1
|
Heusinkveld HJ, Zwart EP, de Haan A, Braeuning A, Alarcan J, van der Ven LTM. The zebrafish embryo as a model for chemically-induced steatosis: A case study with three pesticides. Toxicology 2024; 508:153927. [PMID: 39151607 DOI: 10.1016/j.tox.2024.153927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
There is an increasing incidence and prevalence of fatty liver disease in the western world, with steatosis as the most prevalent variant. Known causes of steatosis include exposure to food-borne chemicals, and overconsumption of alcohol, carbohydrates and fat, and it is a well-known side effect of certain pharmaceuticals such as tetracycline, amiodarone and tamoxifen (drug-induced hepatic steatosis). Mechanistic knowledge on chemical-induced steatosis has greatly evolved and has been organized into adverse outcome pathways (AOPs) describing the chain of events from first molecular interaction of a substance with a biological system to the adverse outcome, intrahepatic lipid accumulation. In this study, three known steatosis-inducing pesticides (imazalil, clothianidin, and thiacloprid) were tested for their ability to induce hepatic triglyceride accumulation in the zebrafish (Danio rerio) embryo (ZFE) at 5 days post fertilization, both as single compounds and equipotent binary mixtures. The results indicate that the ZFE is very well applicable as a higher tier testing model to confirm effects in downstream key events in AOPs, that is, chemically-induced triglyceride accumulation in the whole organism and production of visible steatosis. Moreover, dose addition could be concluded for binary mixtures of substances with similar and with dissimilar modes of action.
Collapse
Affiliation(s)
- Harm J Heusinkveld
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands.
| | - Edwin P Zwart
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Angela de Haan
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Albert Braeuning
- Department of Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, Berlin 10589, Germany
| | - Jimmy Alarcan
- Department of Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, Berlin 10589, Germany
| | - Leo T M van der Ven
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| |
Collapse
|
2
|
Bérubé R, Murray B, Kocarek TA, Gurdziel K, Kassotis CD. Nonylphenol and Cetyl Alcohol Polyethoxylates Disrupt Thyroid Hormone Receptor Signaling to Disrupt Metabolic Health. Endocrinology 2024; 165:bqae149. [PMID: 39497475 PMCID: PMC11574291 DOI: 10.1210/endocr/bqae149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024]
Abstract
Surfactants are molecules with both hydrophobic and hydrophilic structural groups that adsorb at the air-water or oil-water interface and serve to decrease the surface tension. Surfactants combine to form micelles that surround and break down or remove oils, making them ideal for detergents and cleaners. Two of the most important classes of nonionic surfactants are alkylphenol ethoxylates (APEOs) and alcohol ethoxylates (AEOs). APEOs and AEOs are high production-volume chemicals that are used for many industrial and residential purposes, including laundry detergents, hard-surface cleaners, paints, and pesticide adjuvants. Commensurate with better appreciation of the toxicity of APEOs and the base alkylphenols, use of AEOs has increased, and both sets of compounds are now ubiquitous environmental contaminants. We recently demonstrated that diverse APEOs and AEOs induce triglyceride accumulation and/or preadipocyte proliferation in vitro. Both sets of contaminants have also been demonstrated as obesogenic and metabolism-disrupting in a developmental exposure zebrafish model. While these metabolic health effects are consistent across models and species, the mechanisms underlying these effects are less clear. This study sought to evaluate causal mechanisms through reporter gene assays, relative binding affinity assays, coexposure experiments, and use of both human cell and zebrafish models. We report that antagonism of thyroid hormone receptor signaling appears to mediate at least a portion of the polyethoxylate-induced metabolic health effects. These results suggest further evaluation is needed, given the ubiquitous environmental presence of these thyroid-disrupting contaminants and reproducible effects in human cell models and vertebrate animals.
