1
|
Huang R, Liu Z, Pan Y, Ma Z, Wang H, Wan B, Li J, Chang J. Mechanistic insight into the neurodevelopmental toxicity of the novel pesticide pyrifluquinazon (PFQ) and its major metabolite in early-life stage zebrafish. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 366:125469. [PMID: 39643230 DOI: 10.1016/j.envpol.2024.125469] [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: 08/03/2024] [Revised: 11/24/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Pyrifluquinazon (PFQ), a novel insecticide containing a heptafluoroisopropyl moiety, has seen increasing use. However, limited research has been conducted on the toxicological effects and mechanisms of PFQ in aquatic organisms. To investigate the toxicity and underlying mechanisms of PFQ and its primary metabolite dPFQ in aquatic organisms, morphological, behavioral, hormonal, multi-omics analyses, and molecular docking studies were conducted on zebrafish larvae after exposure. The results showed that both PFQ and dPFQ induced developmental abnormalities, behavioral impairment, hormonal disruptions, and alterations in neurologically related metabolites and gene expression in early-stage zebrafish. Notably, delayed retinal vascular development was observed, which is also likely linked to the neurodevelopmental toxicity. Subsequently, identification and relative quantification of PFQ metabolites suggested that its toxicity might be primarily attributed to dPFQ. Finally, an Adverse Outcome Pathway (AOP) was proposed, initiating with the binding of dPFQ to the TRPV4 protein and ultimately leading to neurodevelopmental toxicity. This study delineated the neurodevelopmental toxicity of PFQ and its toxicological mechanisms in zebrafish, emphasizing the hazards posed by pesticide metabolites to non-target organisms and highlighting inherent limitations of extrapolating in vitro toxicity experiments.
Collapse
Affiliation(s)
- Rui Huang
- Laboratory for Chemical Environmental Risk Assessment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Shuangqing RD 18, Beijing, 100085, China; University of Chinese Academy of Sciences, Yuquan RD 19 a, Beijing, 100049, China
| | - Zijun Liu
- Laboratory for Chemical Environmental Risk Assessment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Shuangqing RD 18, Beijing, 100085, China
| | - Yunrui Pan
- Laboratory for Chemical Environmental Risk Assessment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Shuangqing RD 18, Beijing, 100085, China; University of Chinese Academy of Sciences, Yuquan RD 19 a, Beijing, 100049, China
| | - Zheng Ma
- Laboratory for Chemical Environmental Risk Assessment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Shuangqing RD 18, Beijing, 100085, China; University of Chinese Academy of Sciences, Yuquan RD 19 a, Beijing, 100049, China
| | - Huili Wang
- Laboratory for Chemical Environmental Risk Assessment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Shuangqing RD 18, Beijing, 100085, China
| | - Bin Wan
- Laboratory for Chemical Environmental Risk Assessment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Shuangqing RD 18, Beijing, 100085, China; University of Chinese Academy of Sciences, Yuquan RD 19 a, Beijing, 100049, China
| | - Jianzhong Li
- Laboratory for Chemical Environmental Risk Assessment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Shuangqing RD 18, Beijing, 100085, China
| | - Jing Chang
- Laboratory for Chemical Environmental Risk Assessment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Shuangqing RD 18, Beijing, 100085, China.
| |
Collapse
|
2
|
Wu H, Zhao G, Feng W, Yang C, Jiang Y. Fluconazole induces cardiovascular toxicity in zebrafish by promoting oxidative stress, apoptosis, and disruption of key developmental genes. Chem Biol Interact 2025; 408:111391. [PMID: 39828185 DOI: 10.1016/j.cbi.2025.111391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/28/2024] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
This study systematically evaluated the toxic effects of fluconazole on the cardiovascular development of zebrafish. Zebrafish embryos were treated with different concentrations of fluconazole (200, 400, and 800 μg/ml) to observe its impact on heart development, reactive oxygen species (ROS) generation, apoptosis, and hemoglobin production. The results showed that as the concentration of fluconazole increased, significant changes in zebrafish heart structure were observed, along with a notable reduction in heart rate. Pericardial edema and cardiac morphological abnormalities were particularly prominent in the high-dose group. In addition, fluconazole treatment significantly increased ROS levels and induced apoptosis in cardiac cells. Enzyme-linked immunosorbent assay (ELISA) results showed that fluconazole treatment significantly increased the malondialdehyde (MDA) content and reduced superoxide dismutase (SOD) and catalase (CAT) activity, suggesting that oxidative stress and cell death may play a key role in its cardiotoxicity. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis revealed that fluconazole treatment significantly affected the expression of several key genes related to heart development and function, particularly cardiac myosin light chain 2 (cmlc2), ventricular myosin heavy chain (vmhc), and myosin heavy chain 6 (myh6), whose expression changes were closely associated with alterations in heart morphology and function. Transcriptomic analysis showed that several signaling pathways related to cardiac development, apoptosis, and metabolism were affected. In summary, this study reveals the multifaceted cardiotoxic mechanisms of fluconazole in zebrafish and provides new insights into drug safety assessment.
Collapse
Affiliation(s)
- Hanzhi Wu
- Department of Cardiology, Wuxi No.2 People's Hospital, The Affiliated Wuxi Clinical College of Nantong University, Jiangsu, 214000, China
| | - Gang Zhao
- Wuxi Ninth People's Hospital Affiliated to Soochow University, Jiangsu, 214000, China
| | - Wubing Feng
- Department of Orthopedics, Wuxi No.2 People's Hospital, The Affiliated Wuxi Clinical College of Nantong University, Jiangsu, 214000, China
| | - Chenjian Yang
- Department of Cardiology, Wuxi No.2 People's Hospital, The Affiliated Wuxi Clinical College of Nantong University, Jiangsu, 214000, China.
| | - Yu Jiang
- Department of General Practice, Wuxi No.2 People's Hospital, The Affiliated Wuxi Clinical College of Nantong University, Jiangsu, 214000, China.
| |
Collapse
|
3
|
Zheng N, Liao T, Zhang C, Zhang Z, Yan S, Xi X, Ruan F, Yang C, Zhao Q, Deng W, Huang J, Huang Z, Chen Z, Wang X, Qu Q, Zuo Z, He C. Quantum Dots-caused Retinal Degeneration in Zebrafish Regulated by Ferroptosis and Mitophagy in Retinal Pigment Epithelial Cells through Inhibiting Spliceosome. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406343. [PMID: 39420512 PMCID: PMC11633537 DOI: 10.1002/advs.202406343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/23/2024] [Indexed: 10/19/2024]
Abstract
Quantum dots (QDs) are widely used, but their health impact on the visual system is little known. This study aims to elucidate the effects and mechanisms of typical metallic QDs on retinas using zebrafish. Comprehensive histology, imaging, and bulk RNA sequencing reveal that InP/ZnS QDs cause retinal degeneration. Furthermore, single-cell RNA-seq reveals a reduction in the number of retinal pigment epithelial cells (RPE) and short-wave cone UV photoreceptor cells (PR(UV)), accompanied by an increase in middle- and long-wave cone red, green, and blue photoreceptor cells [PR(RGB)]. Mechanistically, after endocytosis by RPE, InP/ZnS QDs inhibit the expression of splicing factor prpf8, resulting in gpx4b mRNA unsplicing, which finally decrease glutathione and induce ferroptosis and mitophagy. The decrease of RPE fails to engulf the damaged outer segments of PR, possibly promoting the differentiation of PR(UV) to PR(RGB). Knockout prpf8 or gpx4b with CRISPR/Cas9 system, the retinal damage is also observed. Whereas, overexpression of prpf8 or gpx4b, or supplement of glutathione can rescue the retinal degenerative damage caused by InP/ZnS QDs. In conclusion, this study illustrates the potential health risks of InP/ZnS QDs on eye development and provides valuable insights into the underlying mechanisms of InP/ZnS QDs-caused retinal degeneration.
Collapse
Affiliation(s)
- Naying Zheng
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Tingting Liao
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Chuchu Zhang
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Zheyang Zhang
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Sen Yan
- Department of ChemistryState Key Laboratory of Physical Chemistry of Solid SurfacesCollaborative Innovation Center of Chemistry for Energy Materials (i‐ChEM)Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM)College of Chemistry and Chemical EngineeringXiamen UniversityXiamen361005China
| | - Xiaohan Xi
- Department of ChemistryState Key Laboratory of Physical Chemistry of Solid SurfacesCollaborative Innovation Center of Chemistry for Energy Materials (i‐ChEM)Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM)College of Chemistry and Chemical EngineeringXiamen UniversityXiamen361005China
| | - Fengkai Ruan
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Chunyan Yang
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Qingliang Zhao
- State Key Laboratory of Vaccines for Infectious DiseasesCenter for Molecular Imaging and Translational MedicineXiang An Biomedicine LaboratorySchool of Public HealthXiamen UniversityXiamenFujian361005China
| | - Wenbo Deng
- Key Laboratory of Reproductive Health ResearchFujian Province UniversitySchool of MedicineXiamen UniversityXiamenFujian361005China
| | - Jialiang Huang
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Zi‐Tao Huang
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk ControlGuangdong‐Hong Kong‐Macao Joint Laboratory for Contaminants Exposure and HealthSchool of Environmental Science and EngineeringGuangdong University of TechnologyGuangzhou510006China
| | - Zhi‐Feng Chen
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk ControlGuangdong‐Hong Kong‐Macao Joint Laboratory for Contaminants Exposure and HealthSchool of Environmental Science and EngineeringGuangdong University of TechnologyGuangzhou510006China
| | - Xiang Wang
- Department of ChemistryState Key Laboratory of Physical Chemistry of Solid SurfacesCollaborative Innovation Center of Chemistry for Energy Materials (i‐ChEM)Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM)College of Chemistry and Chemical EngineeringXiamen UniversityXiamen361005China
| | - Qingming Qu
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Zhenghong Zuo
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Chengyong He
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| |
Collapse
|
4
|
Santos AL, Rodrigues LC, Rodrigues CC, Cirqueira F, Malafaia G, Rocha TL. Polystyrene nanoplastics induce developmental impairments and vasotoxicity in zebrafish (Danio rerio). JOURNAL OF HAZARDOUS MATERIALS 2024; 464:132880. [PMID: 37956561 DOI: 10.1016/j.jhazmat.2023.132880] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023]
Abstract
The exponential use of plastics has significantly increased environmental pollution by nanoplastics (NPs). In the aquatic environment, NPs interact and bioaccumulate in the biota, posing a potential ecotoxicological risk. The present study investigated the developmental toxicity, vasotoxicity, cytotoxicity, ROS induction, and behavioral impairments in zebrafish (Danio rerio) exposed to environmentally relevant polystyrene NPs (PS-NPs) concentrations (0.04, 34 ng L-1, and 34 μg L-1) for 144 h through multiple biomarkers response (mortality, frequency of spontaneous contractions, heart rate, and morphological changes). Furthermore, vasotoxicity (head, yolk sac, tail, and branchial vessels) was evaluated using the transgenic zebrafish tg(Fli1:eGFP). Results showed that PS-NPs interacted mainly with zebrafish chorion, gills, tail, and larvae head. PS-NPs at 34 ng L-1 and 34 μg L-1 induced neurotoxicity (decreased frequency of spontaneous contractions), cardiotoxicity (bradycardia), and morphological changes in the eyes and head, indicating that PS-NPs induce developmental impairments in zebrafish. In addition, cytotoxicity in the caudal region (34 ng L-1), ROS production, decreased mean swimming speed, and distance covered were observed in all tested concentrations. PS-NPs also induced vasotoxicity (yolk sac region) in transgenic zebrafish. Overall, the present study demonstrates the harmful effects of PS-NPs on the early developmental stages of freshwater fish, indicating their environmental risk.
Collapse
Affiliation(s)
- Andressa Liberal Santos
- Laboratory of Environmental Biotechnology and Ecotoxicology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Brazil
| | - Laura Carvalho Rodrigues
- Laboratory of Environmental Biotechnology and Ecotoxicology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Brazil
| | - Cândido Carvalho Rodrigues
- Laboratory of Environmental Biotechnology and Ecotoxicology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Brazil
| | - Felipe Cirqueira
- Laboratory of Environmental Biotechnology and Ecotoxicology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Brazil
| | - Guilherme Malafaia
- Laboratory of Toxicology Applied to the Environment, Goiano Federal Institute, Urutaí, Goiás, Brazil
| | - Thiago Lopes Rocha
- Laboratory of Environmental Biotechnology and Ecotoxicology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Brazil.
| |
Collapse
|
5
|
Lee H, An G, Park J, You J, Song G, Lim W. Mevinphos induces developmental defects via inflammation, apoptosis, and altered MAPK and Akt signaling pathways in zebrafish. Comp Biochem Physiol C Toxicol Pharmacol 2024; 275:109768. [PMID: 37858660 DOI: 10.1016/j.cbpc.2023.109768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/11/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023]
Abstract
Mevinphos, an organophosphate insecticide, is widely used to control pests and enhance crop yield. Because of its high solubility, it can easily flow into water and threaten the aquatic environment, and it is known to be hazardous to non-target organisms. However, little is known about its developmental toxicity and the underlying toxic mechanisms. In this study, we utilized zebrafish, which is frequently used for toxicological research to estimate the toxicity in other aquatic organisms or vertebrates including humans, to elucidate the developmental defects induced by mevinphos. Here, we observed that mevinphos induced various phenotypical abnormalities, such as diminished eyes and head sizes, shortened body length, loss of swim bladder, and increased pericardiac edema. Also, exposure to mevinphos triggered inflammation, apoptosis, and DNA fragmentation in zebrafish larvae. In addition, MAPK and Akt signaling pathways, which control apoptosis, inflammation, and proper development of various organs, were also altered by the treatment of mevinphos. Furthermore, these factors induced various organ defects which were confirmed by various transgenic models. We identified neuronal toxicity through transgenic olig2:dsRed zebrafish, cardiovascular toxicity through transgenic fli1:eGFP zebrafish, and hepatotoxicity and pancreatic toxicity through transgenic lfabp:dsRed;elastase:GFP zebrafish. Overall, our results elucidated the developmental toxicities of mevinphos in zebrafish and provided the parameters for the assessment of toxicities in aquatic environments.
Collapse
Affiliation(s)
- Hojun Lee
- 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
| | - Junho Park
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Jeankyoung You
- 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, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
6
|
Zhang SY, Gan X, Shen B, Jiang J, Shen H, Lei Y, Liang Q, Bai C, Huang C, Wu W, Guo Y, Song Y, Chen J. 6PPD and its metabolite 6PPDQ induce different developmental toxicities and phenotypes in embryonic zebrafish. JOURNAL OF HAZARDOUS MATERIALS 2023; 455:131601. [PMID: 37182464 DOI: 10.1016/j.jhazmat.2023.131601] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 05/05/2023] [Accepted: 05/07/2023] [Indexed: 05/16/2023]
Abstract
The automobile tire antioxidant N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine (6PPD) and its quinone metabolite 6PPDQ have recently received much attention for their acute aquatic toxicity. The present study investigated the mechanistic developmental toxicity of 6PPD and 6PPDQ in embryonic zebrafish. Neither compound induced significant mortality but significantly decreased spontaneous embryo movement and heart rate. Both compounds induced malformations with different phenotypes; the 6PPD-exposed larvae manifested a myopia-like phenotype with a convex eyeball and fusion vessels, while the 6PPDQ-exposed embryonic zebrafish manifested enlarged intestine and blood-coagulated gut, activated neutrophils, and overexpressed enteric neurons. mRNA-Seq and quantitative real-time PCR assays showed that 6PPD- and 6PPDQ-induced distinct differential gene expression aligned with their toxic phenotype. 6PPD activated the retinoic acid metabolic gene cyp26a, but 6PPDQ activated adaptive cellular response to xenobiotics gene cyp1a. 6PPD suppressed the gene expression of the eye involved in retinoic acid metabolism, phototransduction, photoreceptor function and visual perception. In contrast, 6PPDQ perturbed genes involved in inward rectifier K+ and voltage-gated ion channels activities, K+ import across the plasma membrane, iron ion binding, and intestinal immune network for IgA production. The current study advances the present understanding the reason of why many fish species are so adversely impacted by 6PPD and 6PPDQ.
Collapse
Affiliation(s)
- Shu-Yun Zhang
- Zhejiang Provincial Key Laboratory of Watershed Sciences and Health, School of Public health and Management, Wenzhou Medical University, Wenzhou 325035, PR China; School of Medicine, Taizhou University, Taizhou, 318000, PR China
| | - Xiufeng Gan
- Zhejiang Provincial Key Laboratory of Watershed Sciences and Health, School of Public health and Management, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Baoguo Shen
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, PR China
| | - Jian Jiang
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, PR China
| | - Huimin Shen
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 511443, PR China
| | - Yuhang Lei
- Zhejiang Provincial Key Laboratory of Watershed Sciences and Health, School of Public health and Management, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Qiuju Liang
- Zhejiang Provincial Key Laboratory of Watershed Sciences and Health, School of Public health and Management, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Chenglian Bai
- Zhejiang Provincial Key Laboratory of Watershed Sciences and Health, School of Public health and Management, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Changjiang Huang
- Zhejiang Provincial Key Laboratory of Watershed Sciences and Health, School of Public health and Management, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Wencan Wu
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, PR China
| | - Ying Guo
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 511443, PR China.
| | - Yang Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China.
| | - Jiangfei Chen
- Zhejiang Provincial Key Laboratory of Watershed Sciences and Health, School of Public health and Management, Wenzhou Medical University, Wenzhou 325035, PR China; State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China.
| |
Collapse
|
7
|
Ma Y, Zhu Q, Luo S, Zhang F, Liu L, Mengxue Z, Zhang Z, Cao X, Qiu X, Zeng X, Ji D, Li C, Zhong X, Wang J, Wei Y. Environmentally relevant concentrations of fipronil selectively disrupt venous vessel development in zebrafish embryos/larvae. CHEMOSPHERE 2023:139146. [PMID: 37290517 DOI: 10.1016/j.chemosphere.2023.139146] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/29/2023] [Accepted: 06/04/2023] [Indexed: 06/10/2023]
Abstract
The pesticide fipronil is widely dispersed in aquatic environments and frequently detected in the general population. Although the adverse effects on embryonic growth by fipronil exposure have been extensively documented, the early responses for its developmental toxicity are largely unknown. In the present study, we explored the sensitive targets of fipronil, focusing on vascular injury using zebrafish embryos/larvae and cultured human endothelial cells. Exposure to 5-500 μg/L fipronil at the early stage impeded the growth of sub-intestinal venous plexus (SIVP), caudal vein plexus (CVP), and common cardinal veins (CCV). The damages on venous vessels occurred at exposure to the environmentally relevant concentration as low as 5 μg/L fipronil, whereas no significant change was observed in general toxicity indexes. In contrast, vascular development of the dorsal aorta (DA) or intersegmental artery (ISA) was not affected. In addition, the mRNA levels of vascular markers and vessel type-specific function genes exhibited significant decreases in venous genes, including nr2f2, ephb4a, and flt4, but no appreciable change in arterial genes. Likewise, the more pronounced changes in cell death and cytoskeleton disruption were shown in human umbilical vein endothelial cells as compared with human aortic endothelial cells. Furthermore, molecular docking supported a stronger affinity of fipronil and its metabolites to the proteins correlated with venous development, such as BMPR2 and SMARCA4. These results reveal the heterogeneity in developing vasculature responsive to fipronil's exposure. The preferential impacts on the veins confer higher sensitivity, allowing them to be appropriate targets for monitoring fipronil's developmental toxicity.
Collapse
Affiliation(s)
- Ya Ma
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qicheng Zhu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shili Luo
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Fenghong Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China
| | - Lei Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China
| | - Zhi Mengxue
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China
| | - Zhuyi Zhang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiaolian Cao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xuelin Qiu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiangyu Zeng
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Di Ji
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Chenxin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiali Zhong
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jianshe Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China.
| | - Yanhong Wei
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
8
|
Fusco P, Fietta A, Esposito MR, Zanella L, Micheli S, Bastianello A, Bova L, Borile G, Germano G, Cimetta E. miR-210-3p enriched extracellular vesicles from hypoxic neuroblastoma cells stimulate migration and invasion of target cells. Cell Biosci 2023; 13:89. [PMID: 37202777 DOI: 10.1186/s13578-023-01045-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/03/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND Tumor hypoxia stimulates release of extracellular vesicles (EVs) that facilitate short- and long-range intercellular communication and metastatization. Albeit hypoxia and EVs release are known features of Neuroblastoma (NB), a metastasis-prone childhood malignancy of the sympathetic nervous system, whether hypoxic EVs can facilitate NB dissemination is unclear. METHODS Here we isolated and characterized EVs from normoxic and hypoxic NB cell culture supernatants and performed microRNA (miRNA) cargo analysis to identify key mediators of EVs biological effects. We then validated if EVs promote pro-metastatic features both in vitro and in an in vivo zebrafish model. RESULTS EVs from NB cells cultured at different oxygen tensions did not differ for type and abundance of surface markers nor for biophysical properties. However, EVs derived from hypoxic NB cells (hEVs) were more potent than their normoxic counterpart in inducing NB cells migration and colony formation. miR-210-3p was the most abundant miRNA in the cargo of hEVs; mechanistically, overexpression of miR-210-3p in normoxic EVs conferred them pro-metastatic features, whereas miR-210-3p silencing suppressed the metastatic ability of hypoxic EVs both in vitro and in vivo. CONCLUSION Our data identify a role for hypoxic EVs and their miR-210-3p cargo enrichment in the cellular and microenvironmental changes favoring NB dissemination.
Collapse
Affiliation(s)
- Pina Fusco
- Department of Industrial Engineering (DII), University of Padua, Via Marzolo 9, 35131, Padua, Italy
- Fondazione Istituto Di Ricerca Pediatrica Città Della Speranza (IRP), Corso Stati Uniti 4, 35127, Padua, Italy
| | - Anna Fietta
- Fondazione Istituto Di Ricerca Pediatrica Città Della Speranza (IRP), Corso Stati Uniti 4, 35127, Padua, Italy
| | - Maria Rosaria Esposito
- Department of Industrial Engineering (DII), University of Padua, Via Marzolo 9, 35131, Padua, Italy
- Fondazione Istituto Di Ricerca Pediatrica Città Della Speranza (IRP), Corso Stati Uniti 4, 35127, Padua, Italy
| | - Luca Zanella
- Department of Industrial Engineering (DII), University of Padua, Via Marzolo 9, 35131, Padua, Italy
- Fondazione Istituto Di Ricerca Pediatrica Città Della Speranza (IRP), Corso Stati Uniti 4, 35127, Padua, Italy
| | - Sara Micheli
- Department of Industrial Engineering (DII), University of Padua, Via Marzolo 9, 35131, Padua, Italy
- Fondazione Istituto Di Ricerca Pediatrica Città Della Speranza (IRP), Corso Stati Uniti 4, 35127, Padua, Italy
| | - Angelica Bastianello
- Fondazione Istituto Di Ricerca Pediatrica Città Della Speranza (IRP), Corso Stati Uniti 4, 35127, Padua, Italy
| | - Lorenzo Bova
- Department of Industrial Engineering (DII), University of Padua, Via Marzolo 9, 35131, Padua, Italy
| | - Giulia Borile
- Fondazione Istituto Di Ricerca Pediatrica Città Della Speranza (IRP), Corso Stati Uniti 4, 35127, Padua, Italy
| | - Giuseppe Germano
- Fondazione Istituto Di Ricerca Pediatrica Città Della Speranza (IRP), Corso Stati Uniti 4, 35127, Padua, Italy
| | - Elisa Cimetta
- Department of Industrial Engineering (DII), University of Padua, Via Marzolo 9, 35131, Padua, Italy.
- Fondazione Istituto Di Ricerca Pediatrica Città Della Speranza (IRP), Corso Stati Uniti 4, 35127, Padua, Italy.
| |
Collapse
|
9
|
Naija A, Yalcin HC. Evaluation of cadmium and mercury on cardiovascular and neurological systems: Effects on humans and fish. Toxicol Rep 2023; 10:498-508. [PMID: 37396852 PMCID: PMC10313869 DOI: 10.1016/j.toxrep.2023.04.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/09/2023] [Accepted: 04/17/2023] [Indexed: 07/04/2023] Open
Abstract
Chemicals are at the top of public health concerns and metals have received much attention in terms of toxicological studies. Cadmium (Cd) and mercury (Hg) are among the most toxic heavy metals and are widely distributed in the environment. They are considered important factors involved in several organ disturbances. Heart and brain tissues are not among the first exposure sites to Cd and Hg but they are directly affected and may manifest intoxication reactions leading to death. Many cases of human intoxication with Cd and Hg showed that these metals have potential cardiotoxic and neurotoxic effects. Human exposure to heavy metals is through fish consumption which is considered as an excellent source of human nutrients. In the current review, we will summarize the most known cases of human intoxication with Cd and Hg, highlight their toxic effects on fish, and investigate the common signal pathways of both Cd and Hg to affect heart and brain tissues. Also, we will present the most common biomarkers used in the assessment of cardiotoxicity and neurotoxicity using Zebrafish model.
Collapse
|
10
|
Shang Y, Zhang S, Cheng Y, Feng G, Dong Y, Li H, Fan S. Tetrabromobisphenol a exacerbates the overall radioactive hazard to zebrafish (Danio rerio). ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 316:120424. [PMID: 36272602 DOI: 10.1016/j.envpol.2022.120424] [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: 08/12/2022] [Revised: 10/05/2022] [Accepted: 10/10/2022] [Indexed: 06/16/2023]
Abstract
The major health risks of dual exposure to two hazardous factors of plastics and radioactive contamination are obscure. In the present study, we systematically evaluated the combinational toxic effects of tetrabromobisphenol A (TBBPA), one of the most influential plastic ingredients, mainly from electronic wastes, and γ-irradiation in zebrafish for the first time. TBBPA (0.25 μg/mL for embryos and larvae, 300 μg/L for adults) contamination aggravated the radiation (6 Gy for embryos and larvae, 20 Gy for adults)-induced early dysplasia and aberrant angiogenesis of embryos, further impaired the locomotor vitality of irradiated larvae, and worsened the radioactive multiorganic histologic injury, neurobehavioural disturbances and dysgenesis of zebrafish adults as well as the inter-generational neurotoxicity in offspring. TBBPA exaggerated the radiative toxic effects not only by enhancing the inflammatory and apoptotic response but also by further unbalancing the endocrine system and disrupting the underlying gene expression profiles. In conclusion, TBBPA exacerbates radiation-induced injury in zebrafish, including embryos, larvae, adults and even the next generation. Our findings provide new insights into the toxicology of TBBPA and γ-irradiation, shedding light on the severity of cocontamination of MP components and radioactive substances and thereby inspiring novel remediation and rehabilitation strategies for radiation-injured aqueous organisms and radiotherapy patients.
Collapse
Affiliation(s)
- Yue Shang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 238 Baidi Road, 300192, Tianjin, China
| | - Shuqin Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 238 Baidi Road, 300192, Tianjin, China
| | - Yajia Cheng
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 238 Baidi Road, 300192, Tianjin, China
| | - Guoxing Feng
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 238 Baidi Road, 300192, Tianjin, China
| | - Yinping Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 238 Baidi Road, 300192, Tianjin, China
| | - Hang Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 238 Baidi Road, 300192, Tianjin, China
| | - Saijun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 238 Baidi Road, 300192, Tianjin, China.
| |
Collapse
|
11
|
A zebrafish HCT116 xenograft model to predict anandamide outcomes on colorectal cancer. Cell Death Dis 2022; 13:1069. [PMID: 36564370 PMCID: PMC9789132 DOI: 10.1038/s41419-022-05523-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 12/08/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
Colon cancer is one of the leading causes of death worldwide. In recent years, cannabinoids have been extensively studied for their potential anticancer effects and symptom management. Several in vitro studies reported anandamide's (AEA) ability to block cancer cell proliferation and migration, but evidence from in vivo studies is still lacking. Thus, in this study, the effects of AEA exposure in zebrafish embryos transplanted with HCT116 cells were evaluated. Totally, 48 hpf xenografts were exposed to 10 nM AEA, 10 nM AM251, one of the cannabinoid 1 receptor (CB1) antagonist/inverse agonists, and to AEA + AM251, to verify the specific effect of AEA treatment. AEA efficacy was evaluated by confocal microscopy, which demonstrated that these xenografts presented a smaller tumor size, reduced tumor angiogenesis, and lacked micrometastasis formation. To gain deeper evidence into AEA action, microscopic observations were completed by molecular analyses. RNA seq performed on zebrafish transcriptome reported the downregulation of genes involved in cell proliferation, angiogenesis, and the immune system. Conversely, HCT116 cell transcripts resulted not affected by AEA treatment. In vitro HCT116 culture, in fact, confirmed that AEA exposure did not affect cell proliferation and viability, thus suggesting that the reduced tumor size mainly depends on direct effects on the fish rather than on the transplanted cancer cells. AEA reduced cell proliferation and tumor angiogenesis, as suggested by socs3 and pcnp mRNAs and Vegfc protein levels, and exerted anti-inflammatory activity, as indicated by the reduction of il-11a, mhc1uba, and csf3b mRNA. Of note, are the results obtained in groups exposed to AM251, which presence nullifies AEA's beneficial effects. In conclusion, this study promotes the efficacy of AEA in personalized cancer therapy, as suggested by its ability to drive tumor growth and metastasis, and strongly supports the use of zebrafish xenograft as an emerging model platform for cancer studies.
Collapse
|
12
|
Silva Brito R, Canedo A, Farias D, Rocha TL. Transgenic zebrafish (Danio rerio) as an emerging model system in ecotoxicology and toxicology: Historical review, recent advances, and trends. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 848:157665. [PMID: 35907527 DOI: 10.1016/j.scitotenv.2022.157665] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 06/13/2022] [Accepted: 07/23/2022] [Indexed: 06/15/2023]
Abstract
Zebrafish (Danio rerio) is an alternative model system for drug screening, developing new products, and assessing ecotoxic effects of pollutants and biomonitor species in environmental risk assessment. However, the history and current use of transgenic zebrafish lines in ecotoxicology and toxicology studies remain poorly explored. Thus, the present study aimed to summarize and discuss the existing data in the literature about the applications of transgenic zebrafish lines in ecotoxicology and toxicology. The articles were analyzed according to publication year, journal, geographic distribution, and collaborations. Also, the bioassays were evaluated according to the tested chemical, transgenic lines, development stage, biomarkers, and exposure conditions (i.e., concentration, time, type, and route of exposure). Revised data showed that constitutive transgenic lines are the main type of transgenic used in the studies, besides most of uses embryos and larvae under static conditions. Tg(fli1: EGFP) was the main transgenic line, while the GFP and EGFP were the main reporter proteins. Transgenic zebrafish stands out in assessing vasotoxicity, neurotoxicity, systemic toxicity, hepatoxicity, endocrine disruption, cardiotoxicity, immunotoxicity, hematotoxicity, ototoxicity, and pancreotoxicity. This review showed that transgenic zebrafish lines are emerging as a suitable in vivo model system for assessing the mechanism of action and toxicity of chemicals and new biotechnology products, and the effects of traditional and emerging pollutants.
Collapse
Affiliation(s)
- Rafaella Silva Brito
- Laboratory of Environmental Biotechnology and Ecotoxicology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Aryelle Canedo
- Laboratory of Environmental Biotechnology and Ecotoxicology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Davi Farias
- Laboratory for Risk Assessment of Novel Technologies (LabRisk), Center of Exact and Natural Sciences, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | - Thiago Lopes Rocha
- Laboratory of Environmental Biotechnology and Ecotoxicology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil.
| |
Collapse
|
13
|
A low-molecular-weight chitosan fluorometric-based assay for evaluating antiangiogenic drugs. Int J Biol Macromol 2022; 224:927-937. [DOI: 10.1016/j.ijbiomac.2022.10.178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 11/05/2022]
|
14
|
Vinoth S, Selvaraj V, Venkatasubramanian H, Santhakumar K. A Simple Blood Vessel Imaging Protocol for Live Zebrafish Larva. Zebrafish 2022; 19:177-180. [DOI: 10.1089/zeb.2022.0023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- S. Vinoth
- Zebrafish Genetics Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, India
| | - Velanganni Selvaraj
- Zebrafish Genetics Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, India
| | - Hemagowri Venkatasubramanian
- Zebrafish Genetics Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, India
| | - Kirankumar Santhakumar
- Zebrafish Genetics Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, India
| |
Collapse
|
15
|
Tyrkalska SD, Martínez-López A, Arroyo AB, Martínez-Morcillo FJ, Candel S, García-Moreno D, Mesa-del-Castillo P, Cayuela ML, Mulero V. Differential proinflammatory activities of Spike proteins of SARS-CoV-2 variants of concern. SCIENCE ADVANCES 2022; 8:eabo0732. [PMID: 36112681 PMCID: PMC9481140 DOI: 10.1126/sciadv.abo0732] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 07/29/2022] [Indexed: 05/11/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic turned the whole world upside down in a short time. One of the main challenges faced has been to understand COVID-19-associated life-threatening hyperinflammation, the so-called cytokine storm syndrome (CSS). We report here the proinflammatory role of Spike (S) proteins from different severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern in zebrafish. We found that wild-type/Wuhan variant S1 (S1WT) promoted neutrophil and macrophage recruitment, local and systemic hyperinflammation, emergency myelopoiesis, and hemorrhages. In addition, S1γ was more proinflammatory S1δ was less proinflammatory than S1WT, and, notably, S1β promoted delayed and long-lasting inflammation. Pharmacological inhibition of the canonical inflammasome alleviated S1-induced inflammation and emergency myelopoiesis. In contrast, genetic inhibition of angiotensin-converting enzyme 2 strengthened the proinflammatory activity of S1, and angiotensin (1-7) fully rescued S1-induced hyperinflammation and hemorrhages. These results shed light into the mechanisms orchestrating the COVID-19-associated CSS and the host immune response to different SARS-CoV-2 S protein variants.
Collapse
Affiliation(s)
- Sylwia D. Tyrkalska
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alicia Martínez-López
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ana B. Arroyo
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Francisco J. Martínez-Morcillo
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sergio Candel
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Diana García-Moreno
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Pablo Mesa-del-Castillo
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
| | - María L. Cayuela
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
| | - Victoriano Mulero
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
16
|
Diao W, Qian Q, Sheng G, He A, Yan J, Dahlgren RA, Wang X, Wang H. Triclosan targets miR-144 abnormal expression to induce neurodevelopmental toxicity mediated by activating PKC/MAPK signaling pathway. JOURNAL OF HAZARDOUS MATERIALS 2022; 431:128560. [PMID: 35245871 DOI: 10.1016/j.jhazmat.2022.128560] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/16/2022] [Accepted: 02/22/2022] [Indexed: 06/14/2023]
Abstract
Although the previous research confirmed that triclosan (TCS) induced an estrogen effect by acting on a novel G-protein coupled estrogen-membrane receptor (GPER), the underlying mechanisms by which downstream pathways induce neurotoxicity remain unclear after TCS activation of GPER. By employing a series of techniques (Illumina miRNA-seq, RT-qPCR, and artificial intervention of miRNA expression), we screened out four important miRNAs, whose target genes were directly/indirectly involved in neurodevelopment and neurobehavior. Especially, the miR-144 up-regulation caused vascular malformation and severely affected hair-cell development and lateral-line-neuromast formation, thereby causing abnormal motor behavior. After microinjecting 1-2-cell embryos, the similar phenotypic malformations as those induced by TCS were observed, including aberrant neuromast, cuticular-plate development and motor behavior. By KEGG pathway enrichment analysis, these target genes were demonstrated to be mainly related to the PKC/MAPK signaling pathway. When a PKC inhibitor was used to suppress the PKC/MAPK pathway, a substantial alleviation of TCS-induced neurotoxicity was observed. Therefore, TCS acts on GPER to activate the downstream PKC/MAPK signaling pathway, further up-regulating miR-144 expression and causing abnormal modulation of these nerve-related genes to trigger neurodevelopmental toxicity. These findings unravel the molecular mechanisms of TCS-induced neurodegenerative diseases, and offer theoretical guidance for TCS-pollution early warning and management.
Collapse
Affiliation(s)
- Wenqi Diao
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, PR China; School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Qiuhui Qian
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, PR China
| | - Guangyao Sheng
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, PR China
| | - Anfei He
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, PR China
| | - Jin Yan
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, PR China
| | - Randy A Dahlgren
- Department of Land, Air and Water Resources, University of California, Davis, CA 95616, USA
| | - Xuedong Wang
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, PR China.
| | - Huili Wang
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, PR China; School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, PR China.
| |
Collapse
|
17
|
Stream A, Madigan CA. Zebrafish: an underutilized tool for discovery in host-microbe interactions. Trends Immunol 2022; 43:426-437. [PMID: 35527182 PMCID: PMC11302990 DOI: 10.1016/j.it.2022.03.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 03/31/2022] [Accepted: 03/31/2022] [Indexed: 12/31/2022]
Abstract
Zebrafish are relatively new to the field of host-pathogen interactions, although they have been a valuable vertebrate model for decades in developmental biology and neuroscience. Transparent zebrafish larvae have most components of the human innate immune system, and adult zebrafish also produce cells of the adaptive immune system. Recent discoveries using zebrafish infection models include mechanisms of pathogen survival and host cell sensing of microbes. These discoveries were enabled by zebrafish technology, which is constantly evolving and providing new opportunities for immunobiology research. Recent tools include CRISPR/Cas9 mutagenesis, in vivo biotinylation, and genetically encoded biosensors. We argue that the zebrafish model - which remains underutilized in immunology - provides fertile ground for a new understanding of host-microbe interactions in a transparent host.
Collapse
Affiliation(s)
- Alexandra Stream
- Department of Biological Sciences, University of California San Diego (UCSD), San Diego, CA, USA
| | - Cressida A Madigan
- Department of Biological Sciences, University of California San Diego (UCSD), San Diego, CA, USA.
| |
Collapse
|
18
|
5-Azacytidine Downregulates the Proliferation and Migration of Hepatocellular Carcinoma Cells In Vitro and In Vivo by Targeting miR-139-5p/ROCK2 Pathway. Cancers (Basel) 2022; 14:cancers14071630. [PMID: 35406401 PMCID: PMC8996928 DOI: 10.3390/cancers14071630] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/14/2022] [Accepted: 03/18/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary For hepatocellular carcinoma (HCC), the second most common cause of cancer-related death, effective therapeutic approaches are lacking. As aberrant gene methylation is a major contributor to the development of HCC, demethylating drugs such as 5-azacytidine (5-Aza) have been proposed. However, despite the potential efficacy of 5-Aza in HCC, most of its mechanisms of action are still unknown. Here, we investigate the phenotypic/molecular effects of 5-Aza with a focus on miR-139-5p. Using multiple in vitro and in vivo models of HCC, we show for the first time that 5-Aza can impair HCC development via upregulation of miR-139-5p, which in turn downregulates the ROCK2/cyclin D1/E2F1/cyclin B1 pro-proliferative pathway and the ROCK2/MMP-2 pro-migratory pathway. These observations elucidate the mechanisms of action of 5-Aza in HCC, strengthen its therapeutic potential, and provide novel information about the crosstalk among ROCK2/cyclin D1/E2F1/cyclin B1/MMP-2 in HCC. Abstract Background: For hepatocellular carcinoma (HCC), effective therapeutic approaches are lacking. As aberrant gene methylation is a major contributor to HCC development, demethylating drugs such as 5-azacytidine (5-Aza) have been proposed. As most 5-Aza mechanisms of action are unknown, we investigated its phenotypic/molecular effects. Methods: 5-Aza effects were examined in the human HCC cell lines JHH-6/HuH-7 and in the rat cell-line N1-S1. We also employed a xenograft mouse model (HuH-7), a zebrafish model (JHH-6), and an orthotopic syngeneic rat model (N1-S1) of HCC. Results: 5-Aza downregulated cell viability/growth/migration/adhesion by upregulating miR-139-5p, which in turn downregulated ROCK2/cyclin D1/E2F1 and increased p27kip1, resulting in G1/G0 cell accumulation. Moreover, a decrease in cyclin B1 and an increase in p27kip1 led to G2/M accumulation. Finally, we observed a decrease in MMP-2 levels, a stimulator of HCC cell migration. Aza effects were confirmed in the mouse model; in the zebrafish model, we also demonstrated the downregulation of tumor neo-angiogenesis, and in the orthotopic rat model, we observed impaired N1-S1 grafting in a healthy liver. Conclusion: We demonstrate for the first time that 5-Aza can impair HCC development via upregulation of miR-139-5p, which in turn impairs the ROCK2/cyclin D1/E2F1/cyclin B1 pro-proliferative pathway and the ROCK2/MMP-2 pro-migratory pathway. Thus, we provide novel information about 5-Aza mechanisms of action and deepen the knowledge about the crosstalk among ROCK2/cyclin D1/E2F1/cyclin B1/p27kip1/MMP-2 in HCC.
Collapse
|
19
|
Zheng YR, Fan CL, Chen Y, Quan JY, Shi LZ, Tian CY, Shang X, Xu NS, Ye WC, Yu LZ, Liu JS. Anti-inflammatory, anti-angiogenetic and antiviral activities of dammarane-type triterpenoid saponins from the roots of Panax notoginseng. Food Funct 2022; 13:3590-3602. [PMID: 35262135 DOI: 10.1039/d1fo04089h] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Panax notoginseng has been used both as a traditional medicine and as a functional food for hundreds of years in Asia. However, the active constituents from P. notoginseng and their pharmacologic properties still need to be further explored. In this study, one new dammarane-type triterpenoid saponin (1), along with fourteen known analogs (2-15) were isolated and identified from the roots of P. notoginseng. The anti-inflammatory, anti-angiogenetic and anti-dengue virus effects of these isolated compounds were further evaluated. Compounds 1, 3, 5-7 and 10-12 exerted anti-inflammatory effects in two different zebrafish inflammatory models. Among them, 11, with the most significant activities, alleviated the inflammatory response by blocking the MyD88/NF-κB and STAT3 pathways. Moreover, compound 15 showed anti-angiogenetic activities in Tg(fli1:EGFP) and Tg(flk1:GFP) zebrafish, while 3 and 5 only inhibited angiogenesis in Tg(fli1:EGFP) zebrafish. Additionally, compounds 1, 3, 6, 8, 9 and 12 suppressed the replication of dengue virus either at the viral adsorption and entry stages or at the intracellular replication step. In conclusion, these findings enrich knowledge of the diversity of saponins in P. notoginseng and suggest that the dammarane-type triterpenoid saponins from P. notoginseng may be developed as potential functional foods to treat inflammation, angiogenesis or dengue-related diseases.
Collapse
Affiliation(s)
- Yuan-Ru Zheng
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, P. R. China.
| | - Chun-Lin Fan
- Institute of Traditional Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, P. R. China
| | - Ye Chen
- Institute of Traditional Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, P. R. China
| | - Jing-Yu Quan
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, P. R. China.
| | - Ling-Zhu Shi
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, P. R. China.
| | - Chun-Yang Tian
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, P. R. China.
| | - Xiao Shang
- Institute of Traditional Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, P. R. China
| | - Ni-Shan Xu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, P. R. China.
| | - Wen-Cai Ye
- Institute of Traditional Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, P. R. China
| | - Lin-Zhong Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, P. R. China.
| | - Jun-Shan Liu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, P. R. China.
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| |
Collapse
|
20
|
Ji G, Gu J, Guo M, Zhou L, Wang Z, Shi L, Gu A. A systematic comparison of the developmental vascular toxicity of bisphenol A and its alternatives in vivo and in vitro. CHEMOSPHERE 2022; 291:132936. [PMID: 34798105 DOI: 10.1016/j.chemosphere.2021.132936] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/11/2021] [Accepted: 11/14/2021] [Indexed: 06/13/2023]
Abstract
Due to the potential toxicity of bisphenol A (BPA), several bisphenols (BPs), including bisphenol F (BPF), bisphenol S (BPS) and bisphenol AF (BPAF), have been gradually used as its main substitutes, and the levels of these alternatives in different environmental media have been constantly increasing. Although some previous studies have shown that bisphenol substitutes have similar or greater acute toxicity and estrogenic effects than BPA, comparative studies on the cardiovascular toxicity of BPs have not been evaluated. In this study, the developmental vascular toxicity of BPA and three predominant substitutes (BPF, BPS and BPAF) were evaluated using zebrafish embryos and human vascular endothelial cells (HUVECs). BP exposure at a sublethal concentration of 1/10 96 h median lethal concentration (96 h-LC50) significantly hindered intersegmental vessel (ISV) growth, delayed common cardinal vein (CCV) remodeling and decreased subintestinal vessels (SIVs) in Tg (fli1:EGFP) zebrafish embryos. Meanwhile, the results of the endothelial tube formation assay showed that in vitro angiogenesis was inhibited by BP exposure. Mechanistically, BP exposure increased oxidative stress characterized by a significant decrease in superoxide dismutase (SOD) and catalase (CAT) activity, accompanied by increased levels of malondialdehyde (MDA) and reactive oxygen species (ROS) in both zebrafish and HUVECs. Therefore, the vascular toxicity and oxidative stress potency of the BPs were compared and evaluated, ranking as follows: BPAF > BPF > BPA > BPS. To the best of our knowledge, the present work, for the first time, systematically provides direct evidence for BPA and its alternatives on developmental vascular toxicity in vitro and in vivo. Therefore, these findings will provide insight into the rational and safe application of BPA substitutes.
Collapse
Affiliation(s)
- Guixiang Ji
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing, 210042, China
| | - Jie Gu
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing, 210042, China
| | - Min Guo
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing, 210042, China
| | - Linjun Zhou
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing, 210042, China
| | - Zhen Wang
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing, 210042, China
| | - Lili Shi
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing, 210042, China
| | - Aihua Gu
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing, China, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, China, 211166, China.
| |
Collapse
|
21
|
Allen TA, Cullen MM, Hawkey N, Mochizuki H, Nguyen L, Schechter E, Borst L, Yoder JA, Freedman JA, Patierno SR, Cheng K, Eward WC, Somarelli JA. A Zebrafish Model of Metastatic Colonization Pinpoints Cellular Mechanisms of Circulating Tumor Cell Extravasation. Front Oncol 2021; 11:641187. [PMID: 34631514 PMCID: PMC8495265 DOI: 10.3389/fonc.2021.641187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 08/31/2021] [Indexed: 01/18/2023] Open
Abstract
Metastasis is a multistep process in which cells must detach, migrate/invade local structures, intravasate, circulate, extravasate, and colonize. A full understanding of the complexity of this process has been limited by the lack of ability to study these steps in isolation with detailed molecular analyses. Leveraging a comparative oncology approach, we injected canine osteosarcoma cells into the circulation of transgenic zebrafish with fluorescent blood vessels in a biologically dynamic metastasis extravasation model. Circulating tumor cell clusters that successfully extravasated the vasculature as multicellular units were isolated under intravital imaging (n = 6). These extravasation-positive tumor cell clusters sublines were then molecularly profiled by RNA-Seq. Using a systems-level analysis, we pinpointed the downregulation of KRAS signaling, immune pathways, and extracellular matrix (ECM) organization as enriched in extravasated cells (p < 0.05). Within the extracellular matrix remodeling pathway, we identified versican (VCAN) as consistently upregulated and central to the ECM gene regulatory network (p < 0.05). Versican expression is prognostic for a poorer metastasis-free and overall survival in patients with osteosarcoma. Together, our results provide a novel experimental framework to study discrete steps in the metastatic process. Using this system, we identify the versican/ECM network dysregulation as a potential contributor to osteosarcoma circulating tumor cell metastasis.
Collapse
Affiliation(s)
- Tyler A Allen
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States
| | - Mark M Cullen
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States
| | - Nathan Hawkey
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States
| | - Hiroyuki Mochizuki
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Lan Nguyen
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States
| | - Elyse Schechter
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States
| | - Luke Borst
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Jeffrey A Yoder
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Jennifer A Freedman
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States.,Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, United States
| | - Steven R Patierno
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States.,Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, United States
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, United States
| | - William C Eward
- Department of Orthopedics, Duke University Medical Center, Durham, NC, United States
| | - Jason A Somarelli
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States.,Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
22
|
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: 9] [Impact Index Per Article: 2.3] [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
|
23
|
Tonon F, Farra R, Zennaro C, Pozzato G, Truong N, Parisi S, Rizzolio F, Grassi M, Scaggiante B, Zanconati F, Bonazza D, Grassi G, Dapas B. Xenograft Zebrafish Models for the Development of Novel Anti-Hepatocellular Carcinoma Molecules. Pharmaceuticals (Basel) 2021; 14:ph14080803. [PMID: 34451900 PMCID: PMC8400454 DOI: 10.3390/ph14080803] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common type of tumor and the second leading cause of tumor-related death worldwide. Liver cirrhosis is the most important predisposing factor for HCC. Available therapeutic approaches are not very effective, especially for advanced HCC, which is the most common form of the disease at diagnosis. New therapeutic strategies are therefore urgently needed. The use of animal models represents a relevant tool for preclinical screening of new molecules/strategies against HCC. However, several issues, including animal husbandry, limit the use of current models (rodent/pig). One animal model that has attracted the attention of the scientific community in the last 15 years is the zebrafish. This freshwater fish has several attractive features, such as short reproductive time, limited space and cost requirements for husbandry, body transparency and the fact that embryos do not show immune response to transplanted cells. To date, two different types of zebrafish models for HCC have been developed: the transgenic zebrafish and the zebrafish xenograft models. Since transgenic zebrafish models for HCC have been described elsewhere, in this review, we focus on the description of zebrafish xenograft models that have been used in the last five years to test new molecules/strategies against HCC.
Collapse
Affiliation(s)
- Federica Tonon
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, 447, I 34149 Trieste, Italy; (F.T.); (R.F.); (C.Z.); (G.P.); (F.Z.); (D.B.)
| | - Rossella Farra
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, 447, I 34149 Trieste, Italy; (F.T.); (R.F.); (C.Z.); (G.P.); (F.Z.); (D.B.)
| | - Cristina Zennaro
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, 447, I 34149 Trieste, Italy; (F.T.); (R.F.); (C.Z.); (G.P.); (F.Z.); (D.B.)
| | - Gabriele Pozzato
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, 447, I 34149 Trieste, Italy; (F.T.); (R.F.); (C.Z.); (G.P.); (F.Z.); (D.B.)
| | - Nhung Truong
- Stem Cell Research and Application Laboratory, VNUHCM, University of Science, Ho Chi Minh City 72711, Vietnam;
| | - Salvatore Parisi
- Pathology Unit, CRO Aviano, National Cancer Institute, IRCCS, I 33081 Aviano, Italy; (S.P.); (F.R.)
- Doctoral School in Molecular Biomedicine, University of Trieste, I 34127 Trieste, Italy
| | - Flavio Rizzolio
- Pathology Unit, CRO Aviano, National Cancer Institute, IRCCS, I 33081 Aviano, Italy; (S.P.); (F.R.)
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, I 30170 Mestre, Italy
| | - Mario Grassi
- Department of Engineering and Architecture, University of Trieste, Via Valerio 6/A, I 34127 Trieste, Italy;
| | - Bruna Scaggiante
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I 34149 Trieste, Italy; (B.S.); (B.D.)
| | - Fabrizio Zanconati
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, 447, I 34149 Trieste, Italy; (F.T.); (R.F.); (C.Z.); (G.P.); (F.Z.); (D.B.)
| | - Deborah Bonazza
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, 447, I 34149 Trieste, Italy; (F.T.); (R.F.); (C.Z.); (G.P.); (F.Z.); (D.B.)
| | - Gabriele Grassi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, 447, I 34149 Trieste, Italy; (F.T.); (R.F.); (C.Z.); (G.P.); (F.Z.); (D.B.)
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I 34149 Trieste, Italy; (B.S.); (B.D.)
- Correspondence:
| | - Barbara Dapas
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I 34149 Trieste, Italy; (B.S.); (B.D.)
| |
Collapse
|
24
|
Quiroz J, Yazdanyar A. Animal models of diabetic retinopathy. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1272. [PMID: 34532409 PMCID: PMC8421981 DOI: 10.21037/atm-20-6737] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 12/23/2020] [Indexed: 12/16/2022]
Abstract
The retina is the posterior neuro-integrated layer of the eye that conducts impulses induced by light to the optic nerve for human vision. Diseases of the retina often leads to diminished vision and in some cases blindness. Diabetes mellitus (DM) is a worldwide public health issue and globally, there is an estimated 463 million people that are affected by DM and its consequences. Diabetic retinopathy (DR) is a blinding complication of chronic uncontrolled DM and is the most common cause of blindness in the United States between the ages 24-75. It is estimated that the global prevalence of DR will increase to 191.0 million by 2030, of those 56.3 million possessing vision-threatening diabetic retinopathy (VTDR). For the most part, current treatment modalities control the complications of DR without addressing the underlying pathophysiology of the disease. Therefore, there is an unmet need for new therapeutics that not only repair the damaged retinal tissue, but also reverse the course of DR. The key element in developing these treatments is expanding our basic knowledge by studying DR pathogenesis in animal models of proliferative and non-proliferative DR (PDR and NPDR). There are numerous models available for the research of both PDR and NPDR with substantial overlap. Animal models available include those with genetic backgrounds prone to hyperglycemic states, immunologic etiologies, or environmentally induced disease. In this review we aimed to comprehensively summarize the available animal models for DR while also providing insight to each model's ocular therapeutic potential for drug discovery.
Collapse
Affiliation(s)
- Jose Quiroz
- Medical Scientist Training Program, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Amirfarbod Yazdanyar
- Department of Ophthalmology and Visual Sciences, State University of New York (SUNY), Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
25
|
Park H, You HH, Song G. Multiple toxicity of propineb in developing zebrafish embryos: Neurotoxicity, vascular toxicity, and notochord defects in normal vertebrate development. Comp Biochem Physiol C Toxicol Pharmacol 2021; 243:108993. [PMID: 33529709 DOI: 10.1016/j.cbpc.2021.108993] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/06/2021] [Accepted: 01/24/2021] [Indexed: 12/15/2022]
Abstract
A dithiocarbamate (DTC) fungicide, propineb, affects thyroid function and exerts immunotoxicity, cytotoxicity, and neurotoxicity in humans. Long-term exposure to propineb is associated with carcinogenicity, teratogenicity, malfunction of the reproductive system, and abnormalities in vital signs during organ development. However, there is no evidence of acute toxicity attributable to propineb in zebrafish. Therefore, in the present study, we assessed the toxicity of propineb in zebrafish by studying its adverse effects on embryo development, angiogenesis, and notochord development. Embryos with propineb exposure developed morphological and physiological defects and in larvae, apoptosis and notochord defects were induced in the early development stage. Transgenic fli1:eGFP zebrafish exposed to propineb showed abnormal larval development with defects in angiogenesis and deformed vasculature. Propineb induced irreversible damage to the neural development of embryos and neurogenic defects in developing zebrafish in transgenic olig2:dsRED zebrafish. These results show that exposure to propineb triggers abnormalities in different organ systems of zebrafish and suggests the physiological complexity of the response to propineb.
Collapse
Affiliation(s)
- Hahyun Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Hyekyoung Hannah You
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
26
|
El-Sahli S, Hua K, Sulaiman A, Chambers J, Li L, Farah E, McGarry S, Liu D, Zheng P, Lee SH, Cui J, Ekker M, Côté M, Alain T, Li X, D'Costa VM, Wang L, Gadde S. A triple-drug nanotherapy to target breast cancer cells, cancer stem cells, and tumor vasculature. Cell Death Dis 2021; 12:8. [PMID: 33414428 PMCID: PMC7791049 DOI: 10.1038/s41419-020-03308-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 10/20/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, accounting for the majority of breast cancer-related death. Due to the lack of specific therapeutic targets, chemotherapeutic agents (e.g., paclitaxel) remain the mainstay of systemic treatment, but enrich a subpopulation of cells with tumor-initiating capacity and stem-like characteristics called cancer stem cells (CSCs); thus development of a new and effective strategy for TNBC treatment is an unmet medical need. Cancer nanomedicine has transformed the landscape of cancer drug development, allowing for a high therapeutic index. In this study, we developed a new therapy by co-encapsulating clinically approved drugs, such as paclitaxel, verteporfin, and combretastatin (CA4) in polymer-lipid hybrid nanoparticles (NPs) made of FDA-approved biomaterials. Verteporfin is a drug used in the treatment of macular degeneration and has recently been found to inhibit the Hippo/YAP (Yes-associated protein) pathway, which is known to promote the progression of breast cancer and the development of CSCs. CA4 is a vascular disrupting agent and has been tested in phase II/III of clinical trials. We found that our new three drug-NP not only effectively inhibited TNBC cell viability and cell migration, but also significantly diminished paclitaxel-induced and/or CA4-induced CSC enrichment in TNBC cells, partially through inhibiting the upregulated Hippo/YAP signaling. Combination of verteporfin and CA4 was also more effective in suppressing angiogenesis in an in vivo zebrafish model than single drug alone. The efficacy and application potential of our triple drug-NPs were further assessed by using clinically relevant patient-derived xenograft (PDX) models. Triple drug-NP effectively inhibited the viability of PDX organotypic slide cultures ex vivo and stopped the growth of PDX tumors in vivo. This study developed an approach capable of simultaneously inhibiting bulk cancer cells, CSCs, and angiogenesis.
Collapse
Affiliation(s)
- Sara El-Sahli
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Khang Hua
- Department of Biology, Faculty of Science, University of Ottawa, 30 Marie Curie Ottawa, Ottawa, ON, K1N 6N5, Canada
| | - Andrew Sulaiman
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Jason Chambers
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Li Li
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Eliya Farah
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Sarah McGarry
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Dan Liu
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Department of Genetics, School of Basic Medicine, Qiqihar Medical University, No.333 Bukui North Street, Jianhua District, 161006, Qiqihar, Heilongjiang, People's Republic of China
| | - Peiyong Zheng
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, 200032, Shanghai, People's Republic of China
| | - Seung-Hwan Lee
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Jiefeng Cui
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Xue Yuan Road, 200032, Shanghai, People's Republic of China
| | - Marc Ekker
- Department of Biology, Faculty of Science, University of Ottawa, 30 Marie Curie Ottawa, Ottawa, ON, K1N 6N5, Canada
| | - Marceline Côté
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Tommy Alain
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Xuguang Li
- Centre for Biologics Evaluation, Biologics and Genetic Therapies Directorate, Health Canada, Sir Frederick G. Banting Research Centre, 251 Sir Frederick G. Banting, Ottawa, ON, K1Y 0M1, Canada
| | - Vanessa M D'Costa
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| | - Suresh Gadde
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
27
|
Liu P, Wang S, Chang Z, Li L, Xing H, Dong WF. Combined toxicity of silica nanoparticles and cadmium chloride on the cardiovascular system of zebrafish (Danio rerio) larvae. Comp Biochem Physiol C Toxicol Pharmacol 2021; 239:108895. [PMID: 32942051 DOI: 10.1016/j.cbpc.2020.108895] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/04/2020] [Accepted: 09/09/2020] [Indexed: 12/22/2022]
Abstract
Silica nanoparticles (SiNPs) and cadmium are major environmental pollutants that have severe detrimental effects on living organisms. Recent studies have demonstrated that the combined exposure of SiNPs and heavy metals is more toxic than individual exposure. However, studies on the effects of the combined exposure of SiNPs and cadmium, on the cardiovascular system are rare. In this study, zebrafish (Danio rerio) embryos were exposed to a combination of SiNPs and experimentally safe concentrations CdCl2 for studying the alterations in heart morphology, heart rate, apoptosis, and vascular endothelial cells. The results demonstrated that the detrimental effects of SiNPs+CdCl2 exposure on the heart rate and vascular endothelial cells were more severe than those following exposure to SiNPs or CdCl2 alone. Gene expression analysis revealed that apoptosis and inflammation could be the major pathways underlying the toxicity caused by the combined exposure of SiNPs and CdCl2.
Collapse
Affiliation(s)
- Pai Liu
- University of Science and Technology of China, Hefei 230026, People's Republic of China; CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), 88 Keling Road, Suzhou 215163, People's Republic of China
| | - Sheng Wang
- University of Science and Technology of China, Hefei 230026, People's Republic of China; CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), 88 Keling Road, Suzhou 215163, People's Republic of China
| | - Zhimin Chang
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), 88 Keling Road, Suzhou 215163, People's Republic of China
| | - Li Li
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), 88 Keling Road, Suzhou 215163, People's Republic of China
| | - Hao Xing
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), 88 Keling Road, Suzhou 215163, People's Republic of China
| | - Wen-Fei Dong
- University of Science and Technology of China, Hefei 230026, People's Republic of China; CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), 88 Keling Road, Suzhou 215163, People's Republic of China.
| |
Collapse
|
28
|
Extra-Intestinal Effects of C. difficile Toxin A and B: An In Vivo Study Using the Zebrafish Embryo Model. Cells 2020; 9:cells9122575. [PMID: 33271969 PMCID: PMC7760802 DOI: 10.3390/cells9122575] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/27/2020] [Accepted: 11/29/2020] [Indexed: 12/18/2022] Open
Abstract
C.difficile infection (CDI) is not a merely “gut-confined” disease as toxemia could drive the development of CDI-related extra-intestinal effects. These effects could explain the high CDI-associated mortality, not just justified by diarrhea and dehydration. Here, the extra-intestinal effects of toxin A (TcdA) and B (TcdB) produced by C. difficile have been studied in vivo using the zebrafish embryo model. Noteworthy, protective properties of human serum albumin (HSA) towards toxins-induced extra-intestinal effects were also addressed. Zebrafish embryos were treated with TcdA, TcdB and/or HSA at 24 h post-fertilization. Embryos were analyzed for 48 h after treatment to check vital signs and morphological changes. Markers related to cardio-vascular damage and inflammation were evaluated by Real-Time quantitative PCR and/or western blotting. Both toxins induced cardiovascular damage in zebrafish embryos by different mechanisms: (i) direct toxicity (i.e., pericardial edema, cardiac chambers enlargement, endothelial alteration); (ii) increased hormonal production and release (i.e., atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP)), (iii) alteration of the vascular system through the increase of the vascular endothelial growth factor (VEGF-A) levels, as well as of its receptors, (iv) pro-inflammatory response through high cytokines production (i.e., CXCL8, IL1B, IL6 and TNFα) and (v) cell-mediated damage due to the increase in neutrophils number. In addition to cardiovascular damage, we observe skin alteration and inflammation. Finally, our data indicate a protective effect of HSA toward the toxins induced extra-intestinal effects. Together, our findings can serve as a starting point for humans’ studies to substantiate and understand the extra-intestinal effects observed in CDI patients.
Collapse
|
29
|
Lee JY, Yang C, Lim W, Song G. Methiothepin Suppresses Human Ovarian Cancer Cell Growth by Repressing Mitochondrion-Mediated Metabolism and Inhibiting Angiogenesis In Vivo. Pharmaceutics 2020; 12:pharmaceutics12070686. [PMID: 32698407 PMCID: PMC7407284 DOI: 10.3390/pharmaceutics12070686] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/14/2020] [Accepted: 07/19/2020] [Indexed: 01/04/2023] Open
Abstract
Ovarian cancer is the fifth leading cause of cancer-related deaths in women. Despite treatment, most patients experience relapse and the 5-year survival rate of ovarian cancer is less than 50%. Serotonin has cell growth-promoting functions in a variety of carcinomas, but the effect of serotonin receptor antagonists on ovarian cancer cells is unknown. In this study, it was confirmed that methiothepin, a serotonin receptor antagonist, suppresses the viability of, and induces apoptosis in, ovarian cancer cells. Methiothepin also induces mitochondrial dysfunction, represented by depolarization of the mitochondrial membrane and increased mitochondrion-specific Ca2+ levels, and causes metabolic disruption in cancer cells such as decreased ATP production and oxidative phosphorylation. Methiothepin also interferes with vascular development in transgenic zebrafish embryos. Combination treatment with methiothepin improves the anti-cancer effect of paclitaxel, a standard chemotherapeutic agent. In conclusion, this study revealed that methiothepin is a potential novel therapeutic agent for ovarian cancer treatment.
Collapse
Affiliation(s)
- Jin-Young Lee
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Changwon Yang
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, Korea University, Seoul 02841, Korea;
| | - Whasun Lim
- Department of Food and Nutrition, Kookmin University, Seoul 02707, Korea
- Correspondence: (W.L.); (G.S.); Tel.: +82-2-910-4773 (W.L.); +82-2-3290-3012 (G.S.)
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, Korea University, Seoul 02841, Korea;
- Correspondence: (W.L.); (G.S.); Tel.: +82-2-910-4773 (W.L.); +82-2-3290-3012 (G.S.)
| |
Collapse
|
30
|
Wang W, Ru S, Wang L, Qin J, Ru Y, Zhang J, Zhang X. Bisphenol S Induces Ectopic Angiogenesis in Embryos via VEGFR2 Signaling, Leading to Lipid Deposition in Blood Vessels of Larval Zebrafish. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:6822-6831. [PMID: 32348130 DOI: 10.1021/acs.est.9b07080] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Bisphenol S (BPS), used as a bisphenol A substitute, has been detected in various environments. However, the safety of BPS is still unclear. Here, zebrafish embryos were exposed to BPS (0, 1, 10, and 100 μg/L) for 24, 48, 72, 96 h, and 15 days. BPS induced ectopic sprouting of budding blood vessels in embryos, but the blood flow velocity within these lesions was unchanged at 48 h. At 72 h postfertilization (hpf), by observing the subintestinal venous plexus responsible for yolk absorption, we found that VEGFR2 transduced an angiogenic signal and that the subsequent reduction in blood flow velocity inhibited yolk absorption. At 96 hpf, yolk consumption was still delayed because of the disturbed transportation route, resulting in transient extensive lipid retention in the blood vessels. After feeding, obvious atherogenic lipids were discovered in the blood vessels, especially in bends, bifurcations, and stenoses. This dynamic visualization of the pathogenesis demonstrates a plausible mechanistic link between BPS exposure-induced embryonic vessel overgrowth and an increased atherosclerosis risk.
Collapse
Affiliation(s)
- Weiwei Wang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Shaoguo Ru
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Liangliang Wang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Jingyu Qin
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Yiran Ru
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093, United States
| | - Jie Zhang
- 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
|
31
|
Giordo R, Nasrallah GK, Al-Jamal O, Paliogiannis P, Pintus G. Resveratrol Inhibits Oxidative Stress and Prevents Mitochondrial Damage Induced by Zinc Oxide Nanoparticles in Zebrafish ( Danio rerio). Int J Mol Sci 2020; 21:E3838. [PMID: 32481628 PMCID: PMC7312482 DOI: 10.3390/ijms21113838] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 12/22/2022] Open
Abstract
Despite their wide industrial use, Zinc oxide (ZnO) nanoparticles (NPs) exhibit a high toxic potential while concerns of their health-related risks are still present, urging additional in vivo clarification studies. Oxidative stress is recognized as the primary trigger of NP-associated toxicity, suggesting antioxidants as a promising counteractive approach. Here, we investigated the protective effect of the natural antioxidant resveratrol against ZnO NP-induced toxicity in vivo using the zebrafish model. Our findings demonstrate that resveratrol counteracts ZnO NP-induced zebrafish lethality preventing cardiac morphological and functional damage. NP-induced vascular structural abnormalities during embryonic fish development were significantly counteracted by resveratrol treatment. Mechanistically, we further showed that resveratrol inhibits ROS increase, prevents mitochondrial membrane potential dysfunction, and counteracts cell apoptosis/necrosis elicited by ZnO NP. Overall, our data provide further evidence demonstrating the primary role of oxidative stress in NP-induced damage, and highlight new insights concerning the protective mechanism of antioxidants against nanomaterial toxicity.
Collapse
Affiliation(s)
- Roberta Giordo
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar; (R.G.); (O.A.-J.)
| | - Gheyath K. Nasrallah
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar; (R.G.); (O.A.-J.)
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Ola Al-Jamal
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar; (R.G.); (O.A.-J.)
| | - Panagiotis Paliogiannis
- Department of Medical, Surgical and Experimental Surgery, University of Sassari, Viale San Pietro 43, 07100 Sassari, Italy;
| | - Gianfranco Pintus
- Department of Medical Laboratory Sciences, College of Health Sciences and Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, UAE
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100 Sassari, Italy
| |
Collapse
|
32
|
Zheng L, Abdelgawwad MS, Zhang D, Xu L, Wei S, Cao W, Zheng XL. Histone-induced thrombotic thrombocytopenic purpura in adamts13 -/- zebrafish depends on von Willebrand factor. Haematologica 2020; 105:1107-1119. [PMID: 31753928 PMCID: PMC7109750 DOI: 10.3324/haematol.2019.237396] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 11/21/2019] [Indexed: 12/30/2022] Open
Abstract
Thrombotic thrombocytopenic purpura (TTP) is caused by severe deficiency of ADAMTS13 (A13), a plasma metalloprotease that cleaves endothelium-derived von Willebrand factor (VWF). However, severe A13 deficiency alone is often not sufficient to cause an acute TTP; additional factors may be required to trigger the disease. Using CRISPR/Cas9, we created and characterized several novel zebrafish lines carrying a null mutation in a13-/- , vwf, and both. We further used these zebrafish lines to test the hypothesis that inflammation that results in neutrophil activation and release of histone/DNA complexes may trigger TTP. As shown, a13-/- zebrafish exhibit increased levels of plasma VWF antigen, multimer size, and ability of thrombocytes to adhere to a fibrillar collagen-coated surface under flow. The a13-/- zebrafish also show an increased rate of occlusive thrombus formation in the caudal venules after FeCl3 injury. More interestingly, a13-/- zebrafish exhibit ~30% reduction in the number of total, immature, and mature thrombocytes with increased fragmentation of erythrocytes. Administration of a lysine-rich histone results in more severe and persistent thrombocytopenia and a significantly increased mortality rate in a13-/- zebrafish than in wildtype (wt) ones. However, both spontaneous and histone-induced TTP in a13-/- zebrafish are rescued by the deletion of vwf These results demonstrate a potentially mechanistic link between inflammation and the onset of TTP in light of severe A13 deficiency; the novel zebrafish models of TTP may help accelerate our understanding of pathogenic mechanisms and the discoveries of novel therapeutics for TTP and perhaps other arterial thrombotic disorders.
Collapse
Affiliation(s)
| | | | - Di Zhang
- Divisions of Laboratory Medicine
| | | | - Shi Wei
- Divisions of Anatomic Pathology, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | |
Collapse
|
33
|
Moon WK, Atique U, An KG. "Ecological risk assessments and eco-toxicity analyses using chemical, biological, physiological responses, DNA damages and gene-level biomarkers in Zebrafish (Danio rerio) in an urban stream". CHEMOSPHERE 2020; 239:124754. [PMID: 31726531 DOI: 10.1016/j.chemosphere.2019.124754] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/02/2019] [Accepted: 09/03/2019] [Indexed: 06/10/2023]
Abstract
We conducted the ecological risk assessment in an urban stream by using multiple-level approaches ranging from community level, chemical analyses in water and sediments, physiological assays of DNA biomarkers, embryonic development tests, and gene-level marker analyses of cyp1a, c-Fos, CRH, transgenic fli1:GFP and HuC:eGFP in zebrafish (Danio rerio). In water, the chemical perturbations based on nutrients (N,P), organic matter, ionic contents and metals identified in downstream zone. Analogous corroborations verified in sediment samples having hazardous metals (Zn, Pb, Cu, Ni, As, Cd). The chemical contaminations reflected significant damages in fish DNA, based on tDNA, tail length (TL), and tail extent moment (TEM). Zebrafish embryonic development experiments significantly enlightened the chemical contaminants in downstream compared to those in control and reference conditions. Hatching and survival rates rigorously declined in downstream region. Embryonic development delayed and followed by death in the downstream substantiated by the above-mentioned findings. Similar were the findings on heart rate and pigmentation largely affected in the contaminated zone. Pollutants in urban stream reflected significantly at the gene level, and were corroborated through experiments using transgenic zebrafish strains that were influenced by pollutants during the process of occurrence. In conclusion, these studies illuminate the community to gene-level ecological health assessment that could be useful for ecological risk assessments of urban streams and rivers. Further, the gene-level biomarkers and transgenic zebrafish experiments combination propose the procedures could be effectively used as sensitive and efficient biomarkers of ecological health and risk assessment in urban streams from community to gene-level assessments.
Collapse
Affiliation(s)
- Won-Ki Moon
- Department of Bioscience and Biotechnology, Chungnam National University, Daejeon, 34134, Republic of Korea.
| | - Usman Atique
- Department of Bioscience and Biotechnology, Chungnam National University, Daejeon, 34134, Republic of Korea.
| | - Kwang-Guk An
- Department of Bioscience and Biotechnology, Chungnam National University, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
34
|
Zanin R, Pegoraro S, Ros G, Ciani Y, Piazza S, Bossi F, Bulla R, Zennaro C, Tonon F, Lazarevic D, Stupka E, Sgarra R, Manfioletti G. HMGA1 promotes breast cancer angiogenesis supporting the stability, nuclear localization and transcriptional activity of FOXM1. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:313. [PMID: 31311575 PMCID: PMC6636010 DOI: 10.1186/s13046-019-1307-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023]
Abstract
Background Breast cancer is the most common malignancy in women worldwide. Among the breast cancer subtypes, triple-negative breast cancer (TNBC) is the most aggressive and the most difficult to treat. One of the master regulators in TNBC progression is the architectural transcription factor HMGA1. This study aimed to further explore the HMGA1 molecular network to identify molecular mechanisms involved in TNBC progression. Methods RNA from the MDA-MB-231 cell line, silenced for HMGA1 expression, was sequenced and, with a bioinformatic analysis, molecular partners HMGA1 could cooperate with in regulating common downstream gene networks were identified. Among the putative partners, the FOXM1 transcription factor was selected. The relationship occurring between HMGA1 and FOXM1 was explored by qRT-PCR, co-immunoprecipitation and protein stability assays. Subsequently, the transcriptional activity of HMGA1 and FOXM1 was analysed by luciferase assay on the VEGFA promoter. The impact on angiogenesis was assessed in vitro, evaluating the tube formation ability of endothelial cells exposed to the conditioned medium of MDA-MB-231 cells silenced for HMGA1 and FOXM1 and in vivo injecting MDA-MB-231 cells, silenced for the two factors, in zebrafish larvae. Results Here, we discover FOXM1 as a novel molecular partner of HMGA1 in regulating a gene network implicated in several breast cancer hallmarks. HMGA1 forms a complex with FOXM1 and stabilizes it in the nucleus, increasing its transcriptional activity on common target genes, among them, VEGFA, the main inducer of angiogenesis. Furthermore, we demonstrate that HMGA1 and FOXM1 synergistically drive breast cancer cells to promote tumor angiogenesis both in vitro in endothelial cells and in vivo in a zebrafish xenograft model. Moreover, using a dataset of breast cancer patients we show that the co-expression of HMGA1, FOXM1 and VEGFA is a negative prognostic factor of distant metastasis-free survival and relapse-free survival. Conclusions This study reveals FOXM1 as a crucial interactor of HMGA1 and proves that their cooperative action supports breast cancer aggressiveness, by promoting tumor angiogenesis. Therefore, the possibility to target HMGA1/FOXM1 in combination should represent an attractive therapeutic option to counteract breast cancer angiogenesis. Electronic supplementary material The online version of this article (10.1186/s13046-019-1307-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rossella Zanin
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Silvia Pegoraro
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy.
| | - Gloria Ros
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Yari Ciani
- Laboratorio Nazionale CIB, Area Science Park, Padriciano 99, Trieste, Italy.,Present address: Department of Cellular, Computational and Integrative Biology - (CIBIO), University of Trento, Via Sommarive 9, 38123, Trento, Italy
| | - Silvano Piazza
- Department of Cellular, Computational and Integrative Biology - (CIBIO), University of Trento, Via Sommarive 9, 38123, Povo, Trento, Italy
| | - Fleur Bossi
- Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) "Burlo Garofolo", via dell'Istria 65/1, 34134, Trieste, Italy
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Cristina Zennaro
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34149, Trieste, Italy
| | - Federica Tonon
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34149, Trieste, Italy
| | - Dejan Lazarevic
- Center for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elia Stupka
- Center for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Present address: Life Sciences Business Health Catalyst, Cambridge, Via Sommarive 9, 38123, USA
| | - Riccardo Sgarra
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | | |
Collapse
|
35
|
Ma Y, Zang L, Wang D, Jiang J, Wang C, Wang X, Fang F, Wang H. Effects of miR-181a-5p abnormal expression on zebrafish (Danio rerio) vascular development following triclosan exposure. CHEMOSPHERE 2019; 223:523-535. [PMID: 30784759 DOI: 10.1016/j.chemosphere.2019.02.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/07/2019] [Accepted: 02/09/2019] [Indexed: 06/09/2023]
Abstract
Triclosan (TCS), one of the important bactericides, is widely used in personal care products, and its chronic exposure leads to severe toxic effects on the growth and development of blood vessels in zebrafish (Danio rerio). Herein, we screened out three differentially expressed miRNAs (miR-181a-5p, miR-132-3p and miR-128-3p) by sequencing and qRT-PCR analyses of 4-96-hpf TCS-exposed zebrafish, among which miR-181a-5p was found to regulate many signaling pathways involved in fatty acid biosynthesis and phosphatidylimositol signaling systems. By O-dianisidine staining, TCS-exposure resulted in decreased distribution of red blood cells and induced blood hypercoagulable state and thrombotic effects. Defective subintestinal veins (SIVs), and decreased branching and curvature of blood vessels were observed with increasing TCS-exposure concentrations. After microinjection of miR-181a-5p mimic and inhibitor, zebrafish malformation type and percentage were prominently increased such as distorted SIV vessels along with reduced venation and abnormal branches by ALP staining. Overexpressed miR-181a-5p had a greater effect on development and branching patterns of arteries and veins than its knockdown. By laser confocal microscopy observation, the 72-hpf Tg (flk1: mCherry) zebrafish obviously displayed vascular proliferation and ablation in the miR-181a-5p mimic group. Microinjection of miR-181a-5p mimics and inhibitors led to abnormal expressions (20-50%) of two key target genes (pax2a and vash2) by WISH, and increased malformation percentages (18-45%) by IOD analysis. Overexpression of vash2 led to the inhibitory or promoting effects on the expression of PI3K signaling pathway-related genes, proving that the effect of vash2 on development of blood vessels could be realized by inhibiting PI3K signaling pathway. These observations lay theoretical foundation for deep insight into the molecular mechanisms on TCS-induced cardiovascular diseases.
Collapse
Affiliation(s)
- Yan Ma
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Luxiu Zang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Danting Wang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jiahui Jiang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Caihong Wang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xuedong Wang
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China.
| | - Fang Fang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Huili Wang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
36
|
Toxicological Evaluation of SiO₂ Nanoparticles by Zebrafish Embryo Toxicity Test. Int J Mol Sci 2019; 20:ijms20040882. [PMID: 30781642 PMCID: PMC6413002 DOI: 10.3390/ijms20040882] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 02/03/2019] [Accepted: 02/10/2019] [Indexed: 11/21/2022] Open
Abstract
As the use of nanoparticles (NPs) is increasing, the potential toxicity and behavior of NPs in living systems need to be better understood. Our goal was to evaluate the developmental toxicity and bio-distribution of two different sizes of fluorescently-labeled SiO2 NPs, 25 and 115 nm, with neutral surface charge or with different surface functionalization, rendering them positively or negatively charged, in order to predict the effect of NPs in humans. We performed a zebrafish embryo toxicity test (ZFET) by exposing the embryos to SiO2 NPs starting from six hours post fertilization (hpf). Survival rate, hatching time, and gross morphological changes were assessed at 12, 24, 36, 48, 60, and 72 hpf. We evaluated the effect of NPs on angiogenesis by counting the number of sub-intestinal vessels between the second and seventh intersegmental vessels and gene expression analysis of vascular endothelial growth factor (VEGF) and VEGF receptors at 72 hpf. SiO2 NPs did not show any adverse effects on survival rate, hatching time, gross morphology, or physiological angiogenesis. We found that SiO2 NPs were trapped by the chorion up until to the hatching stage. After chemical removal of the chorion (dechorionation), positively surface-charged SiO2 NPs (25 nm) significantly reduced the survival rate of the fish compared to the control group. These results indicate that zebrafish chorion acts as a physical barrier against SiO2 NPs, and removing the chorions in ZFET might be necessary for evaluation of toxicity of NPs.
Collapse
|
37
|
Kteeba SM, El-Ghobashy AE, El-Adawi HI, El-Rayis OA, Sreevidya VS, Guo L, Svoboda KR. Exposure to ZnO nanoparticles alters neuronal and vascular development in zebrafish: Acute and transgenerational effects mitigated with dissolved organic matter. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 242:433-448. [PMID: 30005256 DOI: 10.1016/j.envpol.2018.06.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 05/21/2018] [Accepted: 06/09/2018] [Indexed: 06/08/2023]
Abstract
Exposure to ZnO-nanoparticles (NPs) in embryonic zebrafish reduces hatching rates which can be mitigated with dissolved organic material (DOM). Although hatching rate can be a reliable indicator of toxicity and DOM mitigation potential, a fish that has been exposed to ZnO-NPs or any other toxicant may also exhibit other abnormal phenotypes not readily detected by the unaided eye. In this study, we moved beyond hatching rate analysis to investigate the consequences of ZnO-NPs exposure on the nervous and vascular systems in developing zebrafish. Zebrafish exposed to ZnO-NPs (1-100 ppm) exhibited an array of cellular phenotypes including: abnormal secondary motoneuron (SMN) axonal projections, abnormal dorsal root ganglion development and abnormal blood vessel development. Dissolved Zn (<10 kDa) exposure also caused abnormal SMN axonal projections, but to a lesser extent than ZnO-NPs. The ZnO-NPs-induced abnormal phenotypes were reversed in embryos concurrently exposed with various types of DOM. In these acute mitigation exposure experiments, humic acid and carbohydrate, along with natural organic matter obtained from the Suwannee River in Georgia and Milwaukee River in Wisconsin, were the best mitigators of ZnO-NPs-induced motoneuron toxicity at 96 h post fertilization. Further experiments were performed to determine if the ZnO-NPs-induced, abnormal axonal phenotypes and the DOM mitigated axonal phenotypes could persist across generations. Abnormal SMN axon phenotypes caused by ZnO-NPs-exposure were detected in F1 and F2 generations. These are fish that have not been directly exposed to ZnO-NPs. Fish mitigated with DOM during the acute exposure (F0 generation) had a reduction in abnormal motoneuron axon errors in larvae of subsequent generations. Therefore, ZnO-NPs exposure results in neurotoxicity in developing zebrafish which can persist from one generation to the next. Mitigation with DOM can reverse the abnormal phenotypes in an acute embryonic exposure context, as well as across generations, resulting in healthy fish.
Collapse
Affiliation(s)
- Shimaa M Kteeba
- School of Freshwater Sciences, University of Wisconsin-Milwaukee, 600 E. Greenfield Avenue, Milwaukee, WI, 53204, USA; Zoology Department, Faculty of Science, Damietta University, New Damietta 34511, Damietta, Egypt
| | - Ahmed E El-Ghobashy
- Zoology Department, Faculty of Science, Damietta University, New Damietta 34511, Damietta, Egypt
| | - Hala I El-Adawi
- Medical Biotechnology Department, Genetic Engineering and Biotech Institute, Borg El-Arab City for Scientific Research, Universities and Research District Sector, New Borg El-Arab 21934, Alexandria, Egypt
| | - Osman A El-Rayis
- Oceanography Department, Faculty of Science, Alexandria University, Moharrem Bek 21511, Alexandria, Egypt
| | - Virinchipuram S Sreevidya
- Joseph J. Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI, 53205, USA
| | - Laodong Guo
- School of Freshwater Sciences, University of Wisconsin-Milwaukee, 600 E. Greenfield Avenue, Milwaukee, WI, 53204, USA
| | - Kurt R Svoboda
- Joseph J. Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI, 53205, USA.
| |
Collapse
|
38
|
A Novel In Vivo Model to Study Impaired Tissue Regeneration Mediated by Cigarette Smoke. Sci Rep 2018; 8:10926. [PMID: 30026555 PMCID: PMC6053433 DOI: 10.1038/s41598-018-28687-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/13/2018] [Indexed: 12/27/2022] Open
Abstract
Cigarette smoke is associated with several pathologies including chronic respiratory diseases and cancer. In addition, exposure to cigarette smoke is correlated with impaired wound healing, where a significant decrease in the regenerative capacity of smokers is well documented and broadly considered a negative risk factor after trauma or surgery. So far, some in vitro and in vivo models have been described to study how exposure to cigarette smoke diminishes the regenerative potential in different organisms. However, although useful, many of these models are difficult and expensive to implement and do not allow high-throughput screening approaches. In order to establish a reliable and accessible model, we have evaluated the effects of cigarette smoke extract (CSE) on zebrafish development and regeneration. In this work, zebrafish embryos and larvae were exposed to low doses of aqueous CSE showing severe developmental abnormalities in a dose-dependent manner. Furthermore, when adult zebrafish were subjected to caudal fin amputation, we observed a significant decrease in the regenerative capacity of animals exposed to CSE. The effect was exacerbated in male and aged fish compared to female or young organisms. The establishment of a zebrafish model to assess the consequences of cigarette smoke and its effects on animal physiology could provide a new tool to study the underlying mechanisms involved in impaired tissue regeneration, and aid the development of novel approaches to treat complications associated with cigarette smoke toxicity.
Collapse
|
39
|
Poon KL, Wang X, Lee SGP, Ng AS, Goh WH, Zhao Z, Al-Haddawi M, Wang H, Mathavan S, Ingham PW, McGinnis C, Carney TJ. Editor's Highlight: Transgenic Zebrafish Reporter Lines as Alternative In Vivo Organ Toxicity Models. Toxicol Sci 2018; 156:133-148. [PMID: 28069987 DOI: 10.1093/toxsci/kfw250] [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] [Indexed: 12/15/2022] Open
Abstract
Organ toxicity, particularly liver toxicity, remains one of the major reasons for the termination of drug candidates in the development pipeline as well as withdrawal or restrictions of marketed drugs. A screening-amenable alternative in vivo model such as zebrafish would, therefore, find immediate application in the early prediction of unacceptable organ toxicity. To identify highly upregulated genes as biomarkers of toxic responses in the zebrafish model, a set of well-characterized reference drugs that cause drug-induced liver injury (DILI) in the clinic were applied to zebrafish larvae and adults. Transcriptome microarray analysis was performed on whole larvae or dissected adult livers. Integration of data sets from different drug treatments at different stages identified common upregulated detoxification pathways. Within these were candidate biomarkers which recurred in multiple treatments. We prioritized 4 highly upregulated genes encoding enzymes acting in distinct phases of the drug metabolism pathway. Through promoter isolation and fosmid recombineering, eGFP reporter transgenic zebrafish lines were generated and evaluated for their response to DILI drugs. Three of the 4 generated reporter lines showed a dose and time-dependent induction in endodermal organs to reference drugs and an expanded drug set. In conclusion, through integrated transcriptomics and transgenic approaches, we have developed parallel independent zebrafish in vivo screening platforms able to predict organ toxicities of preclinical drugs.
Collapse
Affiliation(s)
- Kar Lai Poon
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, 138673 Singapore
| | - Xingang Wang
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, 138673 Singapore
| | - Serene G P Lee
- Genome Institute of Singapore, A*STAR (Agency for Science, Technology and Research), 60 Biopolis Street, 138672 Singapore
| | - Ashley S Ng
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, 138673 Singapore
| | - Wei Huang Goh
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, 138673 Singapore
| | - Zhonghua Zhao
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, 138673 Singapore
| | - Muthafar Al-Haddawi
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, 138673 Singapore
| | - Haishan Wang
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, 138673 Singapore
| | - Sinnakaruppan Mathavan
- Genome Institute of Singapore, A*STAR (Agency for Science, Technology and Research), 60 Biopolis Street, 138672 Singapore
| | - Philip W Ingham
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, 138673 Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| | - Claudia McGinnis
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, 138673 Singapore.,Roche Pharmaceutical Research & Early Development (pRED), Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Tom J Carney
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, 138673 Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| |
Collapse
|
40
|
Wang TW, Chang KC, Chen LH, Liao SY, Yeh CW, Chuang YJ. Effects of an injectable functionalized self-assembling nanopeptide hydrogel on angiogenesis and neurogenesis for regeneration of the central nervous system. NANOSCALE 2017; 9:16281-16292. [PMID: 29046917 DOI: 10.1039/c7nr06528k] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Brain injury is a devastating medical condition and represents a major health problem. Tissue and organ reconstruction have been regarded as promising therapeutic strategies. Here, we propose a regenerative methodology focusing on the provision of functionalized nanopeptide scaffolds to facilitate angiogenesis and neurogenesis at the brain injury site. The peptide RADA16-SVVYGLR undergoes self-assembly to construct an interconnected network with intertwining nanofibers, and can be controlled to display various physicochemical properties by the adjustment of microenvironmental factors such as pH and ion concentration. Such scaffolds can support endothelial cells to form tube-like structures and neural stem cells to survive and proliferate. In an in vivo zebrafish brain injury model, sprouting angiogenesis and developmental neurogenesis were achieved, and functional recovery of the severed optic tectum was enhanced in RADA16-SVVYGLR hydrogel-implanted zebrafish. This nanopeptide hydrogel was non-toxic to zebrafish embryos during early developmental stages. This angiogenic self-assembling peptide hydrogel had programmable physical properties, good biocompatibility, and regenerative ability for functional recovery in the injured brain. We suggest that functionalized self-assembling peptides encapsulated with neural stem cells or used alone could be an attractive and effective therapeutic modality for brain injury and diseases (e.g., trauma, stroke, tumor, degenerative neurological disorders, etc.).
Collapse
Affiliation(s)
- Tzu-Wei Wang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan.
| | | | | | | | | | | |
Collapse
|
41
|
Ganta VC, Annex BH. LMO2 (LIM Domain Only 2) and Endothelial Cell Migration in Developmental and Postnatal Angiogenesis. Arterioscler Thromb Vasc Biol 2017; 37:1806-1808. [PMID: 28954807 DOI: 10.1161/atvbaha.117.309953] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Vijay Chaitanya Ganta
- From the Robert M. Berne Cardiovascular Research Center (V.C.G., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Brian H Annex
- From the Robert M. Berne Cardiovascular Research Center (V.C.G., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (B.H.A.), University of Virginia School of Medicine, Charlottesville.
| |
Collapse
|
42
|
Tonon F, Zennaro C, Dapas B, Carraro M, Mariotti M, Grassi G. Rapid and cost-effective xenograft hepatocellular carcinoma model in Zebrafish for drug testing. Int J Pharm 2016; 515:583-591. [PMID: 27989824 DOI: 10.1016/j.ijpharm.2016.10.070] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 10/28/2016] [Indexed: 02/07/2023]
Abstract
We developed a novel, rapid and cost-effective Zebrafish xenograft model of hepatocellular carcinoma (HCC) for drug screening in the disease. Following injection into the yolk sack of Zebrafish larvae of the human HCC cell line JHH6 stained by a vital dye, tumor mass growth was followed by fluorescence microscopy and by human Ki67 quantification. Tumor induced neo-angiogenesis was evaluated by alkaline phosphatase staining of the vessels, by using the Tg(fli1:EGFP)y1 strain of Zebrafish and by the quantification of the zebrafish vascular endothelial growth factor and of its receptor. We show that it is feasible to micro-inject JHH6 in Zebrafish larvae, that injected cells can grow for different days and that this induces a marked neo-angiogenesis. Finally, we show that our model allows testing the effects of anti-HCC drugs such as Bortezomib. Compared to more complex HCC mouse models, our model is far less expensive, faster to set up and does not need immunosuppressant treatment. Finally, the model makes use of JHH6, an aggressive form of HCC cell line never tested before in Zebrafish. In conclusion, the possibility to test anti HCC/neo-angiogenesis drugs makes our JHH6 model useful to select therapeutic molecules for a highly vascularized tumor such as HCC.
Collapse
Affiliation(s)
- Federica Tonon
- Department of Medical, Surgery and Health Sciences, University of Trieste, Cattinara Hospital, Italy
| | - Cristina Zennaro
- Department of Medical, Surgery and Health Sciences, University of Trieste, Cattinara Hospital, Italy
| | - Barbara Dapas
- Department of Life Sciences, University of Trieste, Italy
| | - Michele Carraro
- Department of Medical, Surgery and Health Sciences, University of Trieste, Cattinara Hospital, Italy
| | - Massimo Mariotti
- IRCCS Galeazzi Orthopedic Institute Milan, Italy; Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | | |
Collapse
|
43
|
Gao J, Mahapatra CT, Mapes CD, Khlebnikova M, Wei A, Sepúlveda MS. Vascular toxicity of silver nanoparticles to developing zebrafish (Danio rerio). Nanotoxicology 2016; 10:1363-72. [PMID: 27499207 DOI: 10.1080/17435390.2016.1214763] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Nanoparticles (NPs, 1-100 nm) can enter the environment and result in exposure to humans and other organisms leading to potential adverse health effects. The aim of the present study is to evaluate the effects of early life exposure to polyvinylpyrrolidone-coated silver nanoparticles (PVP-AgNPs, 50 nm), particularly with respect to vascular toxicity on zebrafish embryos and larvae (Danio rerio). Previously published data has suggested that PVP-AgNP exposure can inhibit the expression of genes within the vascular endothelial growth factor (VEGF) signaling pathway, leading to delayed and abnormal vascular development. Here, we show that early acute exposure (0-12 h post-fertilization, hpf) of embryos to PVP-AgNPs at 1 mg/L or higher results in a transient, dose-dependent induction in VEGF-related gene expression that returns to baseline levels at hatching (72 hpf). Hatching results in normoxia, negating the effects of AgNPs on vascular development. Interestingly, increased gene transcription was not followed by the production of associated proteins within the VEGF pathway, which we attribute to NP-induced stress in the endoplasmic reticulum (ER). The impaired translation may be responsible for the observed delays in vascular development at later stages, and for smaller larvae size at hatching. Silver ion (Ag(+)) concentrations were < 0.001 mg/L at all times, with no significant effects on the VEGF pathway. We propose that PVP-AgNPs temporarily delay embryonic vascular development by interfering with oxygen diffusion into the egg, leading to hypoxic conditions and ER stress.
Collapse
Affiliation(s)
- Jiejun Gao
- a Department of Forestry and Natural Resources and Bindley Biosciences Center
| | - Cecon T Mahapatra
- a Department of Forestry and Natural Resources and Bindley Biosciences Center
| | | | - Maria Khlebnikova
- c Department of Chemistry , Purdue University , West Lafayette, IN , USA
| | - Alexander Wei
- c Department of Chemistry , Purdue University , West Lafayette, IN , USA
| | - Marisol S Sepúlveda
- a Department of Forestry and Natural Resources and Bindley Biosciences Center
| |
Collapse
|
44
|
Bodewein L, Schmelter F, Di Fiore S, Hollert H, Fischer R, Fenske M. Differences in toxicity of anionic and cationic PAMAM and PPI dendrimers in zebrafish embryos and cancer cell lines. Toxicol Appl Pharmacol 2016; 305:83-92. [DOI: 10.1016/j.taap.2016.06.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 06/06/2016] [Accepted: 06/07/2016] [Indexed: 12/12/2022]
|
45
|
Kugler J, Luch A, Oelgeschläger M. Transgenic Mouse Models Transferred into the Test Tube: New Perspectives for Developmental Toxicity Testing In Vitro? Trends Pharmacol Sci 2016; 37:822-830. [PMID: 27450043 DOI: 10.1016/j.tips.2016.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 06/24/2016] [Accepted: 06/30/2016] [Indexed: 11/17/2022]
Abstract
Despite our increasing understanding of molecular mechanisms controlling embryogenesis, the identification and characterization of teratogenic substances still heavily relies on animal testing. Embryonic development depends on cell-autonomous and non-autonomous processes including spatiotemporally regulated extracellular signaling activities. These have been elucidated in transgenic mouse models harboring easily detectable reporter genes under the control of evolutionarily conserved signaling cascades. We propose combining these transgenic mouse models and cells derived thereof with existing alternative toxicological testing strategies. This would enable the plausibility of in vitro data to be verified in light of in vivo data and, ultimately, facilitate regulatory acceptance of in vitro test methods.
Collapse
Affiliation(s)
- Josephine Kugler
- German Federal Institute for Risk Assessment (BfR), Department of Chemical and Product Safety, Max-Dohrn-Strasse 8-10, 10589 Berlin, Germany
| | - Andreas Luch
- German Federal Institute for Risk Assessment (BfR), Department of Chemical and Product Safety, Max-Dohrn-Strasse 8-10, 10589 Berlin, Germany.
| | - Michael Oelgeschläger
- German Federal Institute for Risk Assessment (BfR), Department of Experimental Toxicology and ZEBET, Bf3R, Max-Dohrn-Strasse 8-10, 10589 Berlin, Germany
| |
Collapse
|