1
|
Angom RS, Singh M, Muhammad H, Varanasi SM, Mukhopadhyay D. Zebrafish as a Versatile Model for Cardiovascular Research: Peering into the Heart of the Matter. Cells 2025; 14:531. [PMID: 40214485 PMCID: PMC11988917 DOI: 10.3390/cells14070531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/25/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death in the world. A total of 17.5 million people died of CVDs in the year 2012, accounting for 31% of all deaths globally. Vertebrate animal models have been used to understand cardiac disease biology, as the cellular, molecular, and physiological aspects of human CVDs can be replicated closely in these organisms. Zebrafish is a popular model organism offering an arsenal of genetic tools that allow the rapid in vivo analysis of vertebrate gene function and disease conditions. It has a short breeding cycle, high fecundity, optically transparent embryos, rapid internal organ development, and easy maintenance. This review aims to give readers an overview of zebrafish cardiac biology and a detailed account of heart development in zebrafish and its comparison with humans and the conserved genetic circuitry. We also discuss the contributions made in CVD research using the zebrafish model. The first part of this review focuses on detailed information on the morphogenetic and differentiation processes in early cardiac development. The overlap and divergence of the human heart's genetic circuitry, structure, and physiology are emphasized wherever applicable. In the second part of the review, we overview the molecular tools and techniques available to dissect gene function and expression in zebrafish, with special mention of the use of these tools in cardiac biology.
Collapse
Affiliation(s)
- Ramcharan Singh Angom
- Department of Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine and Science, Jacksonville, FL 32224, USA; (R.S.A.); (H.M.); (S.M.V.)
| | - Meghna Singh
- Department of Pathology and Lab Medicine, University of California, Los Angeles, CA 92093, USA;
| | - Huzaifa Muhammad
- Department of Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine and Science, Jacksonville, FL 32224, USA; (R.S.A.); (H.M.); (S.M.V.)
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Sai Manasa Varanasi
- Department of Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine and Science, Jacksonville, FL 32224, USA; (R.S.A.); (H.M.); (S.M.V.)
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine and Science, Jacksonville, FL 32224, USA; (R.S.A.); (H.M.); (S.M.V.)
| |
Collapse
|
2
|
Guo Z, Ai N, Ge W, Xu Q. ZebraVas: A Non-Invasive Microvision System for Vascular Recognition and Blood Flow Monitoring of Zebrafish Larvae. IEEE Trans Nanobioscience 2025; 24:225-233. [PMID: 40030690 DOI: 10.1109/tnb.2024.3520137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Zebrafish have emerged as a powerful model organism in cardiovascular disease research. Accurately identifying zebrafish blood vessels and evaluating blood flow velocity without injury has a wide range of biological applications. This paper presents the design and development of a non-invasive microvision system for vascular recognition and blood flow monitoring of zebrafish larvae. For the first time, a visual algorithm based on color thresholding and discrete Fourier transform filtering is proposed to determine the position of zebrafish dorsal cardinal vein vessels. Next, the blood flow velocity is determined based on the change rate of pixel values near the centroid point of the blood vessel recognition results. Then, an independent software system is developed based on the producer-consumer underlying framework. A user-friendly interface is specifically designed for biomedical workers, and a complete prototype system is built in combination with hardware devices. In addition, relevant experiments were conducted, and the results indicated that the system can effectively recognize the position of vessels and monitor blood flow velocity in zebrafish larvae under different anesthesia concentrations and developmental days. The heart rate information obtained based on blood flow velocity is consistent with the heart beating frequency. Moreover, the system has also been successfully applied to blood flow velocity monitoring under fluorescence conditions. In future work, this system will be applied in drug screening research for cardiovascular-related diseases of zebrafish larvae.
Collapse
|
3
|
Counil H, Silva RO, Rabanel J, Zaouter C, Haddad M, Ben Khedher MR, Brambilla D, Fülöp T, Patten SA, Ramassamy C. Brain penetration of peripheral extracellular vesicles from Alzheimer's patients and induction of microglia activation. JOURNAL OF EXTRACELLULAR BIOLOGY 2025; 4:e70027. [PMID: 39830834 PMCID: PMC11740088 DOI: 10.1002/jex2.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/13/2024] [Accepted: 12/08/2024] [Indexed: 01/22/2025]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative pathology. Brain-derived extracellular vesicles (EVs) have been demonstrated to be implicated in AD pathogenesis by facilitating the propagation of Tau, amyloid-β and inflammatory cytokines. However, the impact of peripheral EVs (pEVs) in AD pathogenesis remains poorly investigated. The objective of our study was to compare the passage of pEVs from adults, cognitively healthy elderly, and AD patients through the blood-brain barrier (BBB), to evaluate their uptake in the brain and to assess their impact on the microglia activity using in vitro and in vivo models. To this end, pEVs were enriched, characterized, and fluorescently labelled. The passage of pEVs through the endothelial bEnd.3 cells was studied in a Transwell device with either neuronal or microglia cells seeded at the bottom of the well. Following the internalization of pEVs from AD patients, microglia adopted an amoeboid morphology and released a heightened level of pro-inflammatory cytokine IL-6. To further assess their in vivo transport across the BBB, pEVs were injected into the blood circulation of 2-days post-fertilization Tg(flk1:EGFP) zebrafish. The biodistribution of pEVs was monitored at 1 and 24 h post-injection using confocal microscopy. We demonstrated that pEVs traverse the BBB by transcytosis and subsequently diffuse progressively into the brain. pEVs were then internalized by neuronal and radial glial cells as seen in Tg(huc:EGFP) and Tg(gfap:EGFP) zebrafish, respectively. Additional experiments were performed with the intrahippocampal injection of pEVs in the mouse, indicating their spreading throughout the brain and their uptake by neuronal and glial cells. These findings contribute to novel insights into the fate of pEVs following their passage through the BBB in vitro and in vivo, and demonstrate for the first time that pEVs from AD patients affect microglia activity. This suggests a potential mechanism through which peripheral tissue cues may contribute to AD pathogenesis.
Collapse
Affiliation(s)
- Hermine Counil
- INRSCentre Armand‐Frappier Santé BiotechnologieLavalQuebecCanada
| | | | - Jean‐Michel Rabanel
- INRSCentre Armand‐Frappier Santé BiotechnologieLavalQuebecCanada
- School of Pharmaceutical Sciences, Faculty of MedecineUniversity of OttawaOttawaOntarioCanada
| | | | - Mohamed Haddad
- INRSCentre Armand‐Frappier Santé BiotechnologieLavalQuebecCanada
| | - Mohamed Raâfet Ben Khedher
- INRSCentre Armand‐Frappier Santé BiotechnologieLavalQuebecCanada
- Higher Institute of Biotechnology of BejaUniversity of JendoubaBejaTunisia
| | - Davide Brambilla
- Université de MontréalFaculté de Pharmacie, Pavillon Jean‐CoutuMontréalQuebecCanada
| | - Tamas Fülöp
- Research Center on Aging, Graduate Program in Immunology, Faculty of Medicine and Health SciencesUniversity of SherbrookeSherbrookeQuebecCanada
| | | | | |
Collapse
|
4
|
Spratt J, Dias JM, Kolonelou C, Kiriako G, Engström E, Petrova E, Karampelias C, Cervenka I, Papanicolaou N, Lentini A, Reinius B, Andersson O, Ambrosetti E, Ruas JL, Teixeira AI. Multivalent insulin receptor activation using insulin-DNA origami nanostructures. NATURE NANOTECHNOLOGY 2024; 19:237-245. [PMID: 37813939 PMCID: PMC10873203 DOI: 10.1038/s41565-023-01507-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 08/15/2023] [Indexed: 10/11/2023]
Abstract
Insulin binds the insulin receptor (IR) and regulates anabolic processes in target tissues. Impaired IR signalling is associated with multiple diseases, including diabetes, cancer and neurodegenerative disorders. IRs have been reported to form nanoclusters at the cell membrane in several cell types, even in the absence of insulin binding. Here we exploit the nanoscale spatial organization of the IR to achieve controlled multivalent receptor activation. To control insulin nanoscale spatial organization and valency, we developed rod-like insulin-DNA origami nanostructures carrying different numbers of insulin molecules with defined spacings. Increasing the insulin valency per nanostructure markedly extended the residence time of insulin-DNA origami nanostructures at the receptors. Both insulin valency and spacing affected the levels of IR activation in adipocytes. Moreover, the multivalent insulin design associated with the highest levels of IR activation also induced insulin-mediated transcriptional responses more effectively than the corresponding monovalent insulin nanostructures. In an in vivo zebrafish model of diabetes, treatment with multivalent-but not monovalent-insulin nanostructures elicited a reduction in glucose levels. Our results show that the control of insulin multivalency and spatial organization with nanoscale precision modulates the IR responses, independent of the insulin concentration. Therefore, we propose insulin nanoscale organization as a design parameter in developing new insulin therapies.
Collapse
Affiliation(s)
- Joel Spratt
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - José M Dias
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Christina Kolonelou
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Georges Kiriako
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Enya Engström
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ekaterina Petrova
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Christos Karampelias
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Igor Cervenka
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Natali Papanicolaou
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Antonio Lentini
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Björn Reinius
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Olov Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Elena Ambrosetti
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Jorge L Ruas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ana I Teixeira
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
5
|
Zebrafish as a powerful alternative model organism for preclinical investigation of nanomedicines. Drug Discov Today 2022; 27:1513-1522. [DOI: 10.1016/j.drudis.2022.02.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 11/28/2021] [Accepted: 02/16/2022] [Indexed: 12/14/2022]
|
6
|
Cardiac forces regulate zebrafish heart valve delamination by modulating Nfat signaling. PLoS Biol 2022; 20:e3001505. [PMID: 35030171 PMCID: PMC8794269 DOI: 10.1371/journal.pbio.3001505] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/27/2022] [Accepted: 12/06/2021] [Indexed: 11/30/2022] Open
Abstract
In the clinic, most cases of congenital heart valve defects are thought to arise through errors that occur after the endothelial–mesenchymal transition (EndoMT) stage of valve development. Although mechanical forces caused by heartbeat are essential modulators of cardiovascular development, their role in these later developmental events is poorly understood. To address this question, we used the zebrafish superior atrioventricular valve (AV) as a model. We found that cellularized cushions of the superior atrioventricular canal (AVC) morph into valve leaflets via mesenchymal–endothelial transition (MEndoT) and tissue sheet delamination. Defects in delamination result in thickened, hyperplastic valves, and reduced heart function. Mechanical, chemical, and genetic perturbation of cardiac forces showed that mechanical stimuli are important regulators of valve delamination. Mechanistically, we show that forces modulate Nfatc activity to control delamination. Together, our results establish the cellular and molecular signature of cardiac valve delamination in vivo and demonstrate the continuous regulatory role of mechanical forces and blood flow during valve formation. Why do developing zebrafish atrioventricular heart valves become hyperplastic under certain hemodynamic conditions? This study suggests that part of the answer lies in how the mechanosensitive Nfat pathway regulates the valve mesenchymal-to-endothelial transition.
Collapse
|
7
|
Zheng Y, Zhao J, Li J, Guo Z, Sheng J, Ye X, Jin G, Wang C, Chai W, Yan J, Liu D, Liang X. SARS-CoV-2 spike protein causes blood coagulation and thrombosis by competitive binding to heparan sulfate. Int J Biol Macromol 2021; 193:1124-1129. [PMID: 34743814 PMCID: PMC8553634 DOI: 10.1016/j.ijbiomac.2021.10.112] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/13/2021] [Accepted: 10/16/2021] [Indexed: 12/24/2022]
Abstract
Thrombotic complication has been an important symptom in critically ill patients with COVID-19. It has not been clear whether the virus spike (S) protein can directly induce blood coagulation in addition to inflammation. Heparan sulfate (HS)/heparin, a key factor in coagulation process, was found to bind SARS-CoV-2 S protein with high affinity. Herein, we found that the S protein can competitively inhibit the bindings of antithrombin and heparin cofactor II to heparin/HS, causing abnormal increase in thrombin activity. SARS-CoV-2 S protein at a similar concentration (~10 μg/mL) as the viral load in critically ill patients can cause directly blood coagulation and thrombosis in zebrafish model. Furthermore, exogenous heparin/HS can significantly reduce coagulation caused by S protein, pointing to a potential new direction to elucidate the etiology of the virus and provide fundamental support for anticoagulant therapy especially for the COVID-19 critically ill patients.
Collapse
Affiliation(s)
- Yi Zheng
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Jinxiang Zhao
- Nantong Laboratory of Development and Diseases, School of Life Science, Co-innovation Center of Neuroregeneration, Key Laboratory of Neuroregeneration of Jiangsu, Ministry of Education, Nantong University, Nantong 226019, China
| | - Jiaqi Li
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Zhimou Guo
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Jiajing Sheng
- Nantong Laboratory of Development and Diseases, School of Life Science, Co-innovation Center of Neuroregeneration, Key Laboratory of Neuroregeneration of Jiangsu, Ministry of Education, Nantong University, Nantong 226019, China
| | - Xianlong Ye
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Gaowa Jin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Chaoran Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Wengang Chai
- Glycosciences Laboratory, Faculty of Medicine, Imperial College London, Hammersmith Campus, London W12 0NN, United Kingdom
| | - Jingyu Yan
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Dong Liu
- Nantong Laboratory of Development and Diseases, School of Life Science, Co-innovation Center of Neuroregeneration, Key Laboratory of Neuroregeneration of Jiangsu, Ministry of Education, Nantong University, Nantong 226019, China.
| | - Xinmiao Liang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| |
Collapse
|
8
|
Tao J, Wei Z, Xu M, Xi L, Cheng Y, Lee SMY, Ge W, Zheng Y. Particle Integrity and Size Effect on the Journey of Polymeric Nanocarriers in Zebrafish Model and the Correlation with Mice. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2103584. [PMID: 34528394 DOI: 10.1002/smll.202103584] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/15/2021] [Indexed: 05/25/2023]
Abstract
Polymeric nanocarriers have high biocompatibility for potential drug delivery applications. After entering bloodstream, nanocarriers will circulate, interact with proteins, dissociate, or be cleared by reticuloendothelial system. Zebrafish as a visual animal model, can serve as a tool for screening nanomedicines and monitoring nanocarrier behaviors in vivo. However, a comprehensive correlation between zebrafish and rodent models is currently deficient. Here, different-sized poly(caprolactone) nanocarriers (PCL NCs) are fabricated with or without PEGylation to investigate correlation between zebrafish and mice regarding their biofate via Förster resonance energy transfer technique. Results show that PEGylated PCL NCs have higher integrity in both zebrafish and mice. Small PEG-PCL NCs have longer circulation, while large PEG-PCL NCs have dramatically higher macrophage sequestration in zebrafish and mice spleen, leading to poor circulation. PCL NCs dissociate rapidly with less macrophage sequestration. Moreover, in 7 days postfertilization (dpf) zebrafish, polymers are eliminated via hepatobiliary pathway, which is not fully functional at earlier stages of development. The effects of nanocarrier integrity on macrophage sequestration in zebrafish and good correlation with mice spleen are pioneered to be demonstrated. The findings suggest that 7 dpf zebrafish are suitable as an in vivo screening model of nanocarriers and predict their biofate in rodents.
Collapse
Affiliation(s)
- Jinsong Tao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Zhengjie Wei
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Meng Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Long Xi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Yaxin Cheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Wei Ge
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Ying Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| |
Collapse
|
9
|
Stabilin-1 is required for the endothelial clearance of small anionic nanoparticles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 34:102395. [PMID: 33838334 DOI: 10.1016/j.nano.2021.102395] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/25/2021] [Accepted: 03/18/2021] [Indexed: 02/07/2023]
Abstract
Clearance of nanoparticles (NPs) after intravenous injection - mainly by the liver - is a critical barrier for the clinical translation of nanomaterials. Physicochemical properties of NPs are known to influence their distribution through cell-specific interactions; however, the molecular mechanisms responsible for liver cellular NP uptake are poorly understood. Liver sinusoidal endothelial cells and Kupffer cells are critical participants in this clearance process. Here we use a zebrafish model for liver-NP interaction to identify the endothelial scavenger receptor Stabilin-1 as a non-redundant receptor for the clearance of small anionic NPs. Furthermore, we show that physiologically, Stabilin-1 is required for the removal of bacterial lipopolysaccharide (LPS/endotoxin) from circulation and that Stabilin-1 cooperates with its homolog Stabilin-2 in the clearance of larger (~100 nm) anionic NPs. Our findings allow optimization of anionic nanomedicine biodistribution and targeting therapies that use Stabilin-1 and -2 for liver endothelium-specific delivery.
Collapse
|
10
|
Zhu XY, Xia B, Ye T, Dai MZ, Yang H, Li CQ, Li P. Ponatinib-induced ischemic stroke in larval zebrafish for drug screening. Eur J Pharmacol 2020; 889:173292. [PMID: 32668288 DOI: 10.1016/j.ejphar.2020.173292] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 05/16/2020] [Accepted: 06/17/2020] [Indexed: 02/07/2023]
Abstract
Conventional mammalian ischemic stroke models for drug screening are technically challenging, laborious and time-consuming. In this study, using Ponatinib as an inducer, we developed and characterized a zebrafish ischemic stroke model. This zebrafish ischemic stroke had the cerebral vascular endothelial injury, thrombosis, reduced blood flow, inflammation and apoptosis as well as the reduced motility. The zebrafish ischemic stroke model was validated with 6 known human therapeutic drugs of ischemic stroke (Aspirin, Clopidogrel, Naoxintong capsules, Edaravone, Xingnaojing injection, Shuxuening injection). The mRNA levels of the neovascularization-related gene (vegfaa) and vascular endothelial growth factor receptor gene (VEGFR), neurodevelopment related genes (mbp and α1-tubulin), brain-derived neurotrophic factor (BDNF) and glial cell derived neurotrophic factor (GDNF) were significantly downregulated; whereas apoptosis-related genes (caspase-3, caspase-7, caspase-9 and bax/bcl-2), and inflammatory factor genes (IL-1β, IL-6, IL-10, TNF-α and NF-κB) were remarkably upregulated in the model. These results suggest that the pathophysiology of Ponatinib-induced zebrafish ischemic stroke is similar to that of human ischemic stroke patients and this whole animal model could be used to study the complex cellular and molecular pathogenesis of ischemic stroke and to rapidly identify therapeutic agents.
Collapse
Affiliation(s)
- Xiao-Yu Zhu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu Province, 210009, PR China; Hunter Biotechnology, Inc, F1A, Building 5, No. 88 Jiangling Road, Binjiang Zone, Hangzhou City, Zhejiang Province, 310051, PR China
| | - Bo Xia
- Hunter Biotechnology, Inc, F1A, Building 5, No. 88 Jiangling Road, Binjiang Zone, Hangzhou City, Zhejiang Province, 310051, PR China
| | - Ting Ye
- Hunter Biotechnology, Inc, F1A, Building 5, No. 88 Jiangling Road, Binjiang Zone, Hangzhou City, Zhejiang Province, 310051, PR China
| | - Ming-Zhu Dai
- Hunter Biotechnology, Inc, F1A, Building 5, No. 88 Jiangling Road, Binjiang Zone, Hangzhou City, Zhejiang Province, 310051, PR China
| | - Hua Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu Province, 210009, PR China
| | - Chun-Qi Li
- Hunter Biotechnology, Inc, F1A, Building 5, No. 88 Jiangling Road, Binjiang Zone, Hangzhou City, Zhejiang Province, 310051, PR China.
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu Province, 210009, PR China.
| |
Collapse
|
11
|
Zhou Y, Zammit P, Zickus V, Taylor JM, Harvey AR. Twin-Airy Point-Spread Function for Extended-Volume Particle Localization. PHYSICAL REVIEW LETTERS 2020; 124:198104. [PMID: 32469536 DOI: 10.1103/physrevlett.124.198104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 11/11/2019] [Accepted: 04/08/2020] [Indexed: 06/11/2023]
Abstract
The localization of point sources in optical microscopy enables nm-precision imaging of single-molecules and biological dynamics. We report a new method of localization microscopy using twin Airy beams that yields precise 3D localization with the key advantages of extended depth range, higher optical throughput, and potential for imaging higher emitter densities than are possible using other techniques. A precision of better than 30 nm was achieved over a depth range in excess of 7 μm using a 60×, 1.4 NA objective. An illustrative application to extended-depth-range blood-flow imaging in a live zebrafish is also demonstrated.
Collapse
Affiliation(s)
- Yongzhuang Zhou
- School of Physics & Astronomy, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Paul Zammit
- School of Physics & Astronomy, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Vytautas Zickus
- School of Physics & Astronomy, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Jonathan M Taylor
- School of Physics & Astronomy, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Andrew R Harvey
- School of Physics & Astronomy, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| |
Collapse
|
12
|
Saez Talens V, Arias-Alpizar G, Makurat DMM, Davis J, Bussmann J, Kros A, Kieltyka RE. Stab2-Mediated Clearance of Supramolecular Polymer Nanoparticles in Zebrafish Embryos. Biomacromolecules 2020; 21:1060-1068. [DOI: 10.1021/acs.biomac.9b01318] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Victorio Saez Talens
- Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, PO Box 9502, 2300 RA Leiden, The Netherlands
| | - Gabriela Arias-Alpizar
- Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, PO Box 9502, 2300 RA Leiden, The Netherlands
| | - D. M. M. Makurat
- Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, PO Box 9502, 2300 RA Leiden, The Netherlands
| | - Joyal Davis
- Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, PO Box 9502, 2300 RA Leiden, The Netherlands
| | - Jeroen Bussmann
- Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, PO Box 9502, 2300 RA Leiden, The Netherlands
| | - Alexander Kros
- Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, PO Box 9502, 2300 RA Leiden, The Netherlands
| | - Roxanne E. Kieltyka
- Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, PO Box 9502, 2300 RA Leiden, The Netherlands
| |
Collapse
|
13
|
Peerzade SAMA, Qin X, Laroche FJ, Palantavida S, Dokukin M, Feng H, Sokolov I. Ultrabright fluorescent silica nanoparticles for in vivo targeting of xenografted human tumors and cancer cells in zebrafish. NANOSCALE 2019; 11:22316-22327. [PMID: 31724677 PMCID: PMC7384872 DOI: 10.1039/c9nr06371d] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
New ultrabright fluorescent silica nanoparticles capable of the fast targeting of epithelial tumors in vivo are presented. The as-synthesized folate-functionalized ultrabright particles of 30-40 nm are 230 times brighter than quantum dots (QD450) and 50% brighter than the polymer dots with similar spectra (excitation 365 nm and emission 486 nm). To decrease non-specific targeting, particles are coated with polyethylene glycol (PEG). We demonstrate the in vivo targeting of xenographic human cervical epithelial tumors (HeLa cells) using zebrafish as a model system. The particles target tumors (and probably even individual HeLa cells) as small as 10-20 microns within 20-30 minutes after blood injection. To demonstrate the advantages of ultrabrightness, we repeated the experiments with similar but 200× less bright particles. Compared to those, ultrabright particles showed ∼3× faster tumor detection and ∼2× higher relative fluorescent contrast of tumors/cancer cells.
Collapse
Affiliation(s)
| | - Xiaodan Qin
- Departments of Pharmacology and Medicine, The Cancer Research Center, Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, USA
| | - Fabrice J.F. Laroche
- Departments of Pharmacology and Medicine, The Cancer Research Center, Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, USA
| | - Shajesh Palantavida
- Department of Mechanical Engineering, Tufts University, Medford, MA 02155, USA
| | - Maxim Dokukin
- Department of Mechanical Engineering, Tufts University, Medford, MA 02155, USA
| | - Hui Feng
- Departments of Pharmacology and Medicine, The Cancer Research Center, Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, USA
| | - Igor Sokolov
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
- Department of Mechanical Engineering, Tufts University, Medford, MA 02155, USA
- Department of Physics, Tufts University, Medford, MA 02155, USA
| |
Collapse
|
14
|
Sieber S, Grossen P, Bussmann J, Campbell F, Kros A, Witzigmann D, Huwyler J. Zebrafish as a preclinical in vivo screening model for nanomedicines. Adv Drug Deliv Rev 2019; 151-152:152-168. [PMID: 30615917 DOI: 10.1016/j.addr.2019.01.001] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/23/2018] [Accepted: 01/02/2019] [Indexed: 12/11/2022]
Abstract
The interactions of nanomedicines with biological environments is heavily influenced by their physicochemical properties. Formulation design and optimization are therefore key steps towards successful nanomedicine development. Unfortunately, detailed assessment of nanomedicine formulations, at a macromolecular level, in rodents is severely limited by the restricted imaging possibilities within these animals. Moreover, rodent in vivo studies are time consuming and expensive, limiting the number of formulations that can be practically assessed in any one study. Consequently, screening and optimisation of nanomedicine formulations is most commonly performed in surrogate biological model systems, such as human-derived cell cultures. However, despite the time and cost advantages of classical in vitro models, these artificial systems fail to reflect and mimic the complex biological situation a nanomedicine will encounter in vivo. This has acutely hampered the selection of potentially successful nanomedicines for subsequent rodent in vivo studies. Recently, zebrafish have emerged as a promising in vivo model, within nanomedicine development pipelines, by offering opportunities to quickly screen nanomedicines under in vivo conditions and in a cost-effective manner so as to bridge the current gap between in vitro and rodent studies. In this review, we outline several advantageous features of the zebrafish model, such as biological conservation, imaging modalities, availability of genetic tools and disease models, as well as their various applications in nanomedicine development. Critical experimental parameters are discussed and the most beneficial applications of the zebrafish model, in the context of nanomedicine development, are highlighted.
Collapse
Affiliation(s)
- Sandro Sieber
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Philip Grossen
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Jeroen Bussmann
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Frederick Campbell
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Alexander Kros
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Dominik Witzigmann
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland; Department of Biochemistry and Molecular Biology, University of British Columbia, Health Sciences Mall, Vancouver, British Columbia, Canada..
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.
| |
Collapse
|
15
|
Nanomaterials meet zebrafish: Toxicity evaluation and drug delivery applications. J Control Release 2019; 311-312:301-318. [PMID: 31446084 DOI: 10.1016/j.jconrel.2019.08.022] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 08/19/2019] [Accepted: 08/21/2019] [Indexed: 12/23/2022]
Abstract
With the rapid development of engineered nanomaterials for various applications, in vivo toxicological studies for evaluating the potential hazardous effects of nanomaterials on environmental and human safety are in urgent need. Zebrafish has long been considered as the "gold standard" for biosafety assessments of chemicals and pollutants due to its high fecundity, cost-effectiveness, well-characterized developmental stages, optical transparency, and so forth. Thus, zebrafish holds great potential for high-throughput nanotoxicity screening. In this review, we summarize the in vivo toxicological profiles of different nanomaterials, including Ag nanoparticles (NPs), CuO NPs, silica NPs, polymeric NPs, quantum dots, nanoscale metal-organic frameworks, etc, in zebrafish and focus on how the physicochemical properties (e.g., size, surface charge, and surface chemistry) of these nanomaterials influence their biosafety. In addition, we also report the recent advances of the in vivo delivery of nanopharmaceuticals using zebrafish as the model organism for therapeutic assessment, biodistribution tracking, and the controlled release of loaded drugs. Limitations and special considerations of zebrafish model are also discussed. Overall, zebrafish is expected to serve as a high-throughput screening platform for nanotoxicity and drug delivery assessment, which may instruct the design of safe nanomaterials and more effective nanomedicines.
Collapse
|
16
|
Cordero-Maldonado ML, Perathoner S, van der Kolk KJ, Boland R, Heins-Marroquin U, Spaink HP, Meijer AH, Crawford AD, de Sonneville J. Deep learning image recognition enables efficient genome editing in zebrafish by automated injections. PLoS One 2019; 14:e0202377. [PMID: 30615627 PMCID: PMC6322765 DOI: 10.1371/journal.pone.0202377] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 12/13/2018] [Indexed: 12/30/2022] Open
Abstract
One of the most popular techniques in zebrafish research is microinjection. This is a rapid and efficient way to genetically manipulate early developing embryos, and to introduce microbes, chemical compounds, nanoparticles or tracers at larval stages. Here we demonstrate the development of a machine learning software that allows for microinjection at a trained target site in zebrafish eggs at unprecedented speed. The software is based on the open-source deep-learning library Inception v3. In a first step, the software distinguishes wells containing embryos at one-cell stage from wells to be skipped with an accuracy of 93%. A second step was developed to pinpoint the injection site. Deep learning allows to predict this location on average within 42 μm to manually annotated sites. Using a Graphics Processing Unit (GPU), both steps together take less than 100 milliseconds. We first tested our system by injecting a morpholino into the middle of the yolk and found that the automated injection efficiency is as efficient as manual injection (~ 80%). Next, we tested both CRISPR/Cas9 and DNA construct injections into the zygote and obtained a comparable efficiency to that of an experienced experimentalist. Combined with a higher throughput, this results in a higher yield. Hence, the automated injection of CRISPR/Cas9 will allow high-throughput applications to knock out and knock in relevant genes to study their mechanisms or pathways of interest in diverse areas of biomedical research.
Collapse
Affiliation(s)
| | - Simon Perathoner
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | | | - Ralf Boland
- Institute of Biology, Leiden University, Leiden, the Netherlands
| | - Ursula Heins-Marroquin
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Herman P. Spaink
- Institute of Biology, Leiden University, Leiden, the Netherlands
| | | | - Alexander D. Crawford
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | | |
Collapse
|
17
|
Zhou Y, Zickus V, Zammit P, Taylor JM, Harvey AR. High-speed extended-volume blood flow measurement using engineered point-spread function. BIOMEDICAL OPTICS EXPRESS 2018; 9:6444-6454. [PMID: 31065441 PMCID: PMC6490974 DOI: 10.1364/boe.9.006444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 09/25/2018] [Indexed: 06/09/2023]
Abstract
Experimental characterization of blood flow in living organisms is crucial for understanding the development and function of cardiovascular systems, but there has been no technique reported for snapshot imaging of thick samples in large volumes with high precision. We have combined computational microscopy and the diffraction-free, self-bending property of Airy-beams to track fluorescent beads with sub-micron precision through an extended axial range (up to 600 μm) within the flowing blood of 3 days post-fertilization (dpf) zebrafish embryos. The spatial trajectories of the tracer beads within flowing blood were recorded during transit through both cardinal and intersegmental vessels, and the trajectories were found to be consistent with the segmentation of the vasculature recorded using selective-plane illumination microscopy (SPIM). This method provides sufficiently precise spatial and temporal measurement of 3D blood flow that has the potential for directly probing key biomechanical quantities such as wall shear stress, as well as exploring the fluidic repercussions of cardiovascular diseases. Although we demonstrate the technique for blood flow, the ten-fold better enhancement in the depth range offers improvements in a wide range of applications of high-speed precision measurement of fluid flow, from microfluidics through measurement of cell dynamics to macroscopic aerosol characterizations.
Collapse
Affiliation(s)
- Yongzhuang Zhou
- School of Physics & Astronomy, University of Glasgow, Glasgow, G12 8QQ,
UK
| | - Vytautas Zickus
- School of Physics & Astronomy, University of Glasgow, Glasgow, G12 8QQ,
UK
| | - Paul Zammit
- School of Physics & Astronomy, University of Glasgow, Glasgow, G12 8QQ,
UK
| | - Jonathan M. Taylor
- School of Physics & Astronomy, University of Glasgow, Glasgow, G12 8QQ,
UK
| | - Andrew R. Harvey
- School of Physics & Astronomy, University of Glasgow, Glasgow, G12 8QQ,
UK
| |
Collapse
|
18
|
Albumin uptake and distribution in the zebrafish liver as observed via correlative imaging. Exp Cell Res 2018; 374:162-171. [PMID: 30496757 DOI: 10.1016/j.yexcr.2018.11.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/20/2018] [Accepted: 11/23/2018] [Indexed: 12/20/2022]
Abstract
Although liver transport routes have been extensively studied in rodents, live imaging under in situ and in vivo conditions of large volumes is still proven to be difficult. In this study, we took advantage of the optical transparency of zebrafish and their small size to explore their usefulness for correlative imaging studies and liver transport experimentations. First, we assessed the micro-architecture of the zebrafish liver and compared its fine structure to the rodent and humans' literature. Next, we investigated the transport routes and cellular distribution of albumin using combined and correlative microscopy approaches. These methods permitted us to track the injected proteins at different time points through the process of liver uptake and clearance of albumin. We demonstrate strong structural and functional resemblance between the zebrafish liver and its rodents and humans' counterparts. In as short as 5 min post-injection, albumin rapidly accumulated within the LSECs. Furthermore, albumin entered the space of Disse where it initially accumulated then subsequently was taken up by the hepatocytes. We propose the zebrafish as a viable alternative experimental model for hepatic transport studies, allowing swift multimodal imaging and direct quantification on the hepatic distribution of supramolecular complexes of interest.
Collapse
|
19
|
Sieber S, Grossen P, Detampel P, Siegfried S, Witzigmann D, Huwyler J. Zebrafish as an early stage screening tool to study the systemic circulation of nanoparticulate drug delivery systems in vivo. J Control Release 2017; 264:180-191. [PMID: 28851572 DOI: 10.1016/j.jconrel.2017.08.023] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 08/20/2017] [Accepted: 08/21/2017] [Indexed: 12/14/2022]
Abstract
Nanomedicines have gained much attention for the delivery of small molecules or nucleic acids as treatment options for many diseases. However, the transfer from experimental systems to in vivo applications remains a challenge since it is difficult to assess their circulation behavior in the body at an early stage of drug discovery. Thus, innovative and improved concepts are urgently needed to overcome this issue and to close the gap between empiric nanoparticle design, in vitro assessment, and first in vivo experiments using rodent animal models. This study was focused on the zebrafish as a vertebrate screening model to assess the circulation in blood and extravasation behavior of nanoparticulate drug delivery systems in vivo. To validate this novel approach, monodisperse preparations of fluorescently labeled liposomes with similar size and zeta potential were injected into transgenic zebrafish lines expressing green fluorescent protein in their vasculature. Phosphatidylcholine-based lipids differed by fatty acid chain length and saturation. Circulation behavior and vascular distribution pattern were evaluated qualitatively and semi-quantitatively using image analysis. Liposomes composed of lipids with lower transition temperature (<28°C) as well as PEGylated liposomes showed longer circulation times and extravasation. In contrast, liposomes composed of lipids with transition temperatures>28°C bound to venous parts of the vasculature. This circulation patterns in the zebrafish model did correlate with published and experimental pharmacokinetic data from mice and rats. Our findings indicate that the zebrafish model is a useful vertebrate screening tool for nanoparticulate drug delivery systems to predict their in vivo circulation behavior with respect to systemic circulation time and exposure.
Collapse
Affiliation(s)
- Sandro Sieber
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Philip Grossen
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Pascal Detampel
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Salome Siegfried
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Dominik Witzigmann
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.
| |
Collapse
|
20
|
Mottaz H, Schönenberger R, Fischer S, Eggen RIL, Schirmer K, Groh KJ. Dose-dependent effects of morphine on lipopolysaccharide (LPS)-induced inflammation, and involvement of multixenobiotic resistance (MXR) transporters in LPS efflux in teleost fish. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2017; 221:105-115. [PMID: 28010888 DOI: 10.1016/j.envpol.2016.11.046] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 10/12/2016] [Accepted: 11/15/2016] [Indexed: 06/06/2023]
Abstract
Opioid drugs, such as morphine (MO), detected in aquatic environments worldwide, may harm fish due to their semi-persistence and ability to potently interact with molecular targets conserved across vertebrates. Here, we established a waterborne bacterial lipopolysaccharide (LPS) challenge assay with zebrafish embryos as a model to investigate chemically-induced disruption of the innate immune system, and used it to study the effects of MO exposure. Exposure to 1 mg/L MO resulted in pronounced immunosuppression, reflected in downregulation of several inflammation-related genes, including myd88, trif, traf6, p38, nfκb2, il-1β, il-8 and ccl34a. Fish exposed to 1 mg/L MO accumulated 11.7 ng/g (wet weight) of MO, a concentration comparable to that reported in blood of chronic drug abusers subject to higher infection rates. Surprisingly, exposure to lower MO concentrations (100 ng/L-100 μg/L) led to exacerbation of LPS-induced inflammation. Two ATP-binding cassette (ABC) transporters known to be involved in the xenobiotic efflux - abcb4 and abcc2, also known as multixenobiotic resistance (MXR) transporters - were downregulated at 100 ng/L MO. We hypothesized that ABC/MXR transporters could modulate the severity of inflammation by being involved in efflux of LPS, thus regulating its accumulation in the organism. Indeed, we could demonstrate that blocking of ABC/MXR transporters by an inhibitor, cyclosporine A, results in stronger inflammation, coinciding with higher LPS accumulation, as visualized with fluorescently labeled LPS. Our work demonstrates that MO can disrupt fish innate immune responses at environmentally relevant concentrations. We also provide evidence for a role of ABC/MXR transporters in LPS efflux in fish. These finding may be applicable across other taxa, as ABC transporters are evolutionary conserved. Since diverse environmentally present chemicals are known to interfere with ABC/MXR transporters' expression or activity, our discovery raises concerns about potential adverse effects of such compounds on the immune system responses in aquatic organisms.
Collapse
Affiliation(s)
- Hélène Mottaz
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, 8600 Dübendorf, Switzerland
| | - Rene Schönenberger
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, 8600 Dübendorf, Switzerland
| | - Stephan Fischer
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, 8600 Dübendorf, Switzerland
| | - Rik I L Eggen
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, 8600 Dübendorf, Switzerland; ETH Zürich, Department of Environmental Systems Science, 8092 Zürich, Switzerland
| | - Kristin Schirmer
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, 8600 Dübendorf, Switzerland; ETH Zürich, Department of Environmental Systems Science, 8092 Zürich, Switzerland; EPF Lausanne, School of Architecture, Civil and Environmental Engineering, 1015 Lausanne, Switzerland.
| | - Ksenia J Groh
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, 8600 Dübendorf, Switzerland
| |
Collapse
|
21
|
Henson HE, Parupalli C, Ju B, Taylor MR. Functional and genetic analysis of choroid plexus development in zebrafish. Front Neurosci 2014; 8:364. [PMID: 25426018 PMCID: PMC4226144 DOI: 10.3389/fnins.2014.00364] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 10/22/2014] [Indexed: 01/30/2023] Open
Abstract
The choroid plexus, an epithelial-based structure localized in the brain ventricle, is the major component of the blood-cerebrospinal fluid barrier. The choroid plexus produces the cerebrospinal fluid and regulates the components of the cerebrospinal fluid. Abnormal choroid plexus function is associated with neurodegenerative diseases, tumor formation in the choroid plexus epithelium, and hydrocephaly. In this study, we used zebrafish (Danio rerio) as a model system to understand the genetic components of choroid plexus development. We generated an enhancer trap line, Et(cp:EGFP)sj2, that expresses enhanced green fluorescent protein (EGFP) in the choroid plexus epithelium. Using immunohistochemistry and fluorescent tracers, we demonstrated that the zebrafish choroid plexus possesses brain barrier properties such as tight junctions and transporter activity. Thus, we have established zebrafish as a functionally relevant model to study choroid plexus development. Using an unbiased approach, we performed a forward genetic dissection of the choroid plexus to identify genes essential for its formation and function. Using Et(cp:EGFP)sj2, we isolated 10 recessive mutant lines with choroid plexus abnormalities, which were grouped into five classes based on GFP intensity, epithelial localization, and overall choroid plexus morphology. We also mapped the mutation for two mutant lines to chromosomes 4 and 21, respectively. The mutants generated in this study can be used to elucidate specific genes and signaling pathways essential for choroid plexus development, function, and/or maintenance and will provide important insights into how these genetic mutations contribute to disease.
Collapse
Affiliation(s)
- Hannah E Henson
- Chemical Biology and Therapeutics, St. Jude Children's Research Hospital Memphis, TN, USA ; Integrated Program in Biomedical Sciences, College of Graduate Health Sciences, University of Tennessee Health Science Center Memphis, TN, USA
| | | | - Bensheng Ju
- Chemical Biology and Therapeutics, St. Jude Children's Research Hospital Memphis, TN, USA
| | - Michael R Taylor
- Chemical Biology and Therapeutics, St. Jude Children's Research Hospital Memphis, TN, USA ; Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison Madison, WI, USA
| |
Collapse
|