1
|
Hong T, Park J, An G, Song J, Song G, Lim W. Evaluation of organ developmental toxicity of environmental toxicants using zebrafish embryos. Mol Cells 2024; 47:100144. [PMID: 39489379 PMCID: PMC11635654 DOI: 10.1016/j.mocell.2024.100144] [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: 08/08/2024] [Revised: 10/04/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024] Open
Abstract
There is increasing global concern about environmental pollutants, such as heavy metals, plastics, pharmaceuticals, personal care products, and pesticides, which have been detected in a variety of environments and are likely to be exposed to nontarget organisms, including humans. Various animal models have been utilized for toxicity assessment, and zebrafish are particularly valuable for studying the toxicity of various compounds owing to their similarity to other aquatic organisms and 70% genetic similarity to humans. Their development is easy to observe, and transgenic models for organs such as the heart, liver, blood vessels, and nervous system enable efficient studies of organ-specific toxicity. This suggests that zebrafish are a valuable tool for evaluating toxicity in specific organs and forecasting the potential impacts on other nontarget species. This review describes organ toxicity caused by various toxic substances and their mechanisms in zebrafish.
Collapse
Affiliation(s)
- Taeyeon Hong
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Junho Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Garam An
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jisoo Song
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| | - Whasun Lim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
2
|
Yin J, Forn-Cuní G, Surendran AM, Lopes-Bastos B, Pouliopoulou N, Jager MJ, Le Dévédec SE, Chen Q, Snaar-Jagalska BE. Lactate secreted by glycolytic conjunctival melanoma cells attracts and polarizes macrophages to drive angiogenesis in zebrafish xenografts. Angiogenesis 2024; 27:703-717. [PMID: 38842752 PMCID: PMC11564320 DOI: 10.1007/s10456-024-09930-y] [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: 01/15/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024]
Abstract
Conjunctival melanoma (CoM) is a rare but potentially lethal cancer of the eye, with limited therapeutic option for metastases. A better understanding how primary CoM disseminate to form metastases is urgently needed in order to develop novel therapies. Previous studies indicated that primary CoM tumors express Vascular Endothelial Growth Factor (VEGF) and may recruit pro-tumorigenic M2-like macrophages. However, due to a lack of proper models, the expected role of angiogenesis in the metastatic dissemination of CoM is still unknown. We show that cells derived from two CoM cell lines induce a strong angiogenic response when xenografted in zebrafish larvae. CoM cells are highly glycolytic and secrete lactate, which recruits and polarizes human and zebrafish macrophages towards a M2-like phenotype. These macrophages elevate the levels of proangiogenic factors such as VEGF, TGF-β, and IL-10 in the tumor microenvironment to induce an angiogenic response towards the engrafted CoM cells in vivo. Chemical ablation of zebrafish macrophages or inhibition of glycolysis in CoM cells terminates this response, suggesting that attraction of lactate-dependent macrophages into engrafted CoM cells drives angiogenesis and serves as a possible dissemination mechanism for glycolytic CoM cells.
Collapse
Affiliation(s)
- Jie Yin
- Institute of Biology, Leiden University, Leiden, 2333 BE, The Netherlands
| | - Gabriel Forn-Cuní
- Institute of Biology, Leiden University, Leiden, 2333 BE, The Netherlands
| | | | - Bruno Lopes-Bastos
- Institute of Biology, Leiden University, Leiden, 2333 BE, The Netherlands
| | - Niki Pouliopoulou
- Institute of Biology, Leiden University, Leiden, 2333 BE, The Netherlands
| | - Martine J Jager
- Department of Ophthalmology, Leiden University Medical Center, Leiden, 2333 ZA, the Netherlands
| | - Sylvia E Le Dévédec
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, 2333 BE, The Netherlands
| | - Quanchi Chen
- Institute of Biology, Leiden University, Leiden, 2333 BE, The Netherlands.
- Division of Spine Surgery, Department of Orthopedic Surgery, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210008, China.
| | | |
Collapse
|
3
|
Vega-Granados K, Escobar-Ibarra P, Palomino-Vizcaino K, Cruz-Reyes J, Valverde-Guillén P, Latorre-Redoli S, Caneda-Santiago CT, Marí-Beffa M, Romero-Sánchez LB. Hexyltrimethylammonium ion enhances potential copper-chelating properties of ammonium thiomolybdate in an in vivo zebrafish model. Arch Biochem Biophys 2024; 758:110077. [PMID: 38942109 DOI: 10.1016/j.abb.2024.110077] [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: 12/12/2023] [Revised: 06/18/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
Ammonium and hexyltrimethylammonium thiomolybdates (ATM and ATM-C6) and thiotungstates (ATT and ATT-C6) were synthesized. Their toxicity was evaluated using both in vitro and in vivo approaches via the zebrafish embryo acute toxicity assay (ZFET), while the copper-thiometallate interaction was studied using cyclic voltammetry, as well as in an in vivo assay. Cyclic voltammetry suggests that all thiometallates form complexes with copper in a 2:1 Cu:thiometallate ratio. Both in vitro and in vivo assays demonstrated low toxicity in BALB/3T3 cells and in zebrafish embryos, with high IC50 and LC50 values. Furthermore, the hexyltrimethylammonium ion played a crucial role in enhancing viability and reducing toxicity during prolonged treatments for ATM and ATT. In particular, the ZEFT assay uncovered the accumulation of ATM in zebrafish yolk, averted by the incorporation of the hexyltrimethylammonium ion. Notably, the copper-thiometallate interaction assay highlighted the improved viability of embryos when cultured in CuCl2 and ATM-C6, even at high CuCl2 concentrations. The hatching assay further confirmed that copper-ATM-C6 interaction mitigates inhibitory effects induced by thiomolybdates and CuCl2 when administered individually. These results suggest that the incorporation of the hexyltrimethylammonium ion in ATM increase its ability to interact with copper and its potential application as a copper chelator.
Collapse
Affiliation(s)
- K Vega-Granados
- Faculty of Chemical Sciences and Engineering, Autonomous University of Baja California, Tijuana, 22390, Mexico
| | - P Escobar-Ibarra
- Faculty of Chemical Sciences and Engineering, Autonomous University of Baja California, Tijuana, 22390, Mexico
| | - K Palomino-Vizcaino
- Faculty of Chemical Sciences and Engineering, Autonomous University of Baja California, Tijuana, 22390, Mexico
| | - J Cruz-Reyes
- Faculty of Chemical Sciences and Engineering, Autonomous University of Baja California, Tijuana, 22390, Mexico
| | - P Valverde-Guillén
- Department of Cell Biology, Genetics and Physiology, Faculty of Science, University of Malaga, Malaga, Spain
| | - S Latorre-Redoli
- Department of Cell Biology, Genetics and Physiology, Faculty of Science, University of Malaga, Malaga, Spain
| | - C T Caneda-Santiago
- Department of Cell Biology, Genetics and Physiology, Faculty of Science, University of Malaga, Malaga, Spain
| | - M Marí-Beffa
- Department of Cell Biology, Genetics and Physiology, Faculty of Science, University of Malaga, Malaga, Spain; Andalusian Centre for Nanomedicine and Biotechnology (IMABIS-BIONAND), Andalusian Institute of Blue Biotechnology and Development (IBYDA), Experimental Centre Grice Hutchinson, University of Malaga, Malaga, Spain
| | - L B Romero-Sánchez
- Faculty of Chemical Sciences and Engineering, Autonomous University of Baja California, Tijuana, 22390, Mexico.
| |
Collapse
|
4
|
Zhang Y, Zhang J, Fan H, Lu R, Nie G. Database construction and comparative genomics analysis of genes involved in nutritional metabolic diseases in fish. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 50:101241. [PMID: 38733902 DOI: 10.1016/j.cbd.2024.101241] [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: 03/17/2024] [Revised: 04/24/2024] [Accepted: 05/04/2024] [Indexed: 05/13/2024]
Abstract
Nutritional metabolic diseases in fish frequently arise in the setting of intensive aquaculture. The etiology and pathogenesis of these conditions involve energy metabolic disorders influenced by both internal genetic factors and external environmental conditions. The exploration of genes associated with nutritional and metabolic disorder has sparked considerable interest within both the aquaculture scientific community and the industry. High-throughput sequencing technology offers researchers extensive genetic information. Effectively mining, analyzing, and securely storing this data is crucial, especially for advancing disease prevention and treatment strategies. Presently, the exploration and application of gene databases concerning nutritional and metabolic disorders in fish are at a nascent stag. Therefore, this study focused on the model organism zebrafish and five primary economic fish species as the subjects of investigation. Using information from KEGG, OMIM, and existing literature, a novel gene database associated with nutritional metabolic diseases in fish was meticulously constructed. This database encompassed 4583 genes for Danio rerio, 6287 for Cyprinus carpio, 3289 for Takifugu rubripes, 3548 for Larimichthys crocea, 3816 for Oreochromis niloticus, and 5708 for Oncorhynchus mykiss. Through a comparative systems biology approach, we discerned a relatively high conservation of genes linked to nutritional metabolic diseases across these fish species, with over 54.9 % of genes being conserved throughout all six species. Additionally, the analysis pinpointed the existence of 13 species-specific genes within the genomes of large yellow croaker, tilapia, and rainbow trout. These genes exhibit the potential to serve as novel candidate targets for addressing nutritional metabolic diseases.
Collapse
Affiliation(s)
- Yuru Zhang
- College of Fisheries, Henan Normal University, Xinxiang 453007, PR China; College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, PR China
| | - Junmei Zhang
- College of Fisheries, Henan Normal University, Xinxiang 453007, PR China; College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, PR China
| | - Haiying Fan
- College of Fisheries, Henan Normal University, Xinxiang 453007, PR China; College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, PR China
| | - Ronghua Lu
- College of Fisheries, Henan Normal University, Xinxiang 453007, PR China; College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, PR China
| | - Guoxing Nie
- College of Fisheries, Henan Normal University, Xinxiang 453007, PR China; College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, PR China.
| |
Collapse
|
5
|
Brix A, Belleri L, Pezzotta A, Pettinato E, Mazzola M, Zoccolillo M, Marozzi A, Monteiro R, Del Bene F, Mortellaro A, Pistocchi A. ADA2 regulates inflammation and hematopoietic stem cell emergence via the A 2bR pathway in zebrafish. Commun Biol 2024; 7:615. [PMID: 38777862 PMCID: PMC11111730 DOI: 10.1038/s42003-024-06286-3] [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: 09/28/2023] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Deficiency of adenosine deaminase 2 (DADA2) is an inborn error of immunity caused by loss-of-function mutations in the adenosine deaminase 2 (ADA2) gene. Clinical manifestations of DADA2 include vasculopathy and immuno-hematological abnormalities, culminating in bone marrow failure. A major gap exists in our knowledge of the regulatory functions of ADA2 during inflammation and hematopoiesis, mainly due to the absence of an ADA2 orthologue in rodents. Exploring these mechanisms is essential for understanding disease pathology and developing new treatments. Zebrafish possess two ADA2 orthologues, cecr1a and cecr1b, with the latter showing functional conservation with human ADA2. We establish a cecr1b-loss-of-function zebrafish model that recapitulates the immuno-hematological and vascular manifestations observed in humans. Loss of Cecr1b disrupts hematopoietic stem cell specification, resulting in defective hematopoiesis. This defect is caused by induced inflammation in the vascular endothelium. Blocking inflammation, pharmacological modulation of the A2r pathway, or the administration of the recombinant human ADA2 corrects these defects, providing insights into the mechanistic link between ADA2 deficiency, inflammation and immuno-hematological abnormalities. Our findings open up potential therapeutic avenues for DADA2 patients.
Collapse
Affiliation(s)
- Alessia Brix
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, L.I.T.A., via Fratelli Cervi 93, Segrate, 20054, Milan, Italy
| | - Laura Belleri
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, L.I.T.A., via Fratelli Cervi 93, Segrate, 20054, Milan, Italy
- Department of Development, Institut de la Vision, 17 Rue Moreau, 75012, Paris, France
| | - Alex Pezzotta
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, L.I.T.A., via Fratelli Cervi 93, Segrate, 20054, Milan, Italy
| | - Emanuela Pettinato
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy
| | - Mara Mazzola
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, L.I.T.A., via Fratelli Cervi 93, Segrate, 20054, Milan, Italy
| | - Matteo Zoccolillo
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy
| | - Anna Marozzi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, L.I.T.A., via Fratelli Cervi 93, Segrate, 20054, Milan, Italy
| | - Rui Monteiro
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, Edgbaston, B15 2TTB, UK
| | - Filippo Del Bene
- Department of Development, Institut de la Vision, 17 Rue Moreau, 75012, Paris, France
| | - Alessandra Mortellaro
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy.
| | - Anna Pistocchi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, L.I.T.A., via Fratelli Cervi 93, Segrate, 20054, Milan, Italy.
| |
Collapse
|
6
|
Yuan J, Que R, Zhao W, Song F, Cao Y, Yu B. Influences of lysine-specific demethylase 1 inhibitors on NO synthase-Kruppel-like factor pathways in human endothelial cells in vitro and zebrafish (Danio rerio) larvae in vivo. J Appl Toxicol 2023; 43:1748-1760. [PMID: 37408164 DOI: 10.1002/jat.4512] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/02/2023] [Accepted: 06/17/2023] [Indexed: 07/07/2023]
Abstract
Lysine-specific demethylase 1 (LSD1) inhibitors are being developed for cancer therapy, but their bioeffects on vasculatures are not clear. In this study, we compared the influences of ORY-1001 (an LSD1 inhibitor being advanced into clinical trials) and 199 (a novel LSD1 inhibitor recently developed by us) to human umbilical vein endothelial cells (HUVECs) in vitro and further verified the bioeffects of ORY-1001 to zebrafish (Danio rerio) larvae in vivo. The results showed that up to 10 μM ORY-1001 or 199 did not significantly affect the cellular viability of HUVECs but substantially reduced the release of inflammatory interleukin-8 (IL-8) and IL-6. The signaling molecule in vasculatures, NO, was also increased in HUVECs. As the mechanism, the protein levels of endothelial NO synthase (eNOS) or p-eNOS, and their regulators Kruppel-like factor 2 (KLF2) or KLF4, were also increased after drug treatment. In vivo, 24 h treatment with up to 100 nM ORY-1001 reduced blood speed without changing morphologies or locomotor activities in zebrafish larvae. ORY-1001 treatment reduced the expression of il8 but promoted the expression of klf2a and nos in the zebrafish model. These data show that LSD1 inhibitors were not toxic but capable to inhibit inflammatory responses and affect the function of blood vessels through the up-regulation of the NOS-KLF pathway.
Collapse
Affiliation(s)
- Jialin Yuan
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Ruiman Que
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Weichao Zhao
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Fengmei Song
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Yi Cao
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Bin Yu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
7
|
Angom RS, Wang Y, Wang E, Dutta S, Mukhopadhyay D. Conditional, Tissue-Specific CRISPR/Cas9 Vector System in Zebrafish Reveals the Role of Nrp1b in Heart Regeneration. Arterioscler Thromb Vasc Biol 2023; 43:1921-1934. [PMID: 37650323 PMCID: PMC10771629 DOI: 10.1161/atvbaha.123.319189] [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: 02/22/2023] [Accepted: 08/04/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat-associated 9) technology-mediated genome editing has significantly improved the targeted inactivation of genes in vitro and in vivo in many organisms. Neuropilins play crucial roles in zebrafish heart regeneration, heart failure in mice, and electrical remodeling after myocardial infarction in rats. But the cell-specific functions of nrp1 have not been described before. In this study, we have investigated the role of nrp1 isoforms, including nrp1a and nrp1b, in cardiomyocytes during cardiac injury and regeneration in adult zebrafish hearts. METHODS In this study, we have reported a novel CRISPR-based vector system for conditional tissue-specific gene ablation in zebrafish. Specifically, the cardiac-specific cmlc2 promoter drives Cas9 expression to silence the nrp1 gene in cardiomyocytes in a heat-shock inducible manner. This vector system establishes a unique tool to regulate the gene knockout in both the developmental and adult stages and hence widens the possibility of loss-of-function studies in zebrafish at different stages of development and adulthood. Using this approach, we investigated the role of neuropilin isoforms nrp1a and nrp1b in response to cardiac injury and regeneration in adult zebrafish hearts. RESULTS We observed that both the isoforms (nrp1a and nrp1b) are upregulated after the cryoinjury. Interestingly, the nrp1b knockout significantly delayed heart regeneration and impaired cardiac function in the adult zebrafish after cryoinjury, demonstrated by reduced heart rate, ejection fractions, and fractional shortening. In addition, we show that the knockdown of nrp1b but not nrp1a induces activation of the cardiac remodeling genes in response to cryoinjury. CONCLUSIONS To our knowledge, this study is novel where we have reported a heat-shock-mediated conditional knockdown of nrp1a and nrp1b isoforms using CRISPR/Cas9 technology in the cardiomyocyte in zebrafish and furthermore have identified a crucial role for the nrp1b isoform in zebrafish cardiac remodeling and eventually heart function in response to injury.
Collapse
Affiliation(s)
- Ramcharan Singh Angom
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, FL 32224
| | - Ying Wang
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Rochester, MN 55905
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, FL 32224
| | - Shamit Dutta
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, FL 32224
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, FL 32224
| |
Collapse
|
8
|
Wu X, Hua X, Xu K, Song Y, Lv T. Zebrafish in Lung Cancer Research. Cancers (Basel) 2023; 15:4721. [PMID: 37835415 PMCID: PMC10571557 DOI: 10.3390/cancers15194721] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Zebrafish is increasingly used as a model organism for cancer research because of its genetic and physiological similarities to humans. Modeling lung cancer (LC) in zebrafish has received significant attention. This review focuses on the insights gained from using zebrafish in LC research. These insights range from investigating the genetic and molecular mechanisms that contribute to the development and progression of LC to identifying potential drug targets, testing the efficacy and toxicity of new therapies, and applying zebrafish for personalized medicine studies. This review provides a comprehensive overview of the current state of LC research performed using zebrafish, highlights the advantages and limitations of this model organism, and discusses future directions in the field.
Collapse
Affiliation(s)
- Xiaodi Wu
- Department of Clinical Medicine, Medical School of Nanjing University, Nanjing 210093, China; (X.W.); (K.X.)
| | - Xin Hua
- Department of Clinical Medicine, Southeast University Medical College, Nanjing 210096, China;
| | - Ke Xu
- Department of Clinical Medicine, Medical School of Nanjing University, Nanjing 210093, China; (X.W.); (K.X.)
| | - Yong Song
- Department of Clinical Medicine, Southeast University Medical College, Nanjing 210096, China;
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Tangfeng Lv
- Department of Clinical Medicine, Medical School of Nanjing University, Nanjing 210093, China; (X.W.); (K.X.)
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| |
Collapse
|
9
|
García-Márquez J, Moreira BR, Valverde-Guillén P, Latorre-Redoli S, Caneda-Santiago CT, Acién G, Martínez-Manzanares E, Marí-Beffa M, Abdala-Díaz RT. In Vitro and In Vivo Effects of Ulvan Polysaccharides from Ulva rigida. Pharmaceuticals (Basel) 2023; 16:ph16050660. [PMID: 37242444 DOI: 10.3390/ph16050660] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/22/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
One of the main bioactive compounds of interest from the Ulva species is the sulfated polysaccharide ulvan, which has recently attracted attention for its anticancer properties. This study investigated the cytotoxic activity of ulvan polysaccharides obtained from Ulva rigida in the following scenarios: (i) in vitro against healthy and carcinogenic cell lines (1064sk (human fibroblasts), HACAT (immortalized human keratinocytes), U-937 (a human leukemia cell line), G-361 (a human malignant melanoma), and HCT-116 (a colon cancer cell line)) and (ii) in vivo against zebrafish embryos. Ulvan exhibited cytotoxic effects on the three human cancer cell lines tested. However, only HCT-116 demonstrated sufficient sensitivity to this ulvan to make it relevant as a potential anticancer treatment, presenting an LC50 of 0.1 mg mL-1. The in vivo assay on the zebrafish embryos showed a linear relationship between the polysaccharide concentration and growth retardation at 7.8 hpf mL mg-1, with an LC50 of about 5.2 mg mL-1 at 48 hpf. At concentrations near the LC50, toxic effects, such as pericardial edema or chorion lysis, could be found in the experimental larvae. Our in vitro study supports the potential use of polysaccharides extracted from U. rigida as candidates for treating human colon cancer. However, the in vivo assay on zebrafish indicated that the potential use of ulvan as a promising, safe compound should be limited to specific concentrations below 0.001 mg mL-1 since it revealed side effects on the embryonic growth rate and osmolar balance.
Collapse
Affiliation(s)
- Jorge García-Márquez
- Department of Microbiology, Faculty of Science, Andalusian Institute of Blue Biotechnology and Development (IBYDA), Malaga University, Campus Universitario de Teatinos s/n, 29071 Malaga, Spain
| | - Bruna Rodrigues Moreira
- Phycology Laboratory, Department of Botany, Biological Sciences Center, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Piedad Valverde-Guillén
- Department of Cell Biology, Genetics and Physiology, Faculty of Science, Andalusian Institute of Blue Biotechnology and Development (IBYDA), Malaga University, Campus Universitario de Teatinos s/n, 29071 Malaga, Spain
| | - Sofía Latorre-Redoli
- Department of Cell Biology, Genetics and Physiology, Faculty of Science, Andalusian Institute of Blue Biotechnology and Development (IBYDA), Malaga University, Campus Universitario de Teatinos s/n, 29071 Malaga, Spain
| | - Candela T Caneda-Santiago
- Department of Cell Biology, Genetics and Physiology, Faculty of Science, Andalusian Institute of Blue Biotechnology and Development (IBYDA), Malaga University, Campus Universitario de Teatinos s/n, 29071 Malaga, Spain
| | - Gabriel Acién
- Department of Chemical Engineering, Almería University, 04120 Almería, Spain
| | - Eduardo Martínez-Manzanares
- Department of Microbiology, Faculty of Science, Andalusian Institute of Blue Biotechnology and Development (IBYDA), Malaga University, Campus Universitario de Teatinos s/n, 29071 Malaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, 29071 Málaga, Spain
| | - Manuel Marí-Beffa
- Department of Cell Biology, Genetics and Physiology, Faculty of Science, Andalusian Institute of Blue Biotechnology and Development (IBYDA), Malaga University, Campus Universitario de Teatinos s/n, 29071 Malaga, Spain
- Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Málaga Biomedical Research Institute and Nanomedicine Platform (IBIMA BIONAND Platform), 29071 Málaga, Spain
| | - Roberto T Abdala-Díaz
- Department of Ecology and Geology, Faculty of Science, Andalusian Institute of Blue Biotechnology and Development (IBYDA), Malaga University, Campus Universitario de Teatinos s/n, 29071 Malaga, Spain
| |
Collapse
|
10
|
Vedder VL, Reinberger T, Haider SMI, Eichelmann L, Odenthal N, Abdelilah-Seyfried S, Aherrahrou Z, Breuer M, Erdmann J. pyHeart4Fish: Chamber-specific heart phenotype quantification of zebrafish in high-content screens. Front Cell Dev Biol 2023; 11:1143852. [PMID: 37113769 PMCID: PMC10126419 DOI: 10.3389/fcell.2023.1143852] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death. Of CVDs, congenital heart diseases are the most common congenital defects, with a prevalence of 1 in 100 live births. Despite the widespread knowledge that prenatal and postnatal drug exposure can lead to congenital abnormalities, the developmental toxicity of many FDA-approved drugs is rarely investigated. Therefore, to improve our understanding of drug side effects, we performed a high-content drug screen of 1,280 compounds using zebrafish as a model for cardiovascular analyses. Zebrafish are a well-established model for CVDs and developmental toxicity. However, flexible open-access tools to quantify cardiac phenotypes are lacking. Here, we provide pyHeart4Fish, a novel Python-based, platform-independent tool with a graphical user interface for automated quantification of cardiac chamber-specific parameters, such as heart rate (HR), contractility, arrhythmia score, and conduction score. In our study, about 10.5% of the tested drugs significantly affected HR at a concentration of 20 µM in zebrafish embryos at 2 days post-fertilization. Further, we provide insights into the effects of 13 compounds on the developing embryo, including the teratogenic effects of the steroid pregnenolone. In addition, analysis with pyHeart4Fish revealed multiple contractility defects induced by seven compounds. We also found implications for arrhythmias, such as atrioventricular block caused by chloropyramine HCl, as well as (R)-duloxetine HCl-induced atrial flutter. Taken together, our study presents a novel open-access tool for heart analysis and new data on potentially cardiotoxic compounds.
Collapse
Affiliation(s)
- Viviana L. Vedder
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
- University Heart Centre Lübeck, Lübeck, Germany
| | - Tobias Reinberger
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
- University Heart Centre Lübeck, Lübeck, Germany
| | - Syed M. I. Haider
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
- University Heart Centre Lübeck, Lübeck, Germany
| | - Luis Eichelmann
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
- University Heart Centre Lübeck, Lübeck, Germany
| | - Nadine Odenthal
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
- University Heart Centre Lübeck, Lübeck, Germany
| | - Salim Abdelilah-Seyfried
- Faculty of Mathematics and Natural Sciences, Institute for Biochemistry and Biology, University Potsdam, Potsdam, Germany
| | - Zouhair Aherrahrou
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
- University Heart Centre Lübeck, Lübeck, Germany
| | - Maximilian Breuer
- Faculty of Mathematics and Natural Sciences, Institute for Biochemistry and Biology, University Potsdam, Potsdam, Germany
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
- University Heart Centre Lübeck, Lübeck, Germany
| |
Collapse
|
11
|
Caruso G, Scalisi EM, Pecoraro R, Cardaci V, Privitera A, Truglio E, Capparucci F, Jarosova R, Salvaggio A, Caraci F, Brundo MV. Effects of carnosine on the embryonic development and TiO 2 nanoparticles-induced oxidative stress on Zebrafish. Front Vet Sci 2023; 10:1148766. [PMID: 37035814 PMCID: PMC10078361 DOI: 10.3389/fvets.2023.1148766] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Abstract
Oxidative stress is due to an unbalance between pro-oxidants, such as reactive oxygen (ROS) and nitrogen (RNS) species, and antioxidants/antioxidant system. Under physiological conditions these species are involved in different cellular processes such as cellular homeostasis and immune response, while an excessive production of ROS/RNS has been linked to the development of various diseases such as cancer, diabetes, and Alzheimer's disease. In this context, the naturally occurring dipeptide carnosine has shown the ability to scavenge ROS, counteract lipid peroxidation, and inhibit proteins oxidation. Titanium dioxide nanoparticles (TiO2-NPs) have been widely used to produce cosmetics, in wastewater treatment, in food industry, and in healthcare product. As consequence, these NPs are often released into aquatic environments. The Danio rerio (commonly called zebrafish) embryos exposure to TiO2-NPs did not affect the hatching rate, but induced oxidative stress. According to this scenario, in the present study, we first investigated the effects of carnosine exposure and of a sub-toxic administration of TiO2-NPs on the development and survival of zebrafish embryos/larvae measured through the acute embryo toxicity test (FET-Test). Zebrafish larvae represent a useful model to study oxidative stress-linked disorders and to test antioxidant molecules, while carnosine was selected based on its well-known multimodal mechanism of action that includes a strong antioxidant activity. Once the basal effects of carnosine were assessed, we then evaluated its effects on TiO2-NPs-induced oxidative stress in zebrafish larvae, measured in terms of total ROS production (measured with 2,7-dichlorodihydrofluorescein diacetate probe) and protein expression by immunohistochemistry of two cellular stress markers, 70 kDa-heat shock protein (Hsp70) and metallothioneins (MTs). We demonstrated that carnosine did not alter the phenotypes of both embryos and larvae of zebrafish at different hours post fertilization. Carnosine was instead able to significantly decrease the enhancement of ROS levels in zebrafish larvae exposed to TiO2-NPs and its antioxidant effect was paralleled by the rescue of the protein expression levels of Hsp70 and MTs. Our results suggest a therapeutic potential of carnosine as a new pharmacological tool in the context of pathologies characterized by oxidative stress such as neurodegenerative disorders.
Collapse
Affiliation(s)
- Giuseppe Caruso
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
- Unit of Neuropharmacology and Translational Neurosciences, Oasi Research Institute-IRCCS, Troina, Italy
| | - Elena Maria Scalisi
- Department of Biological, Geological and Environmental Sciences, University of Catania, Catania, Italy
| | - Roberta Pecoraro
- Department of Biological, Geological and Environmental Sciences, University of Catania, Catania, Italy
| | - Vincenzo Cardaci
- Vita-Salute San Raffaele University, Milan, Italy
- Scuola Superiore di Catania, University of Catania, Catania, Italy
| | - Anna Privitera
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Emanuela Truglio
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Fabiano Capparucci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Romana Jarosova
- Department of Chemistry and R.N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, KS, United States
| | | | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
- Unit of Neuropharmacology and Translational Neurosciences, Oasi Research Institute-IRCCS, Troina, Italy
| | - Maria Violetta Brundo
- Department of Biological, Geological and Environmental Sciences, University of Catania, Catania, Italy
| |
Collapse
|
12
|
Qin JY, Jia W, Ru S, Xiong JQ, Wang J, Wang W, Hao L, Zhang X. Bisphenols induce cardiotoxicity in zebrafish embryos: Role of the thyroid hormone receptor pathway. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 254:106354. [PMID: 36423468 DOI: 10.1016/j.aquatox.2022.106354] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 09/21/2022] [Accepted: 11/11/2022] [Indexed: 06/16/2023]
Abstract
Bisphenols are frequently found in the environment and have been of emerging concern because of their adverse effects on aquatic animals and humans. In this study, we demonstrated that bisphenol A, S, and F (BPA, BPS, BPF) at environmental concentrations induced cardiotoxicity in zebrafish embryos. BPA decreased heart rate at 96 hpf (hours post fertilization) and increased the distance between the sinus venosus (SV) and bulbus arteriosus (BA), in zebrafish. BPF promoted heart pumping and stroke volume, shortened the SV-BAdistance, and increased body weight. Furthermore, we found that BPA increased the expression of the dio3b, thrβ, and myh7 genes but decreased the transcription of dio2. In contrast, BPF downregulated the expression of myh7 but upregulated that of thrβ. Molecular docking results showed that both BPA and BPF are predicted to bind tightly to the active pockets of zebrafish THRβ with affinities of -4.7 and -4.77 kcal/mol, respectively. However, BPS did not significantly affect dio3b, thrβ, and myh7 transcription and had a higher affinity for zebrafish THRβ (-2.13 kcal/mol). These findings suggest that although BPA, BPS, and BPF have similar structures, they may induce cardiotoxicity through different molecular mechanisms involving thyroid hormone systems. This investigation provides novel insights into the potential mechanism of cardiotoxicity from the perspective of thyroid disruption and offer a cautionary role for the use of BPA substitution.
Collapse
Affiliation(s)
- Jing-Yu Qin
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Wenyi Jia
- College of urban and environmental sciences, Peking University, Beijing 100871, China
| | - Shaoguo Ru
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Jiu-Qiang Xiong
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Jun Wang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Weiwei Wang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Liping Hao
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Xiaona Zhang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China.
| |
Collapse
|
13
|
Hong T, Park H, An G, Song G, Lim W. Ethalfluralin induces developmental toxicity in zebrafish via oxidative stress and inflammation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 854:158780. [PMID: 36115403 DOI: 10.1016/j.scitotenv.2022.158780] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/10/2022] [Accepted: 09/11/2022] [Indexed: 06/15/2023]
Abstract
Ethalfluralin, of dinitroaniline herbicide family, is an effective weed controller. Following residue detection in herbicide-treated fields, ethalfluralin was reported to interfere with early stages of implantation in some vertebrate species. However, the role of ethalfluralin in the development of zebrafish embryos has not been elucidated yet. Therefore, in the present study, we investigated the morphological and physiological changes that occur in the embryonic development of zebrafish due to ethalfluralin exposure. Results indicated that ethalfluralin decreased survival rate along with reduction in the hatching ratio and heartbeat. It was observed to cause edema in the heart and yolk sac, and apoptosis in the anterior region of the developing zebrafish larvae; as visualized through acridine orange and TUNEL staining. In addition, ethalfluralin increased the expression of the apoptosis-associated genes including tp53, cyc1, casp8, casp9, and casp3. The Seahorse Mito Stress analysis revealed that ethalfluralin slightly reduced mitochondrial respiration in live zebrafish embryos. Reactive oxygen species (ROS) production was also observed to be elevated in zebrafish larvae in response to ethalfluralin. Treatment with ethalfluralin decreased blood vessel formation in brain and intestine in flk1 transgenic zebrafish embryos. The decrease in angiogenesis related gene expression was specifically observed in vegfc, flt1, and kdrl, and in the intestinal vasculature related genes apoa4a, aqp3, fabp2, and vil1. Moreover, an increase in inflammatory genes such as cox2a, cox2b, cxcl-c1c, il8, mcl1a, mcl1b, and nf-κb was observed using real-time PCR analysis. Collectively, these results indicate that oxidative stress generated by exposure to ethalfluralin induced ROS generation, apoptosis, inflammation and anti-angiogenic effects, and therefore, ethalfluralin may be toxic to the development of zebrafish embryos.
Collapse
Affiliation(s)
- Taeyeon Hong
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hahyun Park
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Garam An
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| | - Whasun Lim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
14
|
Wang M, Liu J, Wang H, Hu T. Spiromesifen contributes vascular developmental toxicity via disrupting endothelial cell proliferation and migration in zebrafish embryos. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2022; 188:105242. [PMID: 36464354 DOI: 10.1016/j.pestbp.2022.105242] [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: 07/28/2022] [Revised: 09/03/2022] [Accepted: 09/13/2022] [Indexed: 06/17/2023]
Abstract
Spiromesifen (SPF) is a specific contact pesticide, which has been widely used to control the growth of sucking insects like mites and whiteflies on crops. Although its residues in crops and effects on organisms has been extensively reported, its impact on the vasculature is still not being reported. In the present study, using human umbilical vein endothelial cells (HUVECs) and zebrafish embryos, we investigated the effects of SPF on blood vessel development and its mechanism of action. SPF exposure triggered abnormal blood vessel development, including vascular deletions and malformations, inhibition of CCV remodeling, and decrease of SIV areas. SPF exposure also obstructed the migration of endothelial cell from caudal hematopoietic tissue in zebrafish embryos. SPF damaged cytoskeleton, caused cell cycle arrest, inhibited the viability and migration of HUVECs. In addition, SPF also inhibited the expression of the VEGF/VEGFR pathway-related genes (hif1a, vegfa, flt1, and kdrl), cell cycle-related genes (ccnd1, ccne1, cdk2, and pcna), and Rho/ROCK pathway-related genes (itgb1, rho, rock, mlc-1, and vim-1). Taken together, SPF may inhibit the proliferation and migration of vascular endothelial cells through disturbing cytoskeleton via the Rho/ ROCK pathway, resulting in vascular malformation. Our study contributes to potential insight into the mechanism of SPF toxicity in angiocardiopathy.
Collapse
Affiliation(s)
- Mingxing Wang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Juan Liu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Huiyun Wang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Tingzhang Hu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China.
| |
Collapse
|
15
|
Macklin BL, Lin YY, Emmerich K, Wisniewski E, Polster BM, Konstantopoulos K, Mumm JS, Gerecht S. Intrinsic epigenetic control of angiogenesis in induced pluripotent stem cell-derived endothelium regulates vascular regeneration. NPJ Regen Med 2022; 7:28. [PMID: 35551465 PMCID: PMC9098630 DOI: 10.1038/s41536-022-00223-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 04/14/2022] [Indexed: 11/17/2022] Open
Abstract
Human-induced pluripotent stem cell-derived endothelial cells (iECs) provide opportunities to study vascular development and regeneration, develop cardiovascular therapeutics, and engineer model systems for drug screening. The differentiation and characterization of iECs are well established; however, the mechanisms governing their angiogenic phenotype remain unknown. Here, we aimed to determine the angiogenic phenotype of iECs and the regulatory mechanism controlling their regenerative capacity. In a comparative study with HUVECs, we show that iECs increased expression of vascular endothelial growth factor receptor 2 (VEGFR2) mediates their highly angiogenic phenotype via regulation of glycolysis enzymes, filopodia formation, VEGF mediated migration, and robust sprouting. We find that the elevated expression of VEGFR2 is epigenetically regulated via intrinsic acetylation of histone 3 at lysine 27 by histone acetyltransferase P300. Utilizing a zebrafish xenograft model, we demonstrate that the ability of iECs to promote the regeneration of the amputated fin can be modulated by P300 activity. These findings demonstrate how the innate epigenetic status of iECs regulates their phenotype with implications for their therapeutic potential.
Collapse
Affiliation(s)
- Bria L Macklin
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Ying-Yu Lin
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Kevin Emmerich
- Department of Ophthalmology, Wilmer Eye Institute and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Emily Wisniewski
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Brian M Polster
- Department of Anesthesiology and Center for Shock, Trauma, and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Jeff S Mumm
- Department of Ophthalmology, Wilmer Eye Institute and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, 21218, USA. .,Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA.
| |
Collapse
|
16
|
Hoareau M, El Kholti N, Debret R, Lambert E. Zebrafish as a Model to Study Vascular Elastic Fibers and Associated Pathologies. Int J Mol Sci 2022; 23:2102. [PMID: 35216218 PMCID: PMC8875079 DOI: 10.3390/ijms23042102] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/09/2022] [Accepted: 02/12/2022] [Indexed: 02/06/2023] Open
Abstract
Many extensible tissues such as skin, lungs, and blood vessels require elasticity to function properly. The recoil of elastic energy stored during a stretching phase is provided by elastic fibers, which are mostly composed of elastin and fibrillin-rich microfibrils. In arteries, the lack of elastic fibers leads to a weakening of the vessel wall with an increased risk to develop cardiovascular defects such as stenosis, aneurysms, and dissections. The development of new therapeutic molecules involves preliminary tests in animal models that recapitulate the disease and whose response to drugs should be as close as possible to that of humans. Due to its superior in vivo imaging possibilities and the broad tool kit for forward and reverse genetics, the zebrafish has become an important model organism to study human pathologies. Moreover, it is particularly adapted to large scale studies, making it an attractive model in particular for the first steps of investigations. In this review, we discuss the relevance of the zebrafish model for the study of elastic fiber-related vascular pathologies. We evidence zebrafish as a compelling alternative to conventional mouse models.
Collapse
Affiliation(s)
- Marie Hoareau
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Institut de Biologie et Chimie des Protéines, Université Lyon 1, 7, Passage du Vercors, CEDEX 07, F-69367 Lyon, France; (N.E.K.); (R.D.)
| | | | | | - Elise Lambert
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Institut de Biologie et Chimie des Protéines, Université Lyon 1, 7, Passage du Vercors, CEDEX 07, F-69367 Lyon, France; (N.E.K.); (R.D.)
| |
Collapse
|
17
|
Chen J, Wang Y, Wang S, Zhao X, Zhao L, Wang Y. Salvianolic acid B and ferulic acid synergistically promote angiogenesis in HUVECs and zebrafish via regulating VEGF signaling. JOURNAL OF ETHNOPHARMACOLOGY 2022; 283:114667. [PMID: 34597652 DOI: 10.1016/j.jep.2021.114667] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/09/2021] [Accepted: 09/19/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Induced vascular growth in the myocardium has been widely acknowledged as a promising intervention strategy for patients with ischemic coronary artery disease. Yet despite long-term efforts on gene, protein or cell-based pro-angiogenic therapies, the clinical translation remains challenging. Noticeably, multiple medicinal herbs have long-term documented effects in promoting blood circulation. Salvia miltiorrhiza and Ligusticum stratum are two representative traditional Chinese medicine herbs with suggested roles in enhancing organ blood supply, and Guanxinning Tablet (GXNT), a botanical drug which is formulated with these two herbs, exhibited significant efficacy against angina pectoris in clinical practices. AIM OF THE STUDY This study aimed to examine the pro-angiogenic activity of GXNT and its major components, as well as to explore their pharmacological mechanism in promoting angiogenesis. MATERIALS AND METHODS In vitro, the pro-angiogenic effects of GXNT and its major components were examined on human umbilical vein endothelial cells by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT), scratch assay, and endothelial cell tube formation assay. In vivo, the pro-angiogenic effects were examined on the ponatinib-induced angiogenesis defective zebrafish model. The active compounds were identified through phenotype-based screening in zebrafish, and their pharmacological mechanism was explored in both in vitro and in vivo models by immunofluorescent staining, cell cycle analysis, quantitative PCR and whole embryo in-situ hybridization. RESULTS We demonstrated strong pro-angiogenic effects of GXNT in both human umbilical vein endothelial cells and zebrafish model. Moreover, through phenotype-based screening in zebrafish for active compounds, pro-angiogenic effects was discovered for salvianolic acid B (Sal B), a major component of Salvia miltiorrhiza, and its activity was further enhanced when co-administered with ferulic acid (FA), which is contained in Ligusticum stratum. On the cellular level, Sal B and FA cotreatment increased endothelial cell proliferation of sprouting arterial intersomitic vessels in zebrafish, as well as largely restored G1-S cell cycle progression and cyclin D1 expression in angiogenic defective HUVECs. Through quantitative transcriptional analysis, increased expression of vegfr2 (kdr, kdrl) and vegfr1 was detected after GXNT or SalB/FA treatment, together with upregulated transcription of their ligands including vegf-a, vegf-b, and pgfb. Bevacizumab, an anti-human VEGF-A monoclonal antibody, was able to significantly, but not completely, block the pro-angiogenic effects of GXNT or SalB/FA, suggesting their multi-targeting properties. CONCLUSIONS In conclusion, from a traditional Chinese medicine with effects in enhancing blood circulation, we demonstrated the synergistic pro-angiogenic effects of Sal B and FA via both in vitro and in vivo models, which function at least partially through regulating the expression of VEGF receptors and ligands. Future studies are warranted to further elaborate the molecular interaction between these two compounds and the key regulators in the process of neovascularization.
Collapse
Affiliation(s)
- Jing Chen
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yingchao Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Shufang Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiaoping Zhao
- College of Preclinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Zhao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Innovation Center in Zhejiang University, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617, China.
| |
Collapse
|
18
|
Sun M, Qin J, Kang Y, Zhang Y, Ba M, Yang H, Duan Y, Yao Y. 2-Methoxydiol derivatives as new tubulin and HDAC dual-targeting inhibitors, displaying antitumor and antiangiogenic response. Bioorg Chem 2022; 120:105625. [DOI: 10.1016/j.bioorg.2022.105625] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/29/2021] [Accepted: 01/11/2022] [Indexed: 12/22/2022]
|
19
|
Chen Y, Evans PC, Wilkinson RN. A Workflow to Track and Analyze Endothelial Migration During Vascular Development in Zebrafish Embryos Using Lightsheet Microscopy. Methods Mol Biol 2022; 2441:19-28. [PMID: 35099725 DOI: 10.1007/978-1-0716-2059-5_2] [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: 11/30/2022]
Abstract
Zebrafish allow unrivalled in vivo imaging of vascular development due to their optical translucency and the availability of transgenic lines which fluorescently label cells and tissues of interest. Advances in light sheet fluorescence microscopy allow longer and faster imaging of live embryos at higher resolutions than previously possible, which facilitates study of dynamic cellular and molecular mechanisms underlying vessel formation and function. Here we describe a workflow using lightsheet microscopy to quantify endothelial cell (EC) migration dynamics during vascular development. Tracking movement of EC nuclei and analyzing the properties of EC migration trajectories permit detailed studies of angiogenesis and vascular remodeling in different contexts.
Collapse
Affiliation(s)
- Yan Chen
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK.
| | - Paul C Evans
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Robert N Wilkinson
- School of Life Sciences, Medical School, Queens Medical Centre, University of Nottingham, Nottingham, UK
| |
Collapse
|
20
|
Karas BF, Hotz JM, Gural BM, Terez KR, DiBona VL, Côrte-Real L, Valente A, Buckley BT, Cooper KR. Anticancer Activity and In Vitro to In Vivo Mechanistic Recapitulation of Novel Ruthenium-Based Metallodrugs in the Zebrafish Model. Toxicol Sci 2021; 182:29-43. [PMID: 33822233 DOI: 10.1093/toxsci/kfab041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Ruthenium is popular as a metal core for chemotherapeutics, due to versatile molecular coordination. Because new metallodrugs are synthesized at high rates, our studies included assays in zebrafish to expedite the initial evaluation as anticancer agents. Here we evaluated novel metallodrugs (PMC79 and LCR134), and cisplatin, a widely used platinum-based chemotherapeutic. We hypothesized that this model could characterize anticancer properties and recapitulate previous in vitro results in vivo. Our findings suggest anticancer properties of PMC79 and LCR134 were similar with less toxicity than cisplatin. Exposures from 24 to 72 h at or below the LOAELs of PMC79 and LCR134 (3.9 µM and 13.5 µm, respectively), impaired blood vessel development and tailfin regeneration. Blood vessel examination through live imaging of larvae revealed distinct regional antiangiogenic impacts. The significant decrease in gene expression of the VEGF-HIF pathway and beta-actin could explain the morphological effects observed in the whole organism following exposure. Tailfin amputation in larvae exposed to PMC79 or LCR134 inhibited tissue regrowth and cell division, but did not impact normal cell proliferation unlike cisplatin. This suggests Ru drugs may be more selective in targeting cancerous cells than cisplatin. Additionally, in vitro mechanisms were confirmed. PMC79 disrupted cytoskeleton formation in larvae and P-glycoprotein transporters in vivo was inhibited at low doses which could limit off-target effects of chemotherapeutics. Our results demonstrate the value for using the zebrafish in metallodrug research to evaluate mechanisms and off-target effects. In light of the findings reported in this article, future investigation of PMC79 and LCR134 are warranted in higher vertebrate models.
Collapse
Affiliation(s)
- Brittany F Karas
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey 08854, USA
| | - Jordan M Hotz
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey 08854, USA.,Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Brian M Gural
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey 08854, USA
| | - Kristin R Terez
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey 08854, USA
| | - Victoria L DiBona
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey 08854, USA
| | - Leonor Côrte-Real
- Centro de Química Estrutural and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa 1749-016, Portugal
| | - Andreia Valente
- Centro de Química Estrutural and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa 1749-016, Portugal
| | - Brian T Buckley
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Keith R Cooper
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey 08854, USA
| |
Collapse
|
21
|
Vega-Granados K, Cruz-Reyes J, Horta-Marrón JF, Marí-Beffa M, Díaz-Rubio L, Córdova-Guerrero I, Chávez-Velasco D, Ocaña MC, Medina MA, Romero-Sánchez LB. Synthesis, characterization and biological evaluation of octyltrimethylammonium tetrathiotungstate. Biometals 2020; 34:107-117. [PMID: 33180255 DOI: 10.1007/s10534-020-00267-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 10/30/2020] [Indexed: 10/23/2022]
Abstract
Octyltrimethylammonium tetrathiotungstate salt (ATT-C8) was synthesized and its ability to chelate copper was evaluated. The biological and toxic aspects were evaluated by in vitro and in vivo assays, using bovine aorta endothelial cells (BAEC) and zebrafish (Danio rerio) embryos. The obtained results suggest that ATT-C8 has better biocompatibility, showing a significantly lower lethal concentration 50 (LC50) value in comparison to ammonium tetrathiotungstate (ATT). Zebrafish embryos assay results indicate that both tetrathiotungstate salts at the studied concentrations increase the hatching time. Even more, an in vivo assay showed that synthesized materials behave as copper antagonists and have the ability to inhibit its toxicological effects. Also, both materials were found to be active for the in vitro 2,2-diphenyl-1-picrylhydrazyl (DPPH) assay. The characterization of the materials was carried out using the following spectroscopic techniques: Ultraviolet-Visible (UV-Vis), Fourier Transform Infrared (FTIR) and proton nuclear magnetic resonance (1H-NRM).