Collapse
Affiliation(s)
- Roxanne Bérubé
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, USA
| | - Brooklynn Murray
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, USA
| | - Thomas A Kocarek
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, USA
| | - Katherine Gurdziel
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, USA
- Genome Sciences Core, Wayne State University, Detroit, MI 48202, USA
| | - Christopher D Kassotis
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
3
|
Tian Z, Wei M, Xue R, Song L, Li H, Ji H, Xiao P, Sun J. Comparative study on the transcriptional activities of grass carp (Ctenopharyngodon idellus) peroxisome proliferator-activated receptors induced by different fatty acids. Comp Biochem Physiol B Biochem Mol Biol 2024; 275:111021. [PMID: 39151662 DOI: 10.1016/j.cbpb.2024.111021] [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: 04/11/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that are part of the nuclear hormone receptor family, playing a crucial role in gene expression regulation. They serve as a connection between lipid metabolism disorders and innate immunity by being activated by fatty acids and their derivatives, facilitating signal transduction between the cell surface and nucleus. However, the specific transcriptional effects of different fatty acids (FAs) in fish are not yet fully understood. In our research, we identified and characterized PPARs in grass carp (Ctenopharyngodon idellus). The complete coding sequences of pparαa, pparαb, pparγ, pparδa, and pparδb were 1443 bp, 1404 bp, 1569 bp, 1551 bp, and 1560 bp in length, respectively. Pparα showed the highest expression in the liver, pparγ was mainly expressed in abdominal adipose tissue, and pparδ exhibited increased expression in the heart compared to other tissues. Gene localization analysis revealed that only pparδa was present in both the nucleus and cytoplasm, while the other four genes were exclusively located in the nucleus. Furthermore, our study explored the influence of various fatty acids (docosahexaenoic acid, palmitic acid, lauric acid and oleic acid at concentrations of 0, 50, 100, and 200 μM) on the transcriptional activities of different PPARs, demonstrating the diverse effects of fatty acid ligands on PPAR transcriptional activity. These results have significant implications for understanding the regulation of PPARs transcriptional activity.
Collapse
Affiliation(s)
- Zhiqi Tian
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Mingkui Wei
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Rongrong Xue
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Lei Song
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Handong Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Hong Ji
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Peizhen Xiao
- Beijing Sunpu Biochemical and Technology Co., Ltd., Beijing 100126, China
| | - Jian Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, China.
| |
Collapse
|
4
|
Gutsfeld S, Wehmas L, Omoyeni I, Schweiger N, Leuthold D, Michaelis P, Howey XM, Gaballah S, Herold N, Vogs C, Wood C, Bertotto L, Wu GM, Klüver N, Busch W, Scholz S, Schor J, Tal T. Investigation of Peroxisome Proliferator-Activated Receptor Genes as Requirements for Visual Startle Response Hyperactivity in Larval Zebrafish Exposed to Structurally Similar Per- and Polyfluoroalkyl Substances (PFAS). ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:77007. [PMID: 39046251 PMCID: PMC11268134 DOI: 10.1289/ehp13667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Per- and polyfluoroalkyl Substances (PFAS) are synthetic chemicals widely detected in humans and the environment. Exposure to perfluorooctanesulfonic acid (PFOS) or perfluorohexanesulfonic acid (PFHxS) was previously shown to cause dark-phase hyperactivity in larval zebrafish. OBJECTIVES The objective of this study was to elucidate the mechanism by which PFOS or PFHxS exposure caused hyperactivity in larval zebrafish. METHODS Swimming behavior was assessed in 5-d postfertilization (dpf) larvae following developmental (1-4 dpf) or acute (5 dpf) exposure to 0.43 - 7.86 μ M PFOS, 7.87 - 120 μ M PFHxS, or 0.4% dimethyl sulfoxide (DMSO). After developmental exposure and chemical washout at 4 dpf, behavior was also assessed at 5-8 dpf. RNA sequencing was used to identify differences in global gene expression to perform transcriptomic benchmark concentration-response (BMC T ) modeling, and predict upstream regulators in PFOS- or PFHxS-exposed larvae. CRISPR/Cas9-based gene editing was used to knockdown peroxisome proliferator-activated receptors (ppars) pparaa/ab, pparda/db, or pparg at day 0. Knockdown crispants were exposed to 7.86 μ M PFOS or 0.4% DMSO from 1-4 dpf and behavior was assessed at 5 dpf. Coexposure with the ppard antagonist GSK3787 and PFOS was also performed. RESULTS Transient dark-phase hyperactivity occurred following developmental or acute exposure to PFOS or PFHxS, relative to the DMSO control. In contrast, visual startle response (VSR) hyperactivity only occurred following developmental exposure and was irreversible up to 8 dpf. Similar global transcriptomic profiles, BMC T estimates, and enriched functions were observed in PFOS- and PFHxS-exposed larvae, and ppars were identified as putative upstream regulators. Knockdown of pparda/db, but not pparaa/ab or pparg, blunted PFOS-dependent VSR hyperactivity to control levels. This finding was confirmed via antagonism of ppard in PFOS-exposed larvae. DISCUSSION This work identifies a novel adverse outcome pathway for VSR hyperactivity in larval zebrafish. We demonstrate that developmental, but not acute, exposure to PFOS triggered persistent VSR hyperactivity that required ppard function. https://doi.org/10.1289/EHP13667.
Collapse
Affiliation(s)
- Sebastian Gutsfeld
- Department of Bioanalytical Ecotoxicology, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
| | - Leah Wehmas
- Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Ifeoluwa Omoyeni
- Department of Bioanalytical Ecotoxicology, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
| | - Nicole Schweiger
- Department of Bioanalytical Ecotoxicology, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
| | - David Leuthold
- Department of Bioanalytical Ecotoxicology, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
| | - Paul Michaelis
- Department of Bioanalytical Ecotoxicology, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
| | - Xia Meng Howey
- Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Shaza Gaballah
- Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Nadia Herold
- Department of Bioanalytical Ecotoxicology, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
| | - Carolina Vogs
- Department of Biomedical Science and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Carmen Wood
- Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Luísa Bertotto
- Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Gi-Mick Wu
- Research and Development Institute for the Agri-Environment, Quebec, Quebec, Canada
| | - Nils Klüver
- Department of Bioanalytical Ecotoxicology, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
| | - Wibke Busch
- Department of Bioanalytical Ecotoxicology, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
| | - Stefan Scholz
- Department of Bioanalytical Ecotoxicology, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
| | - Jana Schor
- Department of Computational Biology and Chemistry, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
| | - Tamara Tal
- Department of Bioanalytical Ecotoxicology, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
- Medical Faculty, University Leipzig, Leipzig, Germany
| |
Collapse
|
5
|
Saini T, Mazumder PM. Current advancement in the preclinical models used for the assessment of diabetic neuropathy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2727-2745. [PMID: 37987794 DOI: 10.1007/s00210-023-02802-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/17/2023] [Indexed: 11/22/2023]
Abstract
Diabetic neuropathy is one of the prevalent and debilitating microvascular complications of diabetes mellitus, affecting a significant portion of the global population. Relational preclinical animal models are essential to understand its pathophysiology and develop effective treatments. This abstract provides an overview of current knowledge and advancements in such models. Various animal models have been developed to mimic the multifaceted aspects of human diabetic neuropathy, including both type 1 and type 2 diabetes. These models involve rodents (rats and mice) and larger animals like rabbits and dogs. Induction of diabetic neuropathy in these models is achieved through chemical, genetic, or dietary interventions, such as diabetogenic agents, genetic modifications, or high-fat diets. Preclinical animal models have greatly contributed to studying the intricate molecular and cellular mechanisms underlying diabetic neuropathy. They have shed light on hyperglycemia-induced oxidative stress, neuroinflammation, mitochondrial dysfunction, and altered neurotrophic factor signaling. Additionally, these models have allowed for the investigation of morphological changes, functional alterations, and behavioral manifestations associated with diabetic neuropathy. These models have also been crucial for evaluating the efficacy and safety of potential therapeutic interventions. Novel pharmacological agents, gene therapies, stem cell-based approaches, exercise, dietary modifications, and neurostimulation techniques have been tested using these models. However, limitations and challenges remain, including physiological differences between humans and animals, complex neuropathy phenotypes, and the need for translational validation. In conclusion, preclinical animal models have played a vital role in advancing our understanding and management of diabetic neuropathy. They have enhanced our knowledge of disease mechanisms, facilitated the development of novel treatments, and provided a platform for translational research. Ongoing efforts to refine and validate these models are crucial for future treatment developments for this debilitating condition.
Collapse
Affiliation(s)
- Tanishk Saini
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, 835215, Ranchi, India
| | - Papiya Mitra Mazumder
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, 835215, Ranchi, India.
| |
Collapse
|
6
|
Liu A, Chen C, Chen K, Shi Y, Grabowski RC, Qiu X. Effects of parental exposure to amitriptyline on the survival, development, behavior, and gene expression in zebrafish offspring. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:169173. [PMID: 38064809 DOI: 10.1016/j.scitotenv.2023.169173] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/28/2023] [Accepted: 12/05/2023] [Indexed: 01/18/2024]
Abstract
In mammals, parental exposure to amitriptyline (AMI) has been proven to contribute to congenital disabilities in their offspring. However, no studies have paid attention to the adverse effects of parental exposure to amitriptyline on fish offspring. In this study, we exposed adult zebrafish (F0) to AMI (0.8 μg/L) for 21 days. Subsequently, these zebrafish (F0) were allowed to mate, and their offspring (F1) were collected to culture in clean water for 5 days. The mortality rate, average hatching time, and heart rate at 48 h post-fertilization (hpf) of F1 were investigated. Our results showed that parental exposure to AMI induced tachycardia and increased mortality in F1 zebrafish. Under a light/dark transition test, F1 larvae born from AMI-exposed parents exhibited lower locomotor activity in the dark period and decreased thigmotaxis in the light period. The transcriptome analysis showed that parental AMI exposure dysregulated some key pathways in their offspring. Through the prediction of key driver analysis, six differentially expressed genes (DEGs) were revealed as key driver genes involved in protein processing in endoplasmic reticulum (hspa5, hsp70.1, hsp90a), ribosome (rps27a) and PPAR signaling pathway (pparab and fabp2). Considering that the concentration of AMI residual components in natural water bodies may be over our test concentration (0.8 μg/L), our findings suggested that toxicity of parental exposure to the offspring of fish should receive greater attention.
Collapse
Affiliation(s)
- Anqi Liu
- Institute of Environmental Health and Ecological Security, School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Chen Chen
- Institute of Environmental Health and Ecological Security, School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Kun Chen
- Institute of Environmental Health and Ecological Security, School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yanhong Shi
- Institute of Environmental Health and Ecological Security, School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Robert C Grabowski
- Centre for Water, Environment and Development, Cranfield University, Cranfield, Bedfordshire MK43 0AL, UK
| | - Xuchun Qiu
- Institute of Environmental Health and Ecological Security, School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Jiangsu Collaborative Innovation Center of Technology and Material of Water Treatment, Suzhou University of Science and Technology, Suzhou 215009, China.
| |
Collapse
|
7
|
Ünal İ, Cansız D, Beler M, Sezer Z, Güzel E, Emekli-Alturfan E. Sodium-dependent glucose co-transporter-2 inhibitor empagliflozin exerts neuroprotective effects in rotenone-induced Parkinson's disease model in zebrafish; mechanism involving ketogenesis and autophagy. Brain Res 2023; 1820:148536. [PMID: 37591458 DOI: 10.1016/j.brainres.2023.148536] [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: 04/28/2023] [Revised: 06/26/2023] [Accepted: 08/14/2023] [Indexed: 08/19/2023]
Abstract
Sodium-dependent glucose co-transporter-2 (SGLT2) inhibitor empagliflozin (EMP), is the new class of oral hypoglycemic agent approved as a treatment for Type 2 diabetes. SGLT2 inhibitors may induce ketogenesis through inhibiting the renal reabsorption of glucose. In recent years, positive effects of ketogenic diets on neurodegenerative diseases such as Parkinson's disease (PD) have been reported by improving autophagy. We aimed to evaluate the effects of EMP treatment as a SGLT2 inhibitor that can mimic the effects of ketogenic diet, in rotenone induced PD model in zebrafish focusing on ketogenesis, autophagy, and molecular pathways related with PD progression including oxidative stress and inflammation. Adult zebrafish were exposed to rotenone and EMP for 30 days. Y-Maze task and locomotor analysis were performed. Neurotransmitter levels were determined by liquid chromatography tandem- mass spectrometry (LC-MS/MS). Lipid peroxidation (LPO), nitric oxide (No), alkaline phosphatase, superoxide dismutase, glutathione, glutathione S-transferase (GST), sialic acid, acetylcholinesterase, and the expressions of autophagy, ketogenesis and PD-related genes were determined. Immunohistochemical staining was performed for the microglial marker L-plastin (Lcp1) and tyrosine hydroxylase (Th). EMP treatment improved DOPAC/DA ratio, Y-Maze task, locomotor activity, expressions of Th and Lcp-1, autophagy and inflammation related (mTor, atg5, tnfα, sirt1, il6, tnfα); PD-related (lrrk2, park2, park7, pink1), and ketone metabolism-related genes (slc16a1b, pparag, and pparab), and oxidant-damage in brain in the rotenone group as evidenced by decreased LPO, No, and improved antioxidant molecules. Our results showed benefical effects of EMP as a SGLT2 inhibitor in neurotoxin-induced PD model in zebrafish. We believe our study, will shed light on the mechanism of the effects of SGLT2 inhibitors, ketogenesis and autopahgy in PD.
Collapse
Affiliation(s)
- İsmail Ünal
- Marmara University, Institute of Health Sciences, Faculty of Pharmacy, Department of Biochemistry, Istanbul, Turkey
| | - Derya Cansız
- Department Medipol University, Faculty of Medicine, Medical Biochemistry, Istanbul, Turkey
| | - Merih Beler
- Marmara University, Institute of Health Sciences, Faculty of Pharmacy, Department of Biochemistry, Istanbul, Turkey
| | - Zehra Sezer
- Department of Histology and Embryology, Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Istanbul 34098, Turkey
| | - Elif Güzel
- Department of Histology and Embryology, Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Istanbul 34098, Turkey
| | - Ebru Emekli-Alturfan
- Marmara University, Faculty of Dentistry, Department of Basic Medical Sciences, Istanbul, Turkey.
| |
Collapse
|
8
|
Chen S, Wang X, Yan J, Wang Z, Qian Q, Wang H. Mechanistic illustration on lipid-metabolism disorders induced by triclosan exposure from the viewpoint of m 6A-RNA epigenetic modification. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 901:165953. [PMID: 37536604 DOI: 10.1016/j.scitotenv.2023.165953] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/23/2023] [Accepted: 07/30/2023] [Indexed: 08/05/2023]
Abstract
As a typically anthropogenic contaminant, the toxicity effects of triclosan (TCS) were investigated in-depth from the viewpoint of m6A-pre-miRNAs modification. Based on miRNAs high-throughput sequencing, we unravelled the underlying molecular mechanisms regarding TCS-induced lipid-metabolism functional disorders. TCS exposure caused severe lipid accumulation in 120 hpf zebrafish liver and reduced their locomotor activity. Both bioinformatics analysis and experimental validation verified that TCS targeted miR-27b up-regulation to further trigger lipid-metabolism disorders and developmental malformations, including shortened body length, yolk cysts, curved spine and delayed yolk absorption. TCS exposure and miR-27b upregulation both caused the enhanced levels of triglyceride and total cholesterol. Knockdown and overexpression of miR-27b regulated the expression changes of several functional genes related to downstream lipid metabolism of miR-27b, and most downstream target genes of miR-27b were suppressed and enriched in the AMPK signaling pathway. The experiments of pathway inhibitors and agonists further evidenced that TCS caused lipid-metabolism disorders by suppressing the AMPK signaling pathway. In upstream of miR-27b, TCS decreased total m6A-RNA level by targeting upregulation of demethylase and downregulation of methylase reader ythdf1. Molecular docking and ythdf1 siRNA interference further confirmed that TCS targeted the expression change of ythdf1. Under ythdf1 knockdown in upstream of miR-27b, both abnormal lipid metabolism and miR-27b upregulation highlighted that TCS-induced lipid-metabolism disorders were attributable to the decreasing m6A-RNA methylation levels in vivo. These perspectives provide an innovative idea for prevention and treatment of the lipid metabolism-related diseases and these findings open a novel avene for TCS's risk assessment and early intervention of the contaminant.
Collapse
Affiliation(s)
- Shuya Chen
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Xuedong Wang
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Jin Yan
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Zejun Wang
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Qiuhui Qian
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Huili Wang
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China.
| |
Collapse
|
9
|
Dong Y, Wei Y, Wang L, Song K, Zhang C, Lu K, Rahimnejad S. Dietary n-3/n-6 polyunsaturated fatty acid ratio modulates growth performance in spotted seabass ( Lateolabrax maculatus) through regulating lipid metabolism, hepatic antioxidant capacity and intestinal health. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2023; 14:20-31. [PMID: 37234947 PMCID: PMC10208799 DOI: 10.1016/j.aninu.2023.04.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 01/12/2023] [Accepted: 04/04/2023] [Indexed: 05/28/2023]
Abstract
An 8-week feeding experiment was carried out to explore the effects of dietary n-3/n-6 polyunsaturated fatty acid (PUFA) ratio on growth performance, lipid metabolism, hepatic antioxidant status, and gut flora of spotted seabass (Lateolabrax maculatus). Six experimental diets were formulated to contain different levels of two purified oil sources including docosahexaenoic and eicosapentaenoic acids enriched oil (n-3) and linoleic acid-enriched oil (n-6) leading to n-3/n-6 PUFA ratios of 0.04, 0.35, 0.66, 1.35, 2.45 and 16.17. Each diet was fed to triplicate groups of juvenile L. maculatus (11.06 ± 0.20 g, 30 fish/tank). Final body weight (FBW), weight gain (WG), specific growth rates (SGR), protein efficiency ratio (PER) and feed utilization efficiency increased as n-3/n-6 PUFA ratio increased up to a certain level, and then decreased thereafter. Fish fed the diet with n-3/n-6 PUFA ratio of 0.66 exhibited the highest FBW, WG, SGR and PER and the lowest feed conversion ratio. Lower n-3/n-6 PUFA ratios induced up-regulated expression of lipid synthesis-related genes (fas, acc2 and srebp-1c) and down-regulated expression of lipolysis related genes (atgl, pparα, cpt-1 and aox). Higher expression of lipolysis-related genes (atgl, pparα and cpt-1) was recorded at moderate n-3/n-6 PUFA ratios (0.66 to 1.35). Moreover, inappropriate n-3/n-6 PUFA ratios triggered up-regulation of pro-inflammatory genes (il-6 and tnf-α) and down-regulation of anti-inflammatory genes (il-4 and il-10) in the intestine. The diet with n-3/n-6 PUFA ratio of 0.66 inhibited intestine inflammation, improved intestinal flora richness, increased the abundance of beneficial bacteria such as Lactobacillus, Alloprevotella and Ruminococcus, and reduced the abundance of harmful bacteria including Escherichia-Shigella and Enterococcus. In summary, it could be suggested that a dietary n-3/n-6 PUFA ratio of 0.66 can improve growth performance and feed utilization in L. maculatus, as is deemed to be mediated through regulation of lipid metabolism and intestinal flora.
Collapse
Affiliation(s)
- Yanzou Dong
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen 361021, China
| | - Yu Wei
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen 361021, China
| | - Ling Wang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen 361021, China
| | - Kai Song
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen 361021, China
| | - Chunxiao Zhang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen 361021, China
| | - Kangle Lu
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen 361021, China
| | - Samad Rahimnejad
- Faculty of Fisheries and Protection of Waters, University of South Bohemia in České Budějovice, South Bohemian Research Centre of Aquaculture and Biodiversity of Hydrocenoses, Vodňany, Zátiší 728, Vodňany 389 25, Czech Republic
- Fish Innate Immune System Group, Department of Cell Biology and Histology, Faculty of Biology, Regional Campus of International Excellence “Campus Mare Nostrum”, University of Murcia, Murcia 30100, Spain
| |
Collapse
|
10
|
Yuan M, Chen S, Zeng C, Fan Y, Ge W, Chen W. Estrogenic and non-estrogenic effects of bisphenol A and its action mechanism in the zebrafish model: An overview of the past two decades of work. ENVIRONMENT INTERNATIONAL 2023; 176:107976. [PMID: 37236126 DOI: 10.1016/j.envint.2023.107976] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/11/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023]
Abstract
Bisphenol A (BPA) is the most simple and predominant component of the Bisphenol family. BPA is widely present in the environment and the human body as a result of its extensive usage in the plastic and epoxy resins of consumer goods like water bottles, food containers, and tableware. Since the 1930s, when BPA's estrogenic activity was first observed, and it was labeled as a "mimic hormone of E2", studies on the endocrine-disrupting effects of BPA then have been widely conducted. As a top vertebrate model for genetic and developmental studies, the zebrafish has caught tremendous attention in the past two decades. By using the zebrafish, the negative effects of BPA either through estrogenic signaling pathways or non-estrogenic signaling pathways were largely found. In this review, we tried to draw a full picture of the current state of knowledge on the estrogenic and non-estrogenic effects of BPA with their mechanisms of action through the zebrafish model of the past two decades, which may help to fully understand the endocrine-disrupting effects of BPA and its action mechanism, and give a direction for the future studies.
Collapse
Affiliation(s)
- Mingzhe Yuan
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Shan Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Chu Zeng
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Yuqin Fan
- Guangdong Provincial Key Laboratory of Conservation and Precision Utilization of Characteristic Agricultural Resources in Mountainous Area, School of Life Sciences, Jiaying University, Meizhou 514015, China
| | - Wei Ge
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| | - Weiting Chen
- Guangdong Provincial Key Laboratory of Conservation and Precision Utilization of Characteristic Agricultural Resources in Mountainous Area, School of Life Sciences, Jiaying University, Meizhou 514015, China.
| |
Collapse
|
11
|
Porto VA, da Rocha Júnior ER, Ursulino JS, Porto RS, da Silva M, de Jesus LWO, Oliveira JMD, Crispim AC, Santos JCC, Aquino TMD. NMR-based metabolomics applied to ecotoxicology with zebrafish (Danio rerio) as a prominent model for metabolic profiling and biomarker discovery: Overviewing the most recent approaches. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 868:161737. [PMID: 36693575 DOI: 10.1016/j.scitotenv.2023.161737] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/28/2022] [Accepted: 01/17/2023] [Indexed: 06/17/2023]
Abstract
Metabolomics is an innovative approach used in the medical, toxicological, and biological sciences. As an interdisciplinary topic, metabolomics and its relation with the environment and toxicological research are extensive. The use of substances, such as drugs and pesticides, contributes to the continuous releasing of xenobiotics into the environment, harming organisms and their habitats. In this context, fish are important bioindicators of the environmental condition and have often been used as model species. Among them, zebrafish (Danio rerio) presents itself as a versatile and straightforward option due to its unique attributes for research. Zebrafish proves to be a valuable model for toxicity assays and also for metabolomics profiling by analytical tools. Thus, NMR-based metabolomics associated with statistical analysis can reasonably assist researchers in critical factors related to discovering and validating biomarkers through accurate diagnosis. Therefore, this review aimed to report the studies that applied zebrafish as a model for (eco)toxicological assays and essentially utilized NMR-based metabolomics analysis to assess the biochemical profile and thus suggest the potential biological marker.
Collapse
Affiliation(s)
- Viviane Amaral Porto
- Research Group on Therapeutic Strategies, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, AL, Brazil.
| | | | - Jeferson Santana Ursulino
- Research Group on Therapeutic Strategies, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, AL, Brazil
| | - Ricardo Silva Porto
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, AL, Brazil
| | - Marciliano da Silva
- Laboratory of Applied Animal Morphophysiology, Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió, AL, Brazil
| | - Lázaro Wender Oliveira de Jesus
- Laboratory of Applied Animal Morphophysiology, Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió, AL, Brazil
| | | | - Alessandre Carmo Crispim
- Research Group on Therapeutic Strategies, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, AL, Brazil
| | | | - Thiago Mendonça de Aquino
- Research Group on Therapeutic Strategies, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, AL, Brazil
| |
Collapse
|
12
|
Analyses of regulatory network and discovery of potential biomarkers for Korean rockfish (Sebastes schlegelii) in responses to starvation stress through transcriptome and metabolome. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2023; 46:101061. [PMID: 36796184 DOI: 10.1016/j.cbd.2023.101061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 01/29/2023] [Accepted: 01/31/2023] [Indexed: 02/11/2023]
Abstract
Whether in aquaculture or in nature, starvation stress limits the growth of fish. The purpose of the study was to clarify the detailed molecular mechanisms underlying starvation stress in Korean rockfish (Sebastes schlegelii) through liver transcriptome and metabolome analysis. Transcriptome results showed that liver genes associated with cell cycle and fatty acid synthesis were down-regulated, whereas those related to fatty acid decomposition were up-regulated in the experimental group (EG; starved for 72 days) compared to the control group (CG; feeding). Metabolomic results showed that there were significant differences in the levels of metabolites related to nucleotide metabolism and energy metabolism, such as purine metabolism, histidine metabolism and oxidative phosphorylation. Five fatty acids (C22:6n-3; C22:5n-3; C20:5n-3; C20:4n-3; C18:3n-6) were selected as possible biomarkers of starvation stress from the differential metabolites of metabolome. Subsequently, correlation between these differential genes of lipid metabolism and cell cycle and differential metabolites were analyzed, and observed that these five fatty acids were significantly correlated with the differential genes. These results provide new clues for understanding the role of fatty acid metabolism and cell cycle in fish under starvation stress. It also provides a reference for promoting the biomarker identification of starvation stress and stress tolerance breeding research.
Collapse
|
13
|
Lei Y, Li F, Mortimer M, Li Z, Peng BX, Li M, Guo LH, Zhuang G. Antibiotics disrupt lipid metabolism in zebrafish (Danio rerio) larvae and 3T3-L1 preadipocytes. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 858:159755. [PMID: 36349636 DOI: 10.1016/j.scitotenv.2022.159755] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/19/2022] [Accepted: 10/23/2022] [Indexed: 06/16/2023]
Abstract
Antibiotics are emerging environmental contaminants with wide attention due to their high consumption and pseudo-persistence in the environment. They have been shown to induce obesity or obesity-related metabolic diseases in experimental animals, but the underlying toxicological mechanisms remain unclear. Here, the disruptive effects of four commonly used antibiotics, namely doxycycline (DC), enrofloxacin (ENR), florfenicol (FF) and sulfamethazine (SMT) on lipid metabolism were investigated in zebrafish (Danio rerio) larvae and murine preadipocyte cell line. Triglyceride (TG) content was reduced after 1 ng/L DC or ENR exposure but was increased at higher concentrations up to 100 mg/L. FF increased and SMT reduced TG content but did not show any concentration dependence. None of the antibiotics had any significant effect on total cholesterol (TC) content in zebrafish except 100 μg/L SMT. Expression levels of 8 lipid metabolism-related genes were also quantified. SMT was most disruptive by up-regulating six genes, followed by FF which up-regulated four genes and down-regulated one gene, whereas DC and ENR both up-regulated one gene. In 3T3-L1 preadipocytes, ENR, FF, and SMT in general increased TG content, while 100 mg/L FF reduced TG substantially. DC did not show any effect up to 10 mg/L, at which TG increased significantly. FF and SMT increased TC slightly at low concentrations but reduced it at high concentrations, whereas TC, DC and ENR had no effect at any tested concentrations. Gene expression measurement also indicated that SMT was most disruptive, followed by FF, DC, and ENR. Reporter gene assays showed that only SMT inhibited the transcriptional activity of peroxisome proliferator-activated receptor γ (PPARγ). The above experimental results and clustering analysis demonstrate that the four antibiotics exerted disruption on lipid metabolism through different mechanisms, and one of the mechanisms for SMT may be inhibition of PPARγ transcriptional activity.
Collapse
Affiliation(s)
- Yuyang Lei
- College of Life Sciences, China Jiliang University, Hangzhou, Zhejiang 310018, China; Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou, Zhejiang 310018, China
| | - Fangfang Li
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou, Zhejiang 310018, China; College of Quality and Safety Engineering, China Jiliang University, Hangzhou, Zhejiang 310018, China
| | - Monika Mortimer
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou, Zhejiang 310018, China; College of Quality and Safety Engineering, China Jiliang University, Hangzhou, Zhejiang 310018, China
| | - Zhi Li
- College of Life Sciences, China Jiliang University, Hangzhou, Zhejiang 310018, China; Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou, Zhejiang 310018, China
| | - Bi-Xia Peng
- College of Life Sciences, China Jiliang University, Hangzhou, Zhejiang 310018, China; Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou, Zhejiang 310018, China
| | - Minjie Li
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou, Zhejiang 310018, China; College of Quality and Safety Engineering, China Jiliang University, Hangzhou, Zhejiang 310018, China.
| | - Liang-Hong Guo
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou, Zhejiang 310018, China; College of Quality and Safety Engineering, China Jiliang University, Hangzhou, Zhejiang 310018, China.
| | - Guoqiang Zhuang
- Research Center for Eco-Environmental Sciences, Chinese Academy of Science, Beijing 100085, China
| |
Collapse
|
14
|
Kim M, Rizvi F, Shin D, Gouon-Evans V. Update on Hepatobiliary Plasticity. Semin Liver Dis 2023; 43:13-23. [PMID: 36764306 PMCID: PMC10005859 DOI: 10.1055/s-0042-1760306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
The liver field has been debating for decades the contribution of the plasticity of the two epithelial compartments in the liver, hepatocytes and biliary epithelial cells (BECs), to derive each other as a repair mechanism. The hepatobiliary plasticity has been first observed in diseased human livers by the presence of biphenotypic cells expressing hepatocyte and BEC markers within bile ducts and regenerative nