause degradation of skeletal muscle along with increased necrosis and atrophy in myocytes, intercellular edema, and increased infiltrations between perimysium sheaths of muscle fibers. The effects of acaricide on myoglobin and periostin, which are associated with oxygen transport and muscle regeneration, respectively, were investigated at the gene and protein levels. RT-PCR results suggested that high concentration yoksorrun-5EC (hexythiazox) can induce myoglobin and periostin genes. Similar results were also obtained in the protein levels of these genes by western blotting analysis. These results suggested that yoksorrun-5EC (hexythiazox)-dependent disruption of skeletal muscle architecture is closely associated with the expression levels of myoglobin and periostin genes in Danio rerio model.
Collapse
Affiliation(s)
- Yücel Başımoğlu Koca
- Department of Biology, Zoology Section, Faculty of Science and Art, Adnan Menderes University, Aydin, Turkey
| | - Serdar Koca
- Department of Biology, General Biology Section, Faculty of Science and Art, Adnan Menderes University, Aydin, Turkey
| | - Zübeyde Öztel
- Department of Biology, Molecular Biology Section, Faculty of Science and Art, Manisa Celal Bayar University, Manisa, Turkey
| | - Erdal Balcan
- Department of Biology, Molecular Biology Section, Faculty of Science and Art, Manisa Celal Bayar University, Manisa, Turkey
| |
Collapse
|
21
|
Production, characterization, microbial inhibition, and in vivo toxicity of cold atmospheric plasma activated water. INNOV FOOD SCI EMERG 2023. [DOI: 10.1016/j.ifset.2022.103265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
22
|
Kwiatkowska I, Hermanowicz JM, Iwinska Z, Kowalczuk K, Iwanowska J, Pawlak D. Zebrafish—An Optimal Model in Experimental Oncology. Molecules 2022; 27:molecules27134223. [PMID: 35807468 PMCID: PMC9268704 DOI: 10.3390/molecules27134223] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/10/2022] [Accepted: 06/28/2022] [Indexed: 02/02/2023] Open
Abstract
A thorough understanding of cancer pathogenesis is a necessary step in the development of more effective and safer therapy. However, due to the complexity of the process and intricate interactions, studying tumor development is an extremely difficult and challenging task. In bringing this issue closer, different scientific models with various advancement levels are helpful. Cell cultures is a system that is too simple and does not allow for multidirectional research. On the other hand, rodent models, although commonly used, are burdened with several limitations. For this reason, new model organisms that will allow for the studying of carcinogenesis stages and factors reliably involved in them are urgently sought after. Danio rerio, an inconspicuous fish endowed with unique features, is gaining in importance in the world of scientific research. Including it in oncological research brings solutions to many challenges afflicting modern medicine. This article aims to illustrate the usefulness of Danio rerio as a model organism which turns out to be a powerful and unique tool for studying the stages of carcinogenesis and solving the hitherto incomprehensible processes that lead to the development of the disease.
Collapse
Affiliation(s)
- Iwona Kwiatkowska
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (J.M.H.); (Z.I.); (J.I.); (D.P.)
- Correspondence: ; Tel./Fax: +48-8574-856-01
| | - Justyna Magdalena Hermanowicz
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (J.M.H.); (Z.I.); (J.I.); (D.P.)
- Department of Clinical Pharmacy, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland
| | - Zaneta Iwinska
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (J.M.H.); (Z.I.); (J.I.); (D.P.)
| | - Krystyna Kowalczuk
- Department of Integrated Medical Care, Medical University of Bialystok, ul. M Skłodowskiej-Curie 7A, 15-096 Bialystok, Poland;
| | - Jolanta Iwanowska
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (J.M.H.); (Z.I.); (J.I.); (D.P.)
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (J.M.H.); (Z.I.); (J.I.); (D.P.)
| |
Collapse
|
23
|
Maciag M, Wnorowski A, Bednarz K, Plazinska A. Evaluation of β-adrenergic ligands for development of pharmacological heart failure and transparency models in zebrafish. Toxicol Appl Pharmacol 2022; 434:115812. [PMID: 34838787 DOI: 10.1016/j.taap.2021.115812] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/25/2021] [Accepted: 11/22/2021] [Indexed: 10/19/2022]
Abstract
Cardiovascular toxicity represents one of the most common reasons for clinical trial failure. Consequently, early identification of novel cardioprotective strategies could prevent the later-stage drug-induced cardiac side effects. The use of zebrafish (Danio rerio) in preclinical studies has greatly increased. High-throughput and low-cost of assays make zebrafish model ideal for initial drug discovery. A common strategy to induce heart failure is a chronic β-adrenergic (βAR) stimulation. Herein, we set out to test a panel of βAR agonists to develop a pharmacological heart failure model in zebrafish. We assessed βAR agonists with respect to the elicited mortality, changes in heart rate, and morphological alterations in zebrafish larvae according to Fish Embryo Acute Toxicity Test. Among the tested βAR agonists, epinephrine elicited the most potent onset of heart stimulation (EC50 = 0.05 mM), which corresponds with its physiological role as catecholamine. However, when used at ten-fold higher dose (0.5 mM), the same compound caused severe heart rate inhibition (-28.70 beats/min), which can be attributed to its cardiotoxicity. Further studies revealed that isoetharine abolished body pigmentation at the sublethal dose of 7.50 mM. Additionally, as a proof of concept that zebrafish can mimic human cardiac physiology, we tested βAR antagonists (propranolol, carvedilol, metoprolol, and labetalol) and verified that they inhibited fish heart rate in a similar fashion as in humans. In conclusion, we proposed two novel pharmacological models in zebrafish; i.e., epinephrine-dependent heart failure and isoetharine-dependent transparent zebrafish. We provided strong evidence that the zebrafish model constitutes a valuable tool for cardiovascular research.
Collapse
Affiliation(s)
- Monika Maciag
- Department of Biopharmacy, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland; Independent Laboratory of Behavioral Studies, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland.
| | - Artur Wnorowski
- Department of Biopharmacy, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland.
| | - Kinga Bednarz
- Department of Biopharmacy, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Anita Plazinska
- Department of Biopharmacy, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland.
| |
Collapse
|
24
|
Leonidis G, Dalezis P, Trafalis D, Beis D, Giardoglou P, Koukiali A, Sigala I, Nikolakaki E, Sarli V. Synthesis and Biological Evaluation of a c(RGDyK) Peptide Conjugate of SRPIN803. ACS OMEGA 2021; 6:28379-28393. [PMID: 34723035 PMCID: PMC8552469 DOI: 10.1021/acsomega.1c04576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/04/2021] [Indexed: 06/13/2023]
Abstract
In the present study, SRPIN803 and c(RGDyK)-SRPIN803 hybrid compounds were efficiently synthesized and evaluated for their stability in human plasma and buffers of pH 7.4 and 5.2. The hybrids were mainly cytostatic against a panel of tested cancer cells, whereas one c(RGDyK)-SRPIN803 hybrid, geo35, was the most active compound in this screen and was cytotoxic against cell lines MCF7 and MRC5 with IC50 values of 61 and 63 μM, respectively. SRPIN803 and geo35 exhibited antiangiogenic activity in zebrafish embryos, and this effect was dose-dependent. Although c(RGDyK)-SRPIN803 hybrid compounds were found less potent compared to SRPIN803, they have shown activities interesting enough to illustrate the potential of this approach for the development of a new class of antiangiogenic compounds.
Collapse
Affiliation(s)
- George Leonidis
- Department
of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Panagiotis Dalezis
- Laboratory
of Pharmacology, Medical School National
and Kapodistrian University of Athens, 75 Mikras Asias Street, Athens 11527, Greece
| | - Dimitrios Trafalis
- Laboratory
of Pharmacology, Medical School National
and Kapodistrian University of Athens, 75 Mikras Asias Street, Athens 11527, Greece
| | - Dimitris Beis
- Zebrafish
Disease Model Lab, Biomedical Research Foundation
Academy of Athens, Athens 115 27, Greece
| | - Panagiota Giardoglou
- Zebrafish
Disease Model Lab, Biomedical Research Foundation
Academy of Athens, Athens 115 27, Greece
| | - Anastasia Koukiali
- Department
of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Ioanna Sigala
- Department
of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Eleni Nikolakaki
- Department
of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Vasiliki Sarli
- Department
of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| |
Collapse
|
25
|
Munekata PES, Pateiro M, Conte-Junior CA, Domínguez R, Nawaz A, Walayat N, Movilla Fierro E, Lorenzo JM. Marine Alkaloids: Compounds with In Vivo Activity and Chemical Synthesis. Mar Drugs 2021; 19:374. [PMID: 34203532 PMCID: PMC8306672 DOI: 10.3390/md19070374] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/24/2021] [Accepted: 06/24/2021] [Indexed: 12/15/2022] Open
Abstract
Marine alkaloids comprise a class of compounds with several nitrogenated structures that can be explored as potential natural bioactive compounds. The scientific interest in these compounds has been increasing in the last decades, and many studies have been published elucidating their chemical structure and biological effects in vitro. Following this trend, the number of in vivo studies reporting the health-related properties of marine alkaloids has been increasing and providing more information about the effects in complex organisms. Experiments with animals, especially mice and zebrafish, are revealing the potential health benefits against cancer development, cardiovascular diseases, seizures, Alzheimer's disease, mental health disorders, inflammatory diseases, osteoporosis, cystic fibrosis, oxidative stress, human parasites, and microbial infections in vivo. Although major efforts are still necessary to increase the knowledge, especially about the translation value of the information obtained from in vivo experiments to clinical trials, marine alkaloids are promising candidates for further experiments in drug development.
Collapse
Affiliation(s)
- Paulo E. S. Munekata
- Centro Tecnológico de la Carne de Galicia, Parque Tecnológico de Galicia, rúa Galicia No. 4, San Cibrao das Viñas, 32900 Ourense, Spain; (P.E.S.M.); (M.P.); (R.D.)
| | - Mirian Pateiro
- Centro Tecnológico de la Carne de Galicia, Parque Tecnológico de Galicia, rúa Galicia No. 4, San Cibrao das Viñas, 32900 Ourense, Spain; (P.E.S.M.); (M.P.); (R.D.)
| | - Carlos A. Conte-Junior
- Centro de Tecnologia, Programa de Pós-Graduação em Ciência de Alimentos, Instituto de Química, Universidade Federal do Rio de Janeiro, Avenida Athos da Silveira Ramos 149, Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil;
| | - Rubén Domínguez
- Centro Tecnológico de la Carne de Galicia, Parque Tecnológico de Galicia, rúa Galicia No. 4, San Cibrao das Viñas, 32900 Ourense, Spain; (P.E.S.M.); (M.P.); (R.D.)
| | - Asad Nawaz
- Jiangsu Key Laboratory of Crop Genetics and Physiology, College of Agriculture, Yangzhou University, Yangzhou 225009, China;
| | - Noman Walayat
- Department of Food Science and Engineering, College of Ocean, Zhejiang University of Technology, Hangzhou 310014, China;
| | | | - José M. Lorenzo
- Centro Tecnológico de la Carne de Galicia, Parque Tecnológico de Galicia, rúa Galicia No. 4, San Cibrao das Viñas, 32900 Ourense, Spain; (P.E.S.M.); (M.P.); (R.D.)
- Área de Tecnología de los Alimentos, Facultad de Ciencias de Ourense, Universidad de Vigo, 32004 Ourense, Spain
| |
Collapse
|
26
|
Saputra F, Lai YH, Fernandez RAT, Macabeo APG, Lai HT, Huang JC, Hsiao CD. Acute and Sub-Chronic Exposure to Artificial Sweeteners at the Highest Environmentally Relevant Concentration Induce Less Cardiovascular Physiology Alterations in Zebrafish Larvae. BIOLOGY 2021; 10:548. [PMID: 34207293 PMCID: PMC8233861 DOI: 10.3390/biology10060548] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 12/16/2022]
Abstract
Artificial sweeteners are widely used food ingredients in beverages and drinks to lower calorie intake which in turn helps prevent lifestyle diseases such as obesity. However, as their popularity has increased, the release of artificial sweetener to the aquatic environment has also increased at a tremendous rate. Thus, our study aims to systematically explore the potential cardiovascular physiology alterations caused by eight commercial artificial sweeteners, including acesulfame-K, alitame, aspartame, sodium cyclamate, dulcin, neotame, saccharine and sucralose, at the highest environmentally relevant concentration on cardiovascular performance using zebrafish (Danio rerio) as a model system. Embryonic zebrafish were exposed to the eight artificial sweeteners at 100 ppb and their cardiovascular performance (heart rate, ejection fraction, fractional shortening, stroke volume, cardiac output, heartbeat variability, and blood flow velocity) was measured and compared. Overall, our finding supports the safety of artificial sweetener exposure. However, several finding like a significant increase in the heart rate and heart rate variability after incubation in several artificial sweeteners are noteworthy. Biomarker testing also revealed that saccharine significantly increase the dopamine level in zebrafish larvae, which is might be the reason for the cardiac physiology changes observed after saccharine exposure.
Collapse
Affiliation(s)
- Ferry Saputra
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320314, Taiwan;
| | - Yu-Heng Lai
- Department of Chemistry, Chinese Culture University, Taipei 11114, Taiwan;
| | - Rey Arturo T. Fernandez
- Laboratory for Organic Reactivity, Discovery and Synthesis (LORDS), Research Center for the Natural and Applied Sciences, University of Santo Tomas, Espana St., Manila 1015, Philippines; (R.A.T.F.); (A.P.G.M.)
| | - Allan Patrick G. Macabeo
- Laboratory for Organic Reactivity, Discovery and Synthesis (LORDS), Research Center for the Natural and Applied Sciences, University of Santo Tomas, Espana St., Manila 1015, Philippines; (R.A.T.F.); (A.P.G.M.)
| | - Hong-Thih Lai
- Department of Aquatic Biosciences, National Chiayi University, Chiayi 600355, Taiwan
| | - Jong-Chin Huang
- Department of Applied Chemistry, National Pingtung University, Pingtung 90003, Taiwan
| | - Chung-Der Hsiao
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320314, Taiwan;
- Center for Nanotechnology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
- Research Center for Aquatic Toxicology and Pharmacology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
| |
Collapse
|
27
|
Lin FJ, Li H, Wu DT, Zhuang QG, Li HB, Geng F, Gan RY. Recent development in zebrafish model for bioactivity and safety evaluation of natural products. Crit Rev Food Sci Nutr 2021; 62:8646-8674. [PMID: 34058920 DOI: 10.1080/10408398.2021.1931023] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The zebrafish is a species of freshwater fish, popular in aquariums and laboratories. Several advantageous features have facilitated zebrafish to be extensively utilized as a valuable vertebrate model in the lab. It has been well-recognized that natural products possess multiple health benefits for humans. With the increasing demand for natural products in the development of functional foods, nutraceuticals, and natural cosmetics, the zebrafish has emerged as an unprecedented tool for rapidly and economically screening and identifying safe and effective substances from natural products. This review first summarized the key factors for the management of zebrafish in the laboratory, followed by highlighting the current progress on the establishment and applications of zebrafish models in the bioactivity evaluation of natural products. In addition, the zebrafish models used for assessing the potential toxicity or health risks of natural products were involved as well. Overall, this review indicates that zebrafish are promising animal models for the bioactivity and safety evaluation of natural products, and zebrafish models can accelerate the discovery of novel natural products with potential health functions.
Collapse
Affiliation(s)
- Fang-Jun Lin
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China.,Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - Hang Li
- Research Center for Plants and Human Health, Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
| | - Ding-Tao Wu
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China
| | - Qi-Guo Zhuang
- China-New Zealand Belt and Road Joint Laboratory on Kiwifruit, Sichuan Provincial Academy of Natural Resource Sciences, Chengdu, China
| | - Hua-Bin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Fang Geng
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China
| | - Ren-You Gan
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China.,Research Center for Plants and Human Health, Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
| |
Collapse
|
28
|
Marrs JA, Sarmah S. The Genius of the Zebrafish Model: Insights on Development and Disease. Biomedicines 2021; 9:biomedicines9050577. [PMID: 34065228 PMCID: PMC8160874 DOI: 10.3390/biomedicines9050577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 05/06/2021] [Indexed: 01/20/2023] Open
|
29
|
Giardoglou P, Bournele D, Park M, Kanoni S, Dedoussis GV, Steinberg SF, Deloukas P, Beis D. A zebrafish forward genetic screen identifies an indispensable threonine residue in the kinase domain of PRKD2. Biol Open 2021; 10:bio.058542. [PMID: 33597201 PMCID: PMC7969590 DOI: 10.1242/bio.058542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Protein kinase D2 belongs to a family of evolutionarily conserved enzymes regulating several biological processes. In a forward genetic screen for zebrafish cardiovascular mutants, we identified a mutation in the prkd2 gene. Homozygous mutant embryos develop as wild type up to 36 h post-fertilization and initiate blood flow, but fail to maintain it, resulting in a complete outflow tract stenosis. We identified a mutation in the prkd2 gene that results in a T757A substitution at a conserved residue in the kinase domain activation loop (T714A in human PRKD2) that disrupts catalytic activity and drives this phenotype. Homozygous mutants survive without circulation for several days, allowing us to study the extreme phenotype of no intracardiac flow, in the background of a functional heart. We show dysregulation of atrioventricular and outflow tract markers in the mutants and higher sensitivity to the Calcineurin inhibitor, Cyclosporin A. Finally we identify TBX5 as a potential regulator of PRKD2. Our results implicate PRKD2 catalytic activity in outflow tract development in zebrafish. This article has an associated First Person interview with the first author of the paper. Summary: We identified, through a zebrafish forward screen, an evolutionarily conserved residue in the catalytic domain of protein kinase D2 and its homologues.
Collapse
Affiliation(s)
- Panagiota Giardoglou
- Zebrafish Disease Model lab, Biomedical Research Foundation Academy of Athens, Athens 115 27, Greece.,Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, Athens 176 71, Greece
| | - Despina Bournele
- Zebrafish Disease Model lab, Biomedical Research Foundation Academy of Athens, Athens 115 27, Greece
| | - Misun Park
- Department of Pharmacology, Columbia University, New York 100 27, USA
| | - Stavroula Kanoni
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Clinical Pharmacology Centre, Queen Mary University of London, London, EC1M 6BQ, UK
| | - George V Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, Athens 176 71, Greece
| | - Susan F Steinberg
- Department of Pharmacology, Columbia University, New York 100 27, USA
| | - Panos Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Clinical Pharmacology Centre, Queen Mary University of London, London, EC1M 6BQ, UK.,Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah 222 52, Saudi Arabia
| | - Dimitris Beis
- Zebrafish Disease Model lab, Biomedical Research Foundation Academy of Athens, Athens 115 27, Greece
| |
Collapse
|
30
|
Moreau A, Reisqs J, Delanoe‐Ayari H, Pierre M, Janin A, Deliniere A, Bessière F, Meli AC, Charrabi A, Lafont E, Valla C, Bauer D, Morel E, Gache V, Millat G, Nissan X, Faucherre A, Jopling C, Richard S, Mejat A, Chevalier P. Deciphering DSC2 arrhythmogenic cardiomyopathy electrical instability: From ion channels to ECG and tailored drug therapy. Clin Transl Med 2021; 11:e319. [PMID: 33784018 PMCID: PMC7908047 DOI: 10.1002/ctm2.319] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Severe ventricular rhythm disturbances are the hallmark of arrhythmogenic cardiomyopathy (ACM), and are often explained by structural conduction abnormalities. However, comprehensive investigations of ACM cell electrical instability are lacking. This study aimed to elucidate early electrical myogenic signature of ACM. METHODS We investigated a 41-year-old ACM patient with a missense mutation (c.394C>T) in the DSC2 gene, which encodes desmocollin 2. Pathogenicity of this variant was confirmed using a zebrafish DSC2 model system. Control and DSC2 patient-derived pluripotent stem cells were reprogrammed and differentiated into cardiomyocytes (hiPSC-CM) to examine the specific electromechanical phenotype and its modulation by antiarrhythmic drugs (AADs). Samples of the patient's heart and hiPSC-CM were examined to identify molecular and cellular alterations. RESULTS A shortened action potential duration was associated with reduced Ca2+ current density and increased K+ current density. This finding led to the elucidation of previously unknown abnormal repolarization dynamics in ACM patients. Moreover, the Ca2+ mobilised during transients was decreased, and the Ca2+ sparks frequency was increased. AAD testing revealed the following: (1) flecainide normalised Ca2+ transients and significantly decreased Ca2+ spark occurrence and (2) sotalol significantly lengthened the action potential and normalised the cells' contractile properties. CONCLUSIONS Thorough analysis of hiPSC-CM derived from the DSC2 patient revealed abnormal repolarization dynamics, prompting the discovery of a short QT interval in some ACM patients. Overall, these results confirm a myogenic origin of ACM electrical instability and provide a rationale for prescribing class 1 and 3 AADs in ACM patients with increased ventricular repolarization reserve.
Collapse
Affiliation(s)
- Adrien Moreau
- PhyMedExpINSERM U1046CNRS UMR9214Université de MontpellierMontpellierFrance
| | - Jean‐Baptiste Reisqs
- PhyMedExpINSERM U1046CNRS UMR9214Université de MontpellierMontpellierFrance
- Neuromyogene InstitutClaude Bernard University, Lyon 1VilleurbanneFrance
| | | | - Marion Pierre
- PhyMedExpINSERM U1046CNRS UMR9214Université de MontpellierMontpellierFrance
| | - Alexandre Janin
- Neuromyogene InstitutClaude Bernard University, Lyon 1VilleurbanneFrance
- Service de RythmologieHospices Civils de LyonLyonFrance
- Laboratoire de Cardiogénétique moléculaireCentre de biologie et pathologie EstBronFrance
| | | | | | - Albano C. Meli
- PhyMedExpINSERM U1046CNRS UMR9214Université de MontpellierMontpellierFrance
| | - Azzouz Charrabi
- PhyMedExpINSERM U1046CNRS UMR9214Université de MontpellierMontpellierFrance
| | - Estele Lafont
- Neuromyogene InstitutClaude Bernard University, Lyon 1VilleurbanneFrance
| | - Camille Valla
- Neuromyogene InstitutClaude Bernard University, Lyon 1VilleurbanneFrance
| | - Delphine Bauer
- Neuromyogene InstitutClaude Bernard University, Lyon 1VilleurbanneFrance
| | - Elodie Morel
- Neuromyogene InstitutClaude Bernard University, Lyon 1VilleurbanneFrance
| | - Vincent Gache
- Neuromyogene InstitutClaude Bernard University, Lyon 1VilleurbanneFrance
| | - Gilles Millat
- Neuromyogene InstitutClaude Bernard University, Lyon 1VilleurbanneFrance
- Service de RythmologieHospices Civils de LyonLyonFrance
- Laboratoire de Cardiogénétique moléculaireCentre de biologie et pathologie EstBronFrance
| | | | | | - Chris Jopling
- IGF, CNRS, INSERMUniversité de MontpellierMontpellierFrance
| | - Sylvain Richard
- PhyMedExpINSERM U1046CNRS UMR9214Université de MontpellierMontpellierFrance
| | - Alexandre Mejat
- Neuromyogene InstitutClaude Bernard University, Lyon 1VilleurbanneFrance
| | - Philippe Chevalier
- Neuromyogene InstitutClaude Bernard University, Lyon 1VilleurbanneFrance
- Service de RythmologieHospices Civils de LyonLyonFrance
| |
Collapse
|
31
|
Martin KE, Waxman JS. Atrial and Sinoatrial Node Development in the Zebrafish Heart. J Cardiovasc Dev Dis 2021; 8:jcdd8020015. [PMID: 33572147 PMCID: PMC7914448 DOI: 10.3390/jcdd8020015] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/31/2021] [Accepted: 02/04/2021] [Indexed: 12/11/2022] Open
Abstract
Proper development and function of the vertebrate heart is vital for embryonic and postnatal life. Many congenital heart defects in humans are associated with disruption of genes that direct the formation or maintenance of atrial and pacemaker cardiomyocytes at the venous pole of the heart. Zebrafish are an outstanding model for studying vertebrate cardiogenesis, due to the conservation of molecular mechanisms underlying early heart development, external development, and ease of genetic manipulation. Here, we discuss early developmental mechanisms that instruct appropriate formation of the venous pole in zebrafish embryos. We primarily focus on signals that determine atrial chamber size and the specialized pacemaker cells of the sinoatrial node through directing proper specification and differentiation, as well as contemporary insights into the plasticity and maintenance of cardiomyocyte identity in embryonic zebrafish hearts. Finally, we integrate how these insights into zebrafish cardiogenesis can serve as models for human atrial defects and arrhythmias.
Collapse
Affiliation(s)
- Kendall E. Martin
- Molecular Genetics, Biochemistry, and Microbiology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Joshua S. Waxman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Correspondence:
| |
Collapse
|
32
|
Lu YA, Jiang Y, Yang HW, Hwang J, Jeon YJ, Ryu B. Diphlorethohydroxycarmalol Isolated from Ishige okamurae Exerts Vasodilatory Effects via Calcium Signaling and PI3K/Akt/eNOS Pathway. Int J Mol Sci 2021; 22:1610. [PMID: 33562632 PMCID: PMC7914902 DOI: 10.3390/ijms22041610] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/11/2022] Open
Abstract
Nitric oxide (NO) is released by endothelial cells in the blood vessel wall to enhance vasodilation. Marine polyphenols are known to have protective effects against vascular dysfunction and hypertension. The present study is the first to investigate how diphlorethohydroxycarmalol (DPHC) isolated from Ishige okamurae affects calcium levels, resulting in enhanced vasodilation. We examined calcium modulation with the well-known receptors, acetylcholine receptor (AchR) and vascular endothelial growth factor 2 (VEGFR2), which are related to NO formation, and further confirmed the vasodilatory effect of DPHC. We confirmed that DPHC stimulated NO production by increasing calcium levels and endothelial nitric oxide synthase (eNOS) expression. DPHC affected AchR and VEGFR2 expression, thereby influencing transient calcium intake. Specific antagonists, atropine and SU5416, were used to verify our findings. Furthermore, based on the results of in vivo experiments, we treated Tg(flk:EGFP) transgenic zebrafish with DPHC to confirm its vasodilatory effect. In conclusion, the present study showed that DPHC modulated calcium transit through AchR and VEGFR2, increasing endothelial-dependent NO production. Thus, DPHC, a natural marine component, can efficiently ameliorate cardiovascular diseases by improving vascular function.
Collapse
Affiliation(s)
- Yu An Lu
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (Y.A.L.); (Y.J.); (H.-W.Y.); (J.H.)
| | - Yunfei Jiang
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (Y.A.L.); (Y.J.); (H.-W.Y.); (J.H.)
| | - Hye-Won Yang
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (Y.A.L.); (Y.J.); (H.-W.Y.); (J.H.)
| | - Jin Hwang
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (Y.A.L.); (Y.J.); (H.-W.Y.); (J.H.)
| | - You-Jin Jeon
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (Y.A.L.); (Y.J.); (H.-W.Y.); (J.H.)
- Marine Science Institute, Jeju National University, Jeju 63333, Korea
| | - Bomi Ryu
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (Y.A.L.); (Y.J.); (H.-W.Y.); (J.H.)
| |
Collapse
|
33
|
Abstract
Inherited cardiac arrhythmias contribute substantially to sudden cardiac death in the young. The underlying pathophysiology remains incompletely understood because of the lack of representative study models and the labour-intensive nature of electrophysiological patch clamp experiments. Whereas patch clamp is still considered the gold standard for investigating electrical properties in a cell, optical mapping of voltage and calcium transients has paved the way for high-throughput studies. Moreover, the development of human-induced pluripotent stem-cell-derived cardiomyocytes (hiPSC-CMs) has enabled the study of patient specific cell lines capturing the full genomic background. Nevertheless, hiPSC-CMs do not fully address the complex interactions between various cell types in the heart. Studies using in vivo models, are therefore necessary. Given the analogies between the human and zebrafish cardiovascular system, zebrafish has emerged as a cost-efficient model for arrhythmogenic diseases. In this review, we describe how hiPSC-CM and zebrafish are employed as models to study primary electrical disorders. We provide an overview of the contemporary electrophysiological phenotyping tools and discuss in more depth the different strategies available for optical mapping. We consider the current advantages and disadvantages of both hiPSC-CM and zebrafish as a model and optical mapping as phenotyping tool and propose strategies for further improvement. Overall, the combination of experimental readouts at cellular (hiPSC-CM) and whole organ (zebrafish) level can raise our understanding of the complexity of inherited cardiac arrhythmia disorders to the next level.
Collapse
|
34
|
Crocins from Crocus sativus L. in the Management of Hyperglycemia. In Vivo Evidence from Zebrafish. Molecules 2020; 25:molecules25225223. [PMID: 33182581 PMCID: PMC7696463 DOI: 10.3390/molecules25225223] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/03/2020] [Accepted: 11/07/2020] [Indexed: 12/11/2022] Open
Abstract
Diabetes mellitus is a disease characterized by persistent high blood glucose levels and accompanied by impaired metabolic pathways. In this study, we used zebrafish to investigate the effect of crocins isolated from Crocus sativus L., on the control of glucose levels and pancreatic β-cells. Embryos were exposed to an aqueous solution of crocins and whole embryo glucose levels were measured at 48 h post-treatment. We showed that the application of crocins reduces zebrafish embryo glucose levels and enhances insulin expression. We also examined whether crocins are implicated in the metabolic pathway of gluconeogenesis. We showed that following a single application of crocins and glucose level reduction, the expression of phosphoenolpyruvate carboxykinase1 (pck1), a key gene involved in glucose metabolism, is increased. We propose a putative role for the crocins in glucose metabolism and insulin management.
Collapse
|
35
|
Ryan R, Moyse BR, Richardson RJ. Zebrafish cardiac regeneration-looking beyond cardiomyocytes to a complex microenvironment. Histochem Cell Biol 2020; 154:533-548. [PMID: 32926230 PMCID: PMC7609419 DOI: 10.1007/s00418-020-01913-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2020] [Indexed: 02/07/2023]
Abstract
The study of heart repair post-myocardial infarction has historically focused on the importance of cardiomyocyte proliferation as the major factor limiting adult mammalian heart regeneration. However, there is mounting evidence that a narrow focus on this one cell type discounts the importance of a complex cascade of cell-cell communication involving a whole host of different cell types. A major difficulty in the study of heart regeneration is the rarity of this process in adult animals, meaning a mammalian template for how this can be achieved is lacking. Here, we review the adult zebrafish as an ideal and unique model in which to study the underlying mechanisms and cell types required to attain complete heart regeneration following cardiac injury. We provide an introduction to the role of the cardiac microenvironment in the complex regenerative process and discuss some of the key advances using this in vivo vertebrate model that have recently increased our understanding of the vital roles of multiple different cell types. Due to the sheer number of exciting studies describing new and unexpected roles for inflammatory cell populations in cardiac regeneration, this review will pay particular attention to these important microenvironment participants.
Collapse
Affiliation(s)
- Rebecca Ryan
- C21a, Biomedical Sciences Building, Faculty of Life Sciences, School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Bethany R Moyse
- C21a, Biomedical Sciences Building, Faculty of Life Sciences, School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Rebecca J Richardson
- C21a, Biomedical Sciences Building, Faculty of Life Sciences, School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol, BS8 1TD, UK.
| |
Collapse
|
36
|
Machikhin AS, Volkov MV, Burlakov AB, Khokhlov DD, Potemkin AV. Blood Vessel Imaging at Pre-Larval Stages of Zebrafish Embryonic Development. Diagnostics (Basel) 2020; 10:diagnostics10110886. [PMID: 33143148 PMCID: PMC7692510 DOI: 10.3390/diagnostics10110886] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/28/2020] [Accepted: 10/28/2020] [Indexed: 01/25/2023] Open
Abstract
The zebrafish (Danio rerio) is an increasingly popular animal model biological system. In cardiovascular research, it has been used to model specific cardiac phenomena as well as to identify novel therapies for human cardiovascular disease. While the zebrafish cardiovascular system functioning is well examined at larval stages, the mechanisms by which vessel activity is initiated remain a subject of intense investigation. In this research, we report on an in vivo stain-free blood vessel imaging technique at pre-larval stages of zebrafish embryonic development. We have developed the algorithm for the enhancement, alignment and spatiotemporal analysis of bright-field microscopy images of zebrafish embryos. It enables the detection, mapping and quantitative characterization of cardiac activity across the whole specimen. To validate the proposed approach, we have analyzed multiple data cubes, calculated vessel images and evaluated blood flow velocity and heart rate dynamics in the absence of any anesthesia. This non-invasive technique may shed light on the mechanism of vessel activity initiation and stabilization as well as the cardiovascular system’s susceptibility to environmental stressors at early developmental stages.
Collapse
Affiliation(s)
- Alexander S. Machikhin
- Laboratory of Acousto-optical Spectroscopy, Scientific and Technological Center of Unique Instrumentation, Russian Academy of Sciences, 117342 Moscow, Russia;
| | - Mikhail V. Volkov
- Department of Applied Optics, University ITMO, 190000 Saint Petersburg, Russia; (M.V.V.); (A.V.P.)
| | - Alexander B. Burlakov
- Department of Ichthyology, Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Demid D. Khokhlov
- Laboratory of Acousto-optical Spectroscopy, Scientific and Technological Center of Unique Instrumentation, Russian Academy of Sciences, 117342 Moscow, Russia;
- Correspondence:
| | - Andrey V. Potemkin
- Department of Applied Optics, University ITMO, 190000 Saint Petersburg, Russia; (M.V.V.); (A.V.P.)
| |
Collapse
|
37
|
Santoso F, Farhan A, Castillo AL, Malhotra N, Saputra F, Kurnia KA, Chen KHC, Huang JC, Chen JR, Hsiao CD. An Overview of Methods for Cardiac Rhythm Detection in Zebrafish. Biomedicines 2020; 8:E329. [PMID: 32899676 PMCID: PMC7554775 DOI: 10.3390/biomedicines8090329] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/29/2020] [Accepted: 09/01/2020] [Indexed: 12/17/2022] Open
Abstract
The heart is the most important muscular organ of the cardiovascular system, which pumps blood and circulates, supplying oxygen and nutrients to peripheral tissues. Zebrafish have been widely explored in cardiotoxicity research. For example, the zebrafish embryo has been used as a human heart model due to its body transparency, surviving several days without circulation, and facilitating mutant identification to recapitulate human diseases. On the other hand, adult zebrafish can exhibit the amazing regenerative heart muscle capacity, while adult mammalian hearts lack this potential. This review paper offers a brief description of the major methodologies used to detect zebrafish cardiac rhythm at both embryonic and adult stages. The dynamic pixel change method was mostly performed for the embryonic stage. Other techniques, such as kymography, laser confocal microscopy, artificial intelligence, and electrocardiography (ECG) have also been applied to study heartbeat in zebrafish embryos. Nevertheless, ECG is widely used for heartbeat detection in adult zebrafish since ECG waveforms' similarity between zebrafish and humans is prominent. High-frequency ultrasound imaging (echocardiography) and modern electronic sensor tag also have been proposed. Despite the fact that each method has its benefits and limitations, it is proved that zebrafish have become a promising animal model for human cardiovascular disease, drug pharmaceutical, and toxicological research. Using those tools, we conclude that zebrafish behaviors as an excellent small animal model to perform real-time monitoring for the developmental heart process with transparent body appearance, to conduct the in vivo cardiovascular performance and gene function assays, as well as to perform high-throughput/high content drug screening.
Collapse
Affiliation(s)
- Fiorency Santoso
- Master Program in Nanotechnology, Chung Yuan Christian University, Chung-Li 320314, Taiwan;
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 320314, Taiwan; (F.S.); (K.A.K.)
| | - Ali Farhan
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Punjab 38000, Pakistan;
| | - Agnes L. Castillo
- Faculty of Pharmacy, The Graduate School and Research Center for the Natural and Applied Sciences, University of Santo Tomas, Manila 1008, Philippines;
| | - Nemi Malhotra
- Department of Biomedical Engineering, Chung Yuan Christian University, Chung-Li 320314, Taiwan;
| | - Ferry Saputra
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 320314, Taiwan; (F.S.); (K.A.K.)
| | - Kevin Adi Kurnia
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 320314, Taiwan; (F.S.); (K.A.K.)
| | - Kelvin H.-C. Chen
- Department of Applied Chemistry, National Pingtung University, Pingtung 900391, Taiwan;
| | - Jong-Chin Huang
- Department of Applied Chemistry, National Pingtung University, Pingtung 900391, Taiwan;
| | - Jung-Ren Chen
- Department of Biological Science & Technology College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Chung-Der Hsiao
- Master Program in Nanotechnology, Chung Yuan Christian University, Chung-Li 320314, Taiwan;
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 320314, Taiwan; (F.S.); (K.A.K.)
- Center of Nanotechnology, Chung Yuan Christian University, Chung-Li 320314, Taiwan
| |
Collapse
|
38
|
Fabian L, Dowling JJ. Zebrafish Models of LAMA2-Related Congenital Muscular Dystrophy (MDC1A). Front Mol Neurosci 2020; 13:122. [PMID: 32742259 PMCID: PMC7364686 DOI: 10.3389/fnmol.2020.00122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/11/2020] [Indexed: 01/28/2023] Open
Abstract
LAMA2-related congenital muscular dystrophy (CMD; LAMA2-MD), also referred to as merosin deficient CMD (MDC1A), is a severe neonatal onset muscle disease caused by recessive mutations in the LAMA2 gene. LAMA2 encodes laminin α2, a subunit of the extracellular matrix (ECM) oligomer laminin 211. There are currently no treatments for MDC1A, and there is an incomplete understanding of disease pathogenesis. Zebrafish, due to their high degree of genetic conservation with humans, large clutch sizes, rapid development, and optical clarity, have emerged as an excellent model system for studying rare Mendelian diseases. They are particularly suitable as a model for muscular dystrophy because they contain at least one orthologue to all major human MD genes, have muscle that is similar to human muscle in structure and function, and manifest obvious and easily measured MD related phenotypes. In this review article, we present the existing zebrafish models of MDC1A, and discuss their contribution to the understanding of MDC1A pathomechanisms and therapy development.
Collapse
Affiliation(s)
- Lacramioara Fabian
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - James J Dowling
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada.,Division of Neurology, Hospital for Sick Children, Toronto, ON, Canada.,Departments of Pediatrics and Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
39
|
Mousavi SE, Patil JG. Light-cardiogram, a simple technique for heart rate determination in adult zebrafish, Danio rerio. Comp Biochem Physiol A Mol Integr Physiol 2020; 246:110705. [DOI: 10.1016/j.cbpa.2020.110705] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/26/2020] [Accepted: 04/07/2020] [Indexed: 12/24/2022]
|
40
|
Song Z, Zhang Y, Zhang H, Rajendran RS, Wang R, Hsiao CD, Li J, Xia Q, Liu K. Isoliquiritigenin triggers developmental toxicity and oxidative stress-mediated apoptosis in zebrafish embryos/larvae via Nrf2-HO1/JNK-ERK/mitochondrion pathway. CHEMOSPHERE 2020; 246:125727. [PMID: 31896010 DOI: 10.1016/j.chemosphere.2019.125727] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 06/10/2023]
Abstract
Isoliquiritigenin (ISL) is an emerging natural flavonoid found in the roots of licorice, exhibits antioxidant, anti-cancer, anti-inflammatory, anti-allergic, cardioprotective, hepatoprotective and neuroprotective properties. However, the effect of ISL in embryonic development is yet to be elucidated, and the mechanisms underlying its target-organ toxicity and harmful side effects are still unclear. In the present study, we employed zebrafish embryos to study the developmental toxicity effect of ISL and its underlying mechanisms. Zebrafish embryos upon treatment with either vehicle control (0.1% DMSO) or ISL solutions for 4-96 h post fertilization (hpf) showed that ISL exposure instigated severe developmental toxicity in heart, liver, and nervous system. Mortality and morphological abnormalities were also observed. High concentrations of ISL exposure resulted in abnormal phenotypes and embryonic malformations including pericardial edema, swim bladder defects, yolk retention, curved body shape and shortening of body length. Moreover, ISL exposure led to significant loss of dopaminergic neurons accompanied by reduced locomotor behaviour. Apoptotic cells were predominantly located in the heart area of 96 hpf embryo. Additionally, ISL significantly increased the levels of reactive oxygen species, lipid peroxidation content and decreased antioxidant enzyme activities. The expressions pattern of apoptosis-related genes Bad, Cyto c, Caspase-9, Caspase-3 and Bax/Bcl-2 indicated that the oxidative stress-induced apoptosis triggered by ISL suggest involvement of Nrf2-HO1/JNK-ERK/mitochondrion pathways. In conclusion, here we provide first evidence that demonstrate ISL-induced dose-dependent developmental toxicity in zebrafish embryos. Furthermore, gene expression patterns in the embryos correlate the above and reveal potential genetic mechanisms of developmental toxicity.
Collapse
Affiliation(s)
- Zhenzhen Song
- School of Pharmacy, Hebei University, Baoding, 071002, China; Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250103, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan, 250103, China
| | - Yun Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250103, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan, 250103, China
| | - Huazheng Zhang
- Shandong Academy of Chinese Medicine, Jinan, 250014, China
| | - R Samuel Rajendran
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250103, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan, 250103, China
| | - Rongchun Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250103, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan, 250103, China
| | - Chung-Der Hsiao
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li, 32023, China
| | - Jianheng Li
- School of Pharmacy, Hebei University, Baoding, 071002, China.
| | - Qing Xia
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250103, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan, 250103, China.
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250103, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan, 250103, China.
| |
Collapse
|
41
|
Shrestha R, Lieberth J, Tillman S, Natalizio J, Bloomekatz J. Using Zebrafish to Analyze the Genetic and Environmental Etiologies of Congenital Heart Defects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1236:189-223. [PMID: 32304074 DOI: 10.1007/978-981-15-2389-2_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Congenital heart defects (CHDs) are among the most common human birth defects. However, the etiology of a large proportion of CHDs remains undefined. Studies identifying the molecular and cellular mechanisms that underlie cardiac development have been critical to elucidating the origin of CHDs. Building upon this knowledge to understand the pathogenesis of CHDs requires examining how genetic or environmental stress changes normal cardiac development. Due to strong molecular conservation to humans and unique technical advantages, studies using zebrafish have elucidated both fundamental principles of cardiac development and have been used to create cardiac disease models. In this chapter we examine the unique toolset available to zebrafish researchers and how those tools are used to interrogate the genetic and environmental contributions to CHDs.
Collapse
Affiliation(s)
- Rabina Shrestha
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | - Jaret Lieberth
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | - Savanna Tillman
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | - Joseph Natalizio
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | | |
Collapse
|
42
|
Exposure to low doses of inorganic arsenic induces transgenerational changes on behavioral and epigenetic markers in zebrafish (Danio rerio). Toxicol Appl Pharmacol 2020; 396:115002. [PMID: 32277946 DOI: 10.1016/j.taap.2020.115002] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/26/2020] [Accepted: 04/07/2020] [Indexed: 02/06/2023]
Abstract
The ability of environmental pollutants to alter the epigenome with resultant development of behavioral alterations has received more attention in recent years. These alterations can be transmitted and affect later generations that have not been directly in contact with the contaminant. Arsenic (As) is a neurotoxicant and potent epigenetic disruptor that is widespread in the environment; however, the precise potential of As to produce transgenerational effects is unknown. Our study focused on the possible transgenerational effects on behavior by ancestral exposure to doses relevant to the environment of As, and the epigenetic mechanisms that could be involved. Embryos of F0 (ancestral generation) were directly exposed to 50 or 500 ppb of As for 150 days. F0 adults were raised to produce the F1 generation (intergeneration) and subsequently the F2 generation (transgeneration). We evaluated motor and cognitive behavior, neurodevelopment-related genes, and epigenetic markers on the F0 and F2 generation. As proposed in our hypothesis, ancestral arsenic exposure altered motor activity through the development and increased anxiety-like behaviors which were transmitted to the F2 generation. Additionally, we found a reduction in brain-derived neurotrophic factor expression between the F0 and F2 generation, and an increase in methylation on histone H3K4me3 in the nervous system.
Collapse
|
43
|
Generation and Characterization of a CRISPR/Cas9 -Induced 3-mst Deficient Zebrafish. Biomolecules 2020; 10:biom10020317. [PMID: 32079278 PMCID: PMC7072312 DOI: 10.3390/biom10020317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 12/12/2022] Open
Abstract
3-mercaptopyruvate sulfurtransferase (3-MST) is an enzyme capable of synthesizing hydrogen sulfide (H2S) and polysulfides. In spite of its ubiquitous presence in mammalian cells, very few studies have investigated its contribution to homeostasis and disease development, thus the role of 3-MST remains largely unexplored. Here, we present a clustered, regularly interspaced, short palindromic repeats (CRISPR)/CRISPR-associated protein-9 (Cas9) induced 3-mst mutant zebrafish line, which will allow the study of 3-MST's role in several biological processes. The 3-mst zebrafish orthologue was identified using a bioinformatic approach and verified by its ability to produce H2S in the presence of 3-mercaptopyruvate (3-MP). Its expression pattern was analyzed during zebrafish early development, indicating predominantly an expression in the heart and central nervous system. As expected, no detectable levels of 3-Mst protein were observed in homozygous mutant larvae. In line with this, H2S levels were reduced in 3-mst-/- zebrafish. Although the mutants showed no obvious morphological deficiencies, they exhibited increased lethality under oxidative stress conditions. The elevated levels of reactive oxygen species, detected following 3-mst deletion, are likely to drive this phenotype. In line with the increased ROS, we observed accelerated fin regenerative capacity in 3-mst deficient zebrafish. Overall, we provide evidence for the expression of 3-mst in zebrafish, confirm its important role in redox homeostasis and indicate the enzyme's possible involvement in the regeneration processes.
Collapse
|
44
|
Pott A, Rottbauer W, Just S. Streamlining drug discovery assays for cardiovascular disease using zebrafish. Expert Opin Drug Discov 2019; 15:27-37. [PMID: 31570020 DOI: 10.1080/17460441.2020.1671351] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: In the last decade, our armamentarium of cardiovascular drug therapy has expanded significantly. Using innovative functional genomics strategies such as genome editing by CRISPR/Cas9 as well as high-throughput assays to identify bioactive small chemical compounds has significantly facilitated elaboration of the underlying pathomechanism in various cardiovascular diseases. However, despite scientific progress approvals for cardiovascular drugs has stagnated significantly compared to other fields of drug discovery and therapy during the past years.Areas covered: In this review, the authors discuss the aspects and pitfalls during the early phase of cardiovascular drug discovery and describe the advantages of zebrafish as an in vivo organism to model human cardiovascular diseases (CVD) as well as an in vivo platform for high-throughput chemical compound screening. They also highlight the emerging, promising techniques of automated read-out systems during high-throughput screening (HTS) for the evaluation of important cardiac functional parameters in zebrafish with the potential to streamline CVD drug discovery.Expert opinion: The successful identification of novel drugs to treat CVD is a major challenge in modern biomedical and clinical research. In this context, the definition of the etiologic fundamentals of human cardiovascular diseases is the prerequisite for an efficient and straightforward drug discovery.
Collapse
Affiliation(s)
- Alexander Pott
- Internal Medicine II, Ulm University Medical Center, Ulm, Germany.,Molecular Cardiology, Internal Medicine II, Ulm University Medical Center, Ulm, Germany
| | | | - Steffen Just
- Molecular Cardiology, Internal Medicine II, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|