Collapse
Affiliation(s)
- Karla Vega-Granados
- Faculty of Chemical Sciences and Engineering, Autonomous University of Baja California (UABC), Universidad 14418, Parque Internacional Industrial Tijuana, 22390, Tijuana, BC, Mexico
| | - Juan Cruz-Reyes
- Faculty of Chemical Sciences and Engineering, Autonomous University of Baja California (UABC), Universidad 14418, Parque Internacional Industrial Tijuana, 22390, Tijuana, BC, Mexico
| | - José F Horta-Marrón
- Faculty of Chemical Sciences and Engineering, Autonomous University of Baja California (UABC), Universidad 14418, Parque Internacional Industrial Tijuana, 22390, Tijuana, BC, Mexico
| | - Manuel Marí-Beffa
- Department of Cell Biology, Genetics and Physiology, University of Malaga, 29071, Malaga, Spain
| | - Laura Díaz-Rubio
- Faculty of Chemical Sciences and Engineering, Autonomous University of Baja California (UABC), Universidad 14418, Parque Internacional Industrial Tijuana, 22390, Tijuana, BC, Mexico
| | - Iván Córdova-Guerrero
- Faculty of Chemical Sciences and Engineering, Autonomous University of Baja California (UABC), Universidad 14418, Parque Internacional Industrial Tijuana, 22390, Tijuana, BC, Mexico
| | - Daniel Chávez-Velasco
- Center for Graduates and Research in Chemistry, National Technological Institute of Mexico/ Technological Institute of Tijuana, 22510, Tijuana, BC, Mexico
| | - M Carmen Ocaña
- Department of Molecular Biology and Biochemistry, Faculty of Science, University of Malaga, 29071, Malaga, Spain
| | - Miguel A Medina
- Department of Molecular Biology and Biochemistry, Faculty of Science, University of Malaga, 29071, Malaga, Spain
| | - Lilian B Romero-Sánchez
- Faculty of Chemical Sciences and Engineering, Autonomous University of Baja California (UABC), Universidad 14418, Parque Internacional Industrial Tijuana, 22390, Tijuana, BC, Mexico.
| |
Collapse
|
22
|
Wu X, Zhou J, Li D. Orientation of the Mitotic Spindle in Blood Vessel Development. Front Cell Dev Biol 2020; 8:583325. [PMID: 33072763 PMCID: PMC7533553 DOI: 10.3389/fcell.2020.583325] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/01/2020] [Indexed: 12/21/2022] Open
Abstract
Angiogenesis requires coordinated endothelial cell specification, proliferation, and collective migration. The orientation of endothelial cell division is tightly regulated during the earliest stages of blood vessel formation in response to morphogenetic cues and the controlled orientation of the mitotic spindle. Consequently, oriented cell division is a vital mechanism in vessel morphogenesis, and defective spindle orientation can perturb the spatial arrangement of daughter cells and consequently contribute to several diseases related to vascular development. Many factors affect endothelial cell proliferation and orientation and therefore blood vessel formation, with the relationship between improper spindle orientation in endothelial cells and various diseases extensively studied. Here we review the molecular mechanisms driving the orientation of endothelial cell division, particularly with respect to the mitotic spindle, and how these processes affect vascular development, disease pathogenesis, and their potential as novel targets.
Collapse
Affiliation(s)
- Xuemei Wu
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Jun Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China.,Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Institute of Biomedical Sciences, Shandong Normal University, Jinan, China
| | - Dengwen Li
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
23
|
Ye M, Chen Y. Zebrafish as an emerging model to study gonad development. Comput Struct Biotechnol J 2020; 18:2373-2380. [PMID: 32994895 PMCID: PMC7498840 DOI: 10.1016/j.csbj.2020.08.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/06/2020] [Accepted: 08/26/2020] [Indexed: 01/24/2023] Open
Abstract
The zebrafish (Danio rerio) has emerged as a popular model organism in developmental biology and pharmacogenetics due to its attribute of pathway conservation. Coupled with the availability of robust genetic and transgenic tools, transparent embryos and rapid larval development, studies of zebrafish allow detailed cellular analysis of many dynamic processes. In recent decades, the cellular and molecular mechanisms involved in the process of gonad development have been the subject of intense research using zebrafish models. In this mini-review, we give a brief overview of these studies, and highlight the essential genes involved in sex determination and gonad development in zebrafish.
Collapse
Affiliation(s)
- Mengling Ye
- Division of Medical Genetics and Genomics, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Genetics, Zhejiang University and Department of Genetics, Zhejiang University School of Medicine, Hangzhou, China
| | - Ye Chen
- Division of Medical Genetics and Genomics, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Genetics, Zhejiang University and Department of Genetics, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
24
|
Lawson ND, Li R, Shin M, Grosse A, Yukselen O, Stone OA, Kucukural A, Zhu L. An improved zebrafish transcriptome annotation for sensitive and comprehensive detection of cell type-specific genes. eLife 2020; 9:55792. [PMID: 32831172 PMCID: PMC7486121 DOI: 10.7554/elife.55792] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 08/21/2020] [Indexed: 02/07/2023] Open
Abstract
The zebrafish is ideal for studying embryogenesis and is increasingly applied to model human disease. In these contexts, RNA-sequencing (RNA-seq) provides mechanistic insights by identifying transcriptome changes between experimental conditions. Application of RNA-seq relies on accurate transcript annotation for a genome of interest. Here, we find discrepancies in analysis from RNA-seq datasets quantified using Ensembl and RefSeq zebrafish annotations. These issues were due, in part, to variably annotated 3' untranslated regions and thousands of gene models missing from each annotation. Since these discrepancies could compromise downstream analyses and biological reproducibility, we built a more comprehensive zebrafish transcriptome annotation that addresses these deficiencies. Our annotation improves detection of cell type-specific genes in both bulk and single cell RNA-seq datasets, where it also improves resolution of cell clustering. Thus, we demonstrate that our new transcriptome annotation can outperform existing annotations, providing an important resource for zebrafish researchers.
Collapse
Affiliation(s)
- Nathan D Lawson
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, United States
| | - Rui Li
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, United States
| | - Masahiro Shin
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, United States
| | - Ann Grosse
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, United States
| | - Onur Yukselen
- Bioinformatics Core, University of Massachusetts Medical School, Worcester, United States
| | - Oliver A Stone
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Alper Kucukural
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States.,Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, United States
| | - Lihua Zhu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, United States.,Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States.,Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, United States
| |
Collapse
|
25
|
Jarque S, Rubio-Brotons M, Ibarra J, Ordoñez V, Dyballa S, Miñana R, Terriente J. Morphometric analysis of developing zebrafish embryos allows predicting teratogenicity modes of action in higher vertebrates. Reprod Toxicol 2020; 96:337-348. [PMID: 32822784 DOI: 10.1016/j.reprotox.2020.08.004] [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] [Received: 03/18/2020] [Revised: 08/03/2020] [Accepted: 08/07/2020] [Indexed: 12/22/2022]
Abstract
The early identification of teratogens in humans and animals is mandatory for drug discovery and development. Zebrafish has emerged as an alternative model to traditional preclinical models for predicting teratogenicity and other potential chemical-induced toxicity hazards. To prove its predictivity, we exposed zebrafish embryos from 0 to 96 h post fertilization to a battery of 31 compounds classified as teratogens or non-teratogens in mammals. The teratogenicity score was based on the measurement of 16 phenotypical parameters, namely heart edema, pigmentation, body length, eye size, yolk size, yolk sac edema, otic vesicle defects, otoliths defects, body axis defects, developmental delay, tail bending, scoliosis, lateral fins absence, hatching ratio, lower jaw malformations and tissue necrosis. Among the 31 compounds, 20 were detected as teratogens and 11 as non-teratogens, resulting in 94.44 % sensitivity, 90.91 % specificity and 87.10 % accuracy compared to rodents. These percentages decreased slightly when referred to humans, with 87.50 % sensitivity, 81.82 % specificity and 74.19 % accuracy, but allowed an increase in the prediction levels reported by rodents for the same compounds. Positive compounds showed a high correlation among teratogenic parameters, pointing out at general developmental delay as major cause to explain the physiological/morphological malformations. A more detailed analysis based on deviations from main trends revealed potential specific modes of action for some compounds such as retinoic acid, DEAB, ochratoxin A, haloperidol, warfarin, valproic acid, acetaminophen, dasatinib, imatinib, dexamethasone, 6-aminonicotinamide and bisphenol A. The high degree of predictivity and the possibility of applying mechanistic approaches makes zebrafish a powerful model for screening teratogenicity.
Collapse
Affiliation(s)
- Sergio Jarque
- ZeClinics SL, Carretera de Can Ruti, Camí de les Escoles, s/n, Edificio IGTP Muntanya, Badalona, 08916 Barcelona, Spain.
| | - Maria Rubio-Brotons
- ZeClinics SL, Carretera de Can Ruti, Camí de les Escoles, s/n, Edificio IGTP Muntanya, Badalona, 08916 Barcelona, Spain
| | - Jone Ibarra
- ZeClinics SL, Carretera de Can Ruti, Camí de les Escoles, s/n, Edificio IGTP Muntanya, Badalona, 08916 Barcelona, Spain
| | - Víctor Ordoñez
- ZeClinics SL, Carretera de Can Ruti, Camí de les Escoles, s/n, Edificio IGTP Muntanya, Badalona, 08916 Barcelona, Spain
| | - Sylvia Dyballa
- ZeClinics SL, Carretera de Can Ruti, Camí de les Escoles, s/n, Edificio IGTP Muntanya, Badalona, 08916 Barcelona, Spain
| | - Rafael Miñana
- ZeClinics SL, Carretera de Can Ruti, Camí de les Escoles, s/n, Edificio IGTP Muntanya, Badalona, 08916 Barcelona, Spain
| | - Javier Terriente
- ZeClinics SL, Carretera de Can Ruti, Camí de les Escoles, s/n, Edificio IGTP Muntanya, Badalona, 08916 Barcelona, Spain.
| |
Collapse
|
26
|
Vedder VL, Aherrahrou Z, Erdmann J. Dare to Compare. Development of Atherosclerotic Lesions in Human, Mouse, and Zebrafish. Front Cardiovasc Med 2020; 7:109. [PMID: 32714944 PMCID: PMC7344238 DOI: 10.3389/fcvm.2020.00109] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/26/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular diseases, such as atherosclerosis, are the leading cause of death worldwide. Although mice are currently the most commonly used model for atherosclerosis, zebrafish are emerging as an alternative, especially for inflammatory and lipid metabolism studies. Here, we review the history of in vivo atherosclerosis models and highlight the potential for future studies on inflammatory responses in lipid deposits in zebrafish, based on known immune reactions in humans and mice, in anticipation of new zebrafish models with more advanced atherosclerotic plaques.
Collapse
Affiliation(s)
- Viviana L Vedder
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany.,University Heart Centre Lübeck, Lübeck, Germany
| | - Zouhair Aherrahrou
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany.,University Heart Centre Lübeck, Lübeck, Germany
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany.,University Heart Centre Lübeck, Lübeck, Germany
| |
Collapse
|
27
|
Endothelial Autophagy: an Effective Target for Radiation-induced Cerebral Capillary Damage. Sci Rep 2020; 10:614. [PMID: 31953486 PMCID: PMC6968992 DOI: 10.1038/s41598-019-57234-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 12/11/2019] [Indexed: 02/05/2023] Open
Abstract
Toxicity to central nervous system tissues is the common side effects for radiotherapy of brain tumor. The radiation toxicity has been thought to be related to the damage of cerebral endothelium. However, because of lacking a suitable high-resolution vivo model, cellular response of cerebral capillaries to radiation remained unclear. Here, we present the flk:eGFP transgenic zebrafish larvae as a feasible model to study the radiation toxicity to cerebral capillary. We showed that, in living zebrafish larvae, radiation could induce acute cerebral capillary shrinkage and blood-flow obstruction, resulting brain hypoxia and glycolysis retardant. Although in vivo neuron damage was also observed after the radiation exposure, further investigation found that they didn’t response to the same dosage of radiation in vitro, indicating that radiation induced neuron damage was a secondary-effect of cerebral vascular function damage. In addition, transgenic labeling and qPCR results showed that the radiation-induced acute cerebral endothelial damage was correlated with intensive endothelial autophagy. Different autophagy inhibitors could significantly alleviate the radiation-induced cerebral capillary damage and prolong the survival of zebrafish larvae. Therefore, we showed that radiation could directly damage cerebral capillary, resulting to blood flow deficiency and neuron death, which suggested endothelial autophagy as a potential target for radiation-induced brain toxicity.
Collapse
|
28
|
Arjmand B, Tayanloo-Beik A, Foroughi Heravani N, Alaei S, Payab M, Alavi-Moghadam S, Goodarzi P, Gholami M, Larijani B. Zebrafish for Personalized Regenerative Medicine; A More Predictive Humanized Model of Endocrine Disease. Front Endocrinol (Lausanne) 2020; 11:396. [PMID: 32765420 PMCID: PMC7379230 DOI: 10.3389/fendo.2020.00396] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 05/18/2020] [Indexed: 12/18/2022] Open
Abstract
Regenerative medicine is a multidisciplinary field that aims to determine different factors and develop various methods to regenerate impaired tissues, organs, and cells in the disease and impairment conditions. When treatment procedures are specified according to the individual's information, the leading role of personalized regenerative medicine will be revealed in developing more effective therapies. In this concept, endocrine disorders can be considered as potential candidates for regenerative medicine application. Diabetes mellitus as a worldwide prevalent endocrine disease causes different damages such as blood vessel damages, pancreatic damages, and impaired wound healing. Therefore, a global effort has been devoted to diabetes mellitus investigations. Hereupon, the preclinical study is a fundamental step. Up to now, several species of animals have been modeled to identify the mechanism of multiple diseases. However, more recent researches have been demonstrated that animal models with the ability of tissue regeneration are more suitable choices for regenerative medicine studies in endocrine disorders, typically diabetes mellitus. Accordingly, zebrafish has been introduced as a model that possesses the capacity to regenerate different organs and tissues. Especially, fine regeneration in zebrafish has been broadly investigated in the regenerative medicine field. In addition, zebrafish is a suitable model for studying a variety of different situations. For instance, it has been used for developmental studies because of the special characteristics of its larva. In this review, we discuss the features of zebrafish that make it a desirable animal model, the advantages of zebrafish and recent research that shows zebrafish is a promising animal model for personalized regenerative diseases. Ultimately, we conclude that as a newly introduced model, zebrafish can have a leading role in regeneration studies of endocrine diseases and provide a good perception of underlying mechanisms.
Collapse
Affiliation(s)
- Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Najmeh Foroughi Heravani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Setareh Alaei
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Moloud Payab
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Goodarzi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Gholami
- Department of Toxicology and Pharmacology, Toxicology and Poisoning Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- *Correspondence: Bagher Larijani
| |
Collapse
|
29
|
Schmöhl F, Peters V, Schmitt CP, Poschet G, Büttner M, Li X, Weigand T, Poth T, Volk N, Morgenstern J, Fleming T, Nawroth PP, Kroll J. CNDP1 knockout in zebrafish alters the amino acid metabolism, restrains weight gain, but does not protect from diabetic complications. Cell Mol Life Sci 2019; 76:4551-4568. [PMID: 31073745 PMCID: PMC11105213 DOI: 10.1007/s00018-019-03127-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/22/2019] [Accepted: 04/30/2019] [Indexed: 12/12/2022]
Abstract
The gene CNDP1 was associated with the development of diabetic nephropathy. Its enzyme carnosinase 1 (CN1) primarily hydrolyzes the histidine-containing dipeptide carnosine but other organ and metabolic functions are mainly unknown. In our study we generated CNDP1 knockout zebrafish, which showed strongly decreased CN1 activity and increased intracellular carnosine levels. Vasculature and kidneys of CNDP1-/- zebrafish were not affected, except for a transient glomerular alteration. Amino acid profiling showed a decrease of certain amino acids in CNDP1-/- zebrafish, suggesting a specific function for CN1 in the amino acid metabolisms. Indeed, we identified a CN1 activity for Ala-His and Ser-His. Under diabetic conditions increased carnosine levels in CNDP1-/- embryos could not protect from respective organ alterations. Although, weight gain through overfeeding was restrained by CNDP1 loss. Together, zebrafish exhibits CN1 functions, while CNDP1 knockout alters the amino acid metabolism, attenuates weight gain but cannot protect organs from diabetic complications.
Collapse
Affiliation(s)
- Felix Schmöhl
- European Center for Angioscience (ECAS), Department of Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Verena Peters
- Center for Paediatric and Adolescent Medicine, University of Heidelberg, Im Neuenheimer Feld 669, 69120, Heidelberg, Germany
| | - Claus Peter Schmitt
- Center for Paediatric and Adolescent Medicine, University of Heidelberg, Im Neuenheimer Feld 669, 69120, Heidelberg, Germany
| | - Gernot Poschet
- Center for Organismal Studies (COS), University of Heidelberg, Im Neuenheimer Feld 360, 69120, Heidelberg, Germany
| | - Michael Büttner
- Center for Organismal Studies (COS), University of Heidelberg, Im Neuenheimer Feld 360, 69120, Heidelberg, Germany
| | - Xiaogang Li
- European Center for Angioscience (ECAS), Department of Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Tim Weigand
- Center for Paediatric and Adolescent Medicine, University of Heidelberg, Im Neuenheimer Feld 669, 69120, Heidelberg, Germany
| | - Tanja Poth
- CMCP-Center for Model System and Comparative Pathology, Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Nadine Volk
- Tissue Bank of the National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Jakob Morgenstern
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Thomas Fleming
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Peter P Nawroth
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Center for Diabetes Research (DZD), 85764, München-Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz-Zentrum, München, Im Neuenheimer Feld 410, F02 Room 02.414-02.434, 69120, Heidelberg, Germany
| | - Jens Kroll
- European Center for Angioscience (ECAS), Department of Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany.
| |
Collapse
|
30
|
Wang G, Xiao Q, Wu Y, Wei YJ, Jing Y, Cao XR, Gong ZN. Design and synthesis of novel celastrol derivative and its antitumor activity in hepatoma cells and antiangiogenic activity in zebrafish. J Cell Physiol 2019; 234:16431-16446. [PMID: 30770566 DOI: 10.1002/jcp.28312] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/20/2019] [Accepted: 01/24/2019] [Indexed: 01/24/2023]
Abstract
Two series of celastrol derivatives were designed and synthesized by modifying carboxylic acid at the 28th position with amino acid, and their intermediates with isobutyrate at the third position. All compounds were evaluated for their antiproliferation activity by four human cancer cell lines (SCG7901, HGC27, HepG2, and Bel7402) and one normal cell LO2. The most promising compound, compound 8, showed superior bioactivity and lower toxicity than others including celastrol. Further underlying tests illustrated that compound 8 induced apoptosis and cell arrest at G2/M and inhibited proliferation and mobility of human hepatoma cells by suppressing the signal transducer and activator of transcription-3 signaling pathway. Besides these, a highly accurate and reproducible high performance liquid chromatography protocol was established to determine celastrol and compound 8 absorption in zebrafish, and results demonstrated that their concentration increased rapidly within 4 hr in a time-dependent manner and the concentration of compound 8 was higher than that of celastrol. In addition, without detection at 12 hr, compound 8 was rapidly metabolized in vivo. These findings are very helpful for the structural modification of celastrol and other bioactive compounds to improve their bioactivity, toxicity, and absorption.
Collapse
Affiliation(s)
- Gang Wang
- Center for New Drug Research and Development, College of Life Science, Nanjing Normal University, Nanjing, People's Republic of China
| | - Qi Xiao
- Center for New Drug Research and Development, College of Life Science, Nanjing Normal University, Nanjing, People's Republic of China
| | - Yao Wu
- Center for New Drug Research and Development, College of Life Science, Nanjing Normal University, Nanjing, People's Republic of China
| | - Ying-Jie Wei
- Key Laboratory of Oral Drug Delivery System of Chinese Meteria Media of State Administration of Tradition Chinese Medicine, Jiangsu Branch of China Academy of Chinese Medical Science, Nanjing, People's Republic of China
| | - Yue Jing
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Xiang-Rong Cao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, People's Republic of China
| | - Zhu-Nan Gong
- Center for New Drug Research and Development, College of Life Science, Nanjing Normal University, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, People's Republic of China
| |
Collapse
|
31
|
O'Brown NM, Megason SG, Gu C. Suppression of transcytosis regulates zebrafish blood-brain barrier function. eLife 2019; 8:e47326. [PMID: 31429822 PMCID: PMC6726461 DOI: 10.7554/elife.47326] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 08/19/2019] [Indexed: 12/15/2022] Open
Abstract
As an optically transparent model organism with an endothelial blood-brain barrier (BBB), zebrafish offer a powerful tool to study the vertebrate BBB. However, the precise developmental profile of functional zebrafish BBB acquisition and the subcellular and molecular mechanisms governing the zebrafish BBB remain poorly characterized. Here, we capture the dynamics of developmental BBB leakage using live imaging, revealing a combination of steady accumulation in the parenchyma and sporadic bursts of tracer leakage. Electron microscopy studies further reveal high levels of transcytosis in brain endothelium early in development that are suppressed later. The timing of this suppression of transcytosis coincides with the establishment of BBB function. Finally, we demonstrate a key mammalian BBB regulator Mfsd2a, which inhibits transcytosis, plays a conserved role in zebrafish, as mfsd2aa mutants display increased BBB permeability due to increased transcytosis. Our findings indicate a conserved developmental program of barrier acquisition between zebrafish and mice.
Collapse
Affiliation(s)
| | - Sean G Megason
- Department of Systems BiologyHarvard Medical SchoolBostonUnited States
| | - Chenghua Gu
- Department of NeurobiologyHarvard Medical SchoolBostonUnited States
| |
Collapse
|
32
|
Abstract
The formation and remodeling of a functional circulatory system is critical for sustaining prenatal and postnatal life. During embryogenesis, newly differentiated endothelial cells require further specification to create the unique features of distinct vessel subtypes needed to support tissue morphogenesis. In this review, we explore signaling pathways and transcriptional regulators that modulate endothelial cell differentiation and specification, as well as applications of these processes to stem cell biology and regenerative medicine. We also summarize recent technical advances, including the growing utilization of single-cell sequencing to study vascular heterogeneity and development.
Collapse
Affiliation(s)
- Jingyao Qiu
- From the Department of Genetics (J.Q., K.K.H.), Yale University School of Medicine, New Haven, CT.,Department of Medicine (J.Q., K.K.H.), Yale University School of Medicine, New Haven, CT.,Yale Cardiovascular Research Center (J.Q., K.K.H.), Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program (J.Q., K.K.H.), Yale University School of Medicine, New Haven, CT
| | - Karen K Hirschi
- From the Department of Genetics (J.Q., K.K.H.), Yale University School of Medicine, New Haven, CT.,Department of Medicine (J.Q., K.K.H.), Yale University School of Medicine, New Haven, CT.,Yale Cardiovascular Research Center (J.Q., K.K.H.), Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program (J.Q., K.K.H.), Yale University School of Medicine, New Haven, CT
| |
Collapse
|
33
|
Chatmethakul T, Roghair RD. Risk of hypertension following perinatal adversity: IUGR and prematurity. J Endocrinol 2019; 242:T21-T32. [PMID: 30657741 PMCID: PMC6594910 DOI: 10.1530/joe-18-0687] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 01/18/2019] [Indexed: 12/12/2022]
Abstract
Consistent with the paradigm shifting observations of David Barker and colleagues that revealed a powerful relationship between decreased weight through 2 years of age and adult disease, intrauterine growth restriction (IUGR) and preterm birth are independent risk factors for the development of subsequent hypertension. Animal models have been indispensable in defining the mechanisms responsible for these associations and the potential targets for therapeutic intervention. Among the modifiable risk factors, micronutrient deficiency, physical immobility, exaggerated stress hormone exposure and deficient trophic hormone production are leading candidates for targeted therapies. With the strong inverse relationship seen between gestational age at delivery and the risk of hypertension in adulthood trumping all other major cardiovascular risk factors, improvements in neonatal care are required. Unfortunately, therapeutic breakthroughs have not kept pace with rapidly improving perinatal survival, and groundbreaking bench-to-bedside studies are urgently needed to mitigate and ultimately prevent the tsunami of prematurity-related adult cardiovascular disease that may be on the horizon. This review highlights our current understanding of the developmental origins of hypertension and draws attention to the importance of increasing the availability of lactation consultants, nutritionists, pharmacists and physical therapists as critical allies in the battle that IUGR or premature infants are waging not just for survival but also for their future cardiometabolic health.
Collapse
Affiliation(s)
- Trassanee Chatmethakul
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Robert D Roghair
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
34
|
Pelster B, Egg M. Hypoxia-inducible transcription factors in fish: expression, function and interconnection with the circadian clock. J Exp Biol 2018; 221:221/13/jeb163709. [DOI: 10.1242/jeb.163709] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
ABSTRACT
The hypoxia-inducible transcription factors are key regulators for the physiological response to low oxygen availability. In vertebrates, typically three Hif-α isoforms, Hif-1α, Hif-2α and Hif-3α, are expressed, each of which, together with Hif-1β, may form a functional heterodimer under hypoxic conditions, controlling expression of hundreds of genes. A teleost-specific whole-genome duplication complicates the analysis of isoform-specific functions in fish, but recent studies suggest that the existence of paralogues of a specific isoform opens up the possibility for a subfunctionalization. In contrast to during development inside the uterus, fish eggs are freely accessible and studies analyzing Hif expression in fish embryos during development have revealed that Hif proteins are not only controlling the hypoxic response, but are also crucial for proper development and organ differentiation. Significant advances have been made in our knowledge about tissue-specific functions of Hif proteins, especially with respect to gill or gonadal tissue. The hypoxia signalling pathway is known to be tightly and mutually intertwined with the circadian clock in zebrafish and mammals. Recently, a mechanistic explanation for the hypoxia-induced dampening of the transcriptional clock was detected in zebrafish, including also metabolically induced alterations of cellular redox signalling. In turn, MAP kinase-mediated H2O2 signalling modulates the temporal expression of Hif-1α protein, similar to the redox regulation of the circadian clock itself. Once again, the zebrafish has emerged as an excellent model organism with which to explore these specific functional aspects of basic eukaryotic cell biology.
Collapse
Affiliation(s)
- Bernd Pelster
- Institute of Zoology, University of Innsbruck, Technikerstr. 25, A-6020 Innsbruck, Austria
- Center for Molecular Biosciences, University of Innsbruck, A-6020 Innsbruck, Austria
| | - Margit Egg
- Institute of Zoology, University of Innsbruck, Technikerstr. 25, A-6020 Innsbruck, Austria
| |
Collapse
|
35
|
Patton C, Farr GH, An D, Martini PG, Maves L. Lipid Nanoparticle Packaging Is an Effective and Nontoxic mRNA Delivery Platform in Embryonic Zebrafish. Zebrafish 2018; 15:217-227. [DOI: 10.1089/zeb.2017.1511] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Clay Patton
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington
| | - Gist H. Farr
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington
| | - Ding An
- Moderna Therapeutics, Cambridge, Massachusetts
| | | | - Lisa Maves
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington
- Department of Pediatrics, University of Washington, Seattle, Washington
| |
Collapse
|
36
|
García-Caballero M, Quesada AR, Medina MA, Marí-Beffa M. Fishing anti(lymph)angiogenic drugs with zebrafish. Drug Discov Today 2017; 23:366-374. [PMID: 29081356 DOI: 10.1016/j.drudis.2017.10.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 10/13/2017] [Accepted: 10/19/2017] [Indexed: 10/18/2022]
Abstract
Zebrafish, an amenable small teleost fish with a complex mammal-like circulatory system, is being increasingly used for drug screening and toxicity studies. It combines the biological complexity of in vivo models with a higher-throughput screening capability compared with other available animal models. Externally growing, transparent embryos, displaying well-defined blood and lymphatic vessels, allow the inexpensive, rapid, and automatable evaluation of drug candidates that are able to inhibit neovascularisation. Here, we briefly review zebrafish as a model for the screening of anti(lymph)angiogenic drugs, with emphasis on the advantages and limitations of the different zebrafish-based in vivo assays.
Collapse
Affiliation(s)
- Melissa García-Caballero
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, and IBIMA (Biomedical Research Institute of Málaga), University of Málaga, Andalucía Tech, Málaga, Spain; Unit 741 of CIBER de Enfermedades Raras, Málaga, Spain
| | - Ana R Quesada
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, and IBIMA (Biomedical Research Institute of Málaga), University of Málaga, Andalucía Tech, Málaga, Spain; Unit 741 of CIBER de Enfermedades Raras, Málaga, Spain
| | - Miguel A Medina
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, and IBIMA (Biomedical Research Institute of Málaga), University of Málaga, Andalucía Tech, Málaga, Spain; Unit 741 of CIBER de Enfermedades Raras, Málaga, Spain.
| | - Manuel Marí-Beffa
- Department of Cellular Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Málaga, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, Málaga, Spain.
| |
Collapse
|
37
|
Ibrahim M, Richardson MK. Beyond organoids: In vitro vasculogenesis and angiogenesis using cells from mammals and zebrafish. Reprod Toxicol 2017; 73:292-311. [PMID: 28697965 DOI: 10.1016/j.reprotox.2017.07.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/12/2017] [Accepted: 07/05/2017] [Indexed: 12/24/2022]
Abstract
The ability to culture complex organs is currently an important goal in biomedical research. It is possible to grow organoids (3D organ-like structures) in vitro; however, a major limitation of organoids, and other 3D culture systems, is the lack of a vascular network. Protocols developed for establishing in vitro vascular networks typically use human or rodent cells. A major technical challenge is the culture of functional (perfused) networks. In this rapidly advancing field, some microfluidic devices are now getting close to the goal of an artificially perfused vascular network. Another development is the emergence of the zebrafish as a complementary model to mammals. In this review, we discuss the culture of endothelial cells and vascular networks from mammalian cells, and examine the prospects for using zebrafish cells for this objective. We also look into the future and consider how vascular networks in vitro might be successfully perfused using microfluidic technology.
Collapse
Affiliation(s)
- Muhammad Ibrahim
- Animal Science and Health Cluster, Institute of Biology Leiden, Leiden University, The Netherlands; Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar, Pakistan
| | - Michael K Richardson
- Animal Science and Health Cluster, Institute of Biology Leiden, Leiden University, The Netherlands.
| |
Collapse
|
38
|
Harrington JK, Sorabella R, Tercek A, Isler JR, Targoff KL. Nkx2.5 is essential to establish normal heart rate variability in the zebrafish embryo. Am J Physiol Regul Integr Comp Physiol 2017; 313:R265-R271. [PMID: 28615160 DOI: 10.1152/ajpregu.00223.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 06/02/2017] [Accepted: 06/06/2017] [Indexed: 12/11/2022]
Abstract
Heart rate variability (HRV) has become an important clinical marker of cardiovascular health and a research measure for the study of the cardiac conduction system and its autonomic controls. While the zebrafish (Danio rerio) is an ideal vertebrate model for understanding heart development, HRV has only recently been investigated in this system. We have previously demonstrated that nkx2.5 and nkx2.7, two homologues of Nkx2-5 expressed in zebrafish cardiomyocytes, play vital roles in maintaining cardiac chamber-specific characteristics. Given observed defects in ventricular and atrial chamber identities in nkx2.5-/- embryos coupled with conduction system abnormalities in murine models of Nkx2.5 insufficiency, we postulated that reduced HRV would serve as a marker of poor cardiac health in nkx2.5 mutants and in other zebrafish models of human congenital heart disease. Using live video image acquisition, we derived beat-to-beat intervals to compare HRV in wild-type and nkx2.5-/- embryos. Our data illustrate that the nkx2.5 loss-of-function model exhibits increased heart rate and decreased HRV when compared with wild type during embryogenesis. These findings validate HRV analysis as a useful quantitative tool for assessment of cardiac health in zebrafish and underscore the importance of nkx2.5 in maintaining normal heart rate and HRV during early conduction system development.
Collapse
Affiliation(s)
- Jamie K Harrington
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Robert Sorabella
- Division of Cardiothoracic Surgery, Department of Surgery, College of Physicians and Surgeons, Columbia University, New York, New York; and
| | - Abigail Tercek
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Joseph R Isler
- Division of Neonatology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Kimara L Targoff
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York;
| |
Collapse
|
39
|
In vitro development of zebrafish vascular networks. Reprod Toxicol 2017; 70:102-115. [DOI: 10.1016/j.reprotox.2017.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 01/27/2017] [Accepted: 02/08/2017] [Indexed: 12/28/2022]
|
40
|
Zhai Y, Zhao X, Hu GF, Sheng J, Xu Z. Pro-angiogenic effect of ribonuclease like 1 in zebrafish embryo development. GENE REPORTS 2017. [DOI: 10.1016/j.genrep.2016.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
41
|
Baxendale S, van Eeden F, Wilkinson R. The Power of Zebrafish in Personalised Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1007:179-197. [PMID: 28840558 DOI: 10.1007/978-3-319-60733-7_10] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The goal of personalised medicine is to develop tailor-made therapies for patients in whom currently available therapeutics fail. This approach requires correlating individual patient genotype data to specific disease phenotype data and using these stratified data sets to identify bespoke therapeutics. Applications for personalised medicine include common complex diseases which may have multiple targets, as well as rare monogenic disorders, for which the target may be unknown. In both cases, whole genome sequence analysis (WGS) is discovering large numbers of disease associated mutations in new candidate genes and potential modifier genes. Currently, the main limiting factor is the determination of which mutated genes are important for disease progression and therefore represent potential targets for drug discovery. Zebrafish have gained popularity as a model organism for understanding developmental processes, disease mechanisms and more recently for drug discovery and toxicity testing. In this chapter, we will examine the diverse roles that zebrafish can make in the expanding field of personalised medicine, from generating humanised disease models to xenograft screening of different cancer cell lines, through to finding new drugs via in vivo phenotypic screens. We will discuss the tools available for zebrafish research and recent advances in techniques, highlighting the advantages and potential of using zebrafish for high throughput disease modeling and precision drug discovery.
Collapse
Affiliation(s)
- Sarah Baxendale
- The Bateson Centre, Department of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, UK.
| | - Freek van Eeden
- The Bateson Centre, Department of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, UK
| | - Robert Wilkinson
- The Bateson Centre, Department of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, UK.,Department of Infection, Immunity and Cardiovascular Disease, Medical School, Beech Hill Rd, University of Sheffield, Sheffield, S10 2RX, UK
| |
Collapse
|
42
|
Laviña B. Brain Vascular Imaging Techniques. Int J Mol Sci 2016; 18:ijms18010070. [PMID: 28042833 PMCID: PMC5297705 DOI: 10.3390/ijms18010070] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/13/2016] [Accepted: 12/26/2016] [Indexed: 12/13/2022] Open
Abstract
Recent major improvements in a number of imaging techniques now allow for the study of the brain in ways that could not be considered previously. Researchers today have well-developed tools to specifically examine the dynamic nature of the blood vessels in the brain during development and adulthood; as well as to observe the vascular responses in disease situations in vivo. This review offers a concise summary and brief historical reference of different imaging techniques and how these tools can be applied to study the brain vasculature and the blood-brain barrier integrity in both healthy and disease states. Moreover, it offers an overview on available transgenic animal models to study vascular biology and a description of useful online brain atlases.
Collapse
Affiliation(s)
- Bàrbara Laviña
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden.
| |
Collapse
|
43
|
Serbanovic-Canic J, de Luca A, Warboys C, Ferreira PF, Luong LA, Hsiao S, Gauci I, Mahmoud M, Feng S, Souilhol C, Bowden N, Ashton JP, Walczak H, Firmin D, Krams R, Mason JC, Haskard DO, Sherwin S, Ridger V, Chico TJA, Evans PC. Zebrafish Model for Functional Screening of Flow-Responsive Genes. Arterioscler Thromb Vasc Biol 2016; 37:130-143. [PMID: 27834691 PMCID: PMC5172514 DOI: 10.1161/atvbaha.116.308502] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 10/23/2016] [Indexed: 12/22/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Atherosclerosis is initiated at branches and bends of arteries exposed to disturbed blood flow that generates low shear stress. This mechanical environment promotes lesions by inducing endothelial cell (EC) apoptosis and dysfunction via mechanisms that are incompletely understood. Although transcriptome-based studies have identified multiple shear-responsive genes, most of them have an unknown function. To address this, we investigated whether zebrafish embryos can be used for functional screening of mechanosensitive genes that regulate EC apoptosis in mammalian arteries. Approach and Results— First, we demonstrated that flow regulates EC apoptosis in developing zebrafish vasculature. Specifically, suppression of blood flow in zebrafish embryos (by targeting cardiac troponin) enhanced that rate of EC apoptosis (≈10%) compared with controls exposed to flow (≈1%). A panel of candidate regulators of apoptosis were identified by transcriptome profiling of ECs from high and low shear stress regions of the porcine aorta. Genes that displayed the greatest differential expression and possessed 1 to 2 zebrafish orthologues were screened for the regulation of apoptosis in zebrafish vasculature exposed to flow or no-flow conditions using a knockdown approach. A phenotypic change was observed in 4 genes; p53-related protein (PERP) and programmed cell death 2–like protein functioned as positive regulators of apoptosis, whereas angiopoietin-like 4 and cadherin 13 were negative regulators. The regulation of perp, cdh13, angptl4, and pdcd2l by shear stress and the effects of perp and cdh13 on EC apoptosis were confirmed by studies of cultured EC exposed to flow. Conclusions— We conclude that a zebrafish model of flow manipulation coupled to gene knockdown can be used for functional screening of mechanosensitive genes in vascular ECs, thus providing potential therapeutic targets to prevent or treat endothelial injury at atheroprone sites.
Collapse
Affiliation(s)
- Jovana Serbanovic-Canic
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Amalia de Luca
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Christina Warboys
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Pedro F Ferreira
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Le A Luong
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Sarah Hsiao
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Ismael Gauci
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Marwa Mahmoud
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Shuang Feng
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Celine Souilhol
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Neil Bowden
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - John-Paul Ashton
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Henning Walczak
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - David Firmin
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Rob Krams
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Justin C Mason
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Dorian O Haskard
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Spencer Sherwin
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Victoria Ridger
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Timothy J A Chico
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Paul C Evans
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom.
| |
Collapse
|
44
|
Tulotta C, He S, van der Ent W, Chen L, Groenewoud A, Spaink HP, Snaar-Jagalska BE. Imaging Cancer Angiogenesis and Metastasis in a Zebrafish Embryo Model. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 916:239-63. [PMID: 27165357 DOI: 10.1007/978-3-319-30654-4_11] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Tumor angiogenesis and metastasis are key steps of cancer progression. In vitro and animal model studies have contributed to partially elucidating the mechanisms involved in these processes and in developing therapies. Besides the improvements in fundamental research and the optimization of therapeutic regimes, cancer still remains a major health threatening condition and therefore the development of new models is needed. The zebrafish is a powerful tool to study tumor angiogenesis and metastasis, because it allows the visualization of fluorescently labelled tumor cells inducing vessel remodeling, disseminating and invading surrounding tissues in a whole transparent embryo. The embryo model has also been used to address the contribution of the tumor stroma in sustaining tumor angiogenesis and spreading. Simultaneously, new anti-angiogenic drugs and compounds affecting malignant cell survival and migration can be tested by simply adding the compound into the water of living embryos. Therefore the zebrafish model offers the opportunity to gain more knowledge on cancer angiogenesis and metastasis in vivo with the final aim of providing new translational insights into therapeutic approaches to help patients.
Collapse
Affiliation(s)
- C Tulotta
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - S He
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - W van der Ent
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - L Chen
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - A Groenewoud
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - H P Spaink
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - B E Snaar-Jagalska
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|
45
|
Gaspar D, Zeugolis DI. Engineering in vitro complex pathophysiologies for drug discovery purposes. Drug Discov Today 2016; 21:1341-1344. [DOI: 10.1016/j.drudis.2016.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
46
|
Nijoukubo D, Tanaka Y, Okuno Y, Yin G, Kitazawa T, Peterson RE, Kubota A, Teraoka H. Protective effect of prostacyclin against pre-cardiac edema caused by 2,3,7,8-tetrachlorodibenzo-p-dioxin and a thromboxane receptor agonist in developing zebrafish. CHEMOSPHERE 2016; 156:111-117. [PMID: 27174823 DOI: 10.1016/j.chemosphere.2016.04.107] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 04/22/2016] [Accepted: 04/25/2016] [Indexed: 06/05/2023]
Abstract
The role of prostaglandin pathways has been suggested in some toxicological responses to dioxins. Cyclooxygenase type 2b (COX2b), thromboxane synthase, and the thromboxane receptor (TP) pathway have been implicated in mediating 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced pre-cardiac edema in developing zebrafish at 55 h post fertilization (hpf). Pre-cardiac edema refers to edema located in a small cavity between the heart and body wall of zebrafish eleutheroembryos. In the present study, we assessed the role of prostacyclin, which counteracts some biological effects of thromboxane, in TCDD-induced pre-cardiac edema. Pre-cardiac edema induced by TCDD exposure (0.5 and 1 ppb) beginning at 24 hpf was markedly inhibited by exposure to beraprost (5 and 10 μM), a prostacyclin receptor (IP) agonist, beginning at 33 hpf. The preventive effect of beraprost was reduced by exposure to CAY10441 (10 μM), an IP antagonist starting at 33 hpf. Knockdowns of the IP receptor (IP-KD) with two different morpholinos caused edema by themselves and enhanced pre-cardiac edema caused by the low concentration of TCDD (0.5 ppb). On the other hand, short exposure beginning at 48 hpf to U46619 (7.5-30 μM), a thromboxane receptor agonist caused pre-cardiac edema, which was inhibited by exposure beginning at 48 hpf to both ICI-192,605 (24 μM), a TP antagonist, and beraprost. Expression of prostacyclin synthase was increased from fertilization, plateaued by 48 hpf, and was maintained until at least 96 hpf. Overall, the results demonstrate a preventive effect of prostacyclin on TCDD-induced pre-cardiac edema in developing zebrafish.
Collapse
Affiliation(s)
- Daisuke Nijoukubo
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Yasuaki Tanaka
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Yuki Okuno
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Guojun Yin
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan; Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| | - Takio Kitazawa
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | | | - Akira Kubota
- Diagnostic Center for Animal Health and Food Safety, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Hiroki Teraoka
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan.
| |
Collapse
|
47
|
Rezzola S, Paganini G, Semeraro F, Presta M, Tobia C. Zebrafish ( Danio rerio ) embryo as a platform for the identification of novel angiogenesis inhibitors of retinal vascular diseases. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1291-6. [DOI: 10.1016/j.bbadis.2016.04.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 04/05/2016] [Accepted: 04/11/2016] [Indexed: 12/26/2022]
|
48
|
Planchart A, Mattingly CJ, Allen D, Ceger P, Casey W, Hinton D, Kanungo J, Kullman SW, Tal T, Bondesson M, Burgess SM, Sullivan C, Kim C, Behl M, Padilla S, Reif DM, Tanguay RL, Hamm J. Advancing toxicology research using in vivo high throughput toxicology with small fish models. ALTEX 2016; 33:435-452. [PMID: 27328013 PMCID: PMC5270630 DOI: 10.14573/altex.1601281] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 05/31/2016] [Indexed: 12/18/2022]
Abstract
Small freshwater fish models, especially zebrafish, offer advantages over traditional rodent models, including low maintenance and husbandry costs, high fecundity, genetic diversity, physiology similar to that of traditional biomedical models, and reduced animal welfare concerns. The Collaborative Workshop on Aquatic Models and 21st Century Toxicology was held at North Carolina State University on May 5-6, 2014, in Raleigh, North Carolina, USA. Participants discussed the ways in which small fish are being used as models to screen toxicants and understand mechanisms of toxicity. Workshop participants agreed that the lack of standardized protocols is an impediment to broader acceptance of these models, whereas development of standardized protocols, validation, and subsequent regulatory acceptance would facilitate greater usage. Given the advantages and increasing application of small fish models, there was widespread interest in follow-up workshops to review and discuss developments in their use. In this article, we summarize the recommendations formulated by workshop participants to enhance the utility of small fish species in toxicology studies, as well as many of the advances in the field of toxicology that resulted from using small fish species, including advances in developmental toxicology, cardiovascular toxicology, neurotoxicology, and immunotoxicology. We alsoreview many emerging issues that will benefit from using small fish species, especially zebrafish, and new technologies that will enable using these organisms to yield results unprecedented in their information content to better understand how toxicants affect development and health.
Collapse
Affiliation(s)
- Antonio Planchart
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
| | - Carolyn J. Mattingly
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
| | - David Allen
- Integrated Laboratory Systems, Inc., Research Triangle Park, NC, USA
| | - Patricia Ceger
- Integrated Laboratory Systems, Inc., Research Triangle Park, NC, USA
| | - Warren Casey
- National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - David Hinton
- Nicholas School of the Environment, Duke University, Durham, NC, USA
| | - Jyotshna Kanungo
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Seth W. Kullman
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
| | - Tamara Tal
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Maria Bondesson
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, USA
| | | | - Con Sullivan
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME, USA
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Carol Kim
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME, USA
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Mamta Behl
- Division of National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Stephanie Padilla
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - David M. Reif
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
| | - Robert L. Tanguay
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR, USA
| | - Jon Hamm
- Integrated Laboratory Systems, Inc., Research Triangle Park, NC, USA
| |
Collapse
|
49
|
Yang S, Ott CJ, Rossmann MP, Superdock M, Zon LI, Zhou Y. Chromatin immunoprecipitation and an open chromatin assay in zebrafish erythrocytes. Methods Cell Biol 2016; 135:387-412. [PMID: 27443937 DOI: 10.1016/bs.mcb.2016.04.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Zebrafish is an excellent genetic and developmental model for the study of vertebrate development and disease. Its ability to produce an abundance of transparent, externally developed embryos has facilitated large-scale genetic and chemical screens for the identification of critical genes and chemical factors that modulate developmental pathways. These studies can have profound implications for the diagnosis and treatment of a variety of human diseases. Recent advancements in molecular and genomic studies have provided valuable tools and resources for comprehensive and high-resolution analysis of epigenomes during cell specification and lineage differentiation throughout development. In this chapter, we describe two simple methods to evaluate protein-DNA interaction and chromatin architecture in erythrocytes from adult zebrafish. These are chromatin immunoprecipitation coupled with next-generation sequencing (ChIP-seq) and an assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq). These techniques, together with gene expression profiling, are useful for analyzing epigenomic regulation of cell specification, differentiation, and function during zebrafish development in both normal and disease models.
Collapse
Affiliation(s)
- S Yang
- Boston Children's Hospital, Boston, MA, United States; Dana Farber Cancer Institute, Harvard Stem Cell Institute, Boston, MA, United States; Harvard Medical School and Howard Hughes Medical Institute, Boston, MA, United States
| | - C J Ott
- Dana Farber Cancer Institute, Harvard Stem Cell Institute, Boston, MA, United States
| | - M P Rossmann
- Harvard University, Harvard, Cambridge, MA, United States
| | - M Superdock
- Boston Children's Hospital, Boston, MA, United States; Dana Farber Cancer Institute, Harvard Stem Cell Institute, Boston, MA, United States; Harvard Medical School and Howard Hughes Medical Institute, Boston, MA, United States
| | - L I Zon
- Boston Children's Hospital, Boston, MA, United States; Dana Farber Cancer Institute, Harvard Stem Cell Institute, Boston, MA, United States; Harvard Medical School and Howard Hughes Medical Institute, Boston, MA, United States; Harvard University, Harvard, Cambridge, MA, United States
| | - Y Zhou
- Boston Children's Hospital, Boston, MA, United States; Dana Farber Cancer Institute, Harvard Stem Cell Institute, Boston, MA, United States; Harvard Medical School and Howard Hughes Medical Institute, Boston, MA, United States; Harvard University, Harvard, Cambridge, MA, United States
| |
Collapse
|
50
|
Chávez MN, Aedo G, Fierro FA, Allende ML, Egaña JT. Zebrafish as an Emerging Model Organism to Study Angiogenesis in Development and Regeneration. Front Physiol 2016; 7:56. [PMID: 27014075 PMCID: PMC4781882 DOI: 10.3389/fphys.2016.00056] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 02/05/2016] [Indexed: 01/04/2023] Open
Abstract
Angiogenesis is the process through which new blood vessels are formed from preexisting ones and plays a critical role in several conditions including embryonic development, tissue repair and disease. Moreover, enhanced therapeutic angiogenesis is a major goal in the field of regenerative medicine and efficient vascularization of artificial tissues and organs is one of the main hindrances in the implementation of tissue engineering approaches, while, on the other hand, inhibition of angiogenesis is a key therapeutic target to inhibit for instance tumor growth. During the last decades, the understanding of cellular and molecular mechanisms involved in this process has been matter of intense research. In this regard, several in vitro and in vivo models have been established to visualize and study migration of endothelial progenitor cells, formation of endothelial tubules and the generation of new vascular networks, while assessing the conditions and treatments that either promote or inhibit such processes. In this review, we address and compare the most commonly used experimental models to study angiogenesis in vitro and in vivo. In particular, we focus on the implementation of the zebrafish (Danio rerio) as a model to study angiogenesis and discuss the advantages and not yet explored possibilities of its use as model organism.
Collapse
Affiliation(s)
- Myra N Chávez
- Department of Plastic Surgery and Hand Surgery, University Hospital rechts der Isar, Technische Universität MünchenMunich, Germany; Department of Biology, FONDAP Center for Genome Regulation, Faculty of Science, Universidad de ChileSantiago, Chile; Department of Biochemistry and Molecular Biology, FONDAP Advanced Center for Chronic Diseases (ACCDiS) and Center for Molecular Studies of the Cell (CEMC), Faculty of Chemical and Pharmaceutical Sciences, Faculty of Medicine, University of ChileSantiago, Chile
| | - Geraldine Aedo
- Department of Biology, FONDAP Center for Genome Regulation, Faculty of Science, Universidad de Chile Santiago, Chile
| | - Fernando A Fierro
- Department of Cell Biology and Human Anatomy, University of California Davis, Sacramento, CA, USA
| | - Miguel L Allende
- Department of Biology, FONDAP Center for Genome Regulation, Faculty of Science, Universidad de Chile Santiago, Chile
| | - José T Egaña
- Institute for Medical and Biological Engineering, Schools of Engineering, Biological Sciences and Medicine, Pontifícia Universidad Católica de Chile Santiago, Chile
| |
Collapse
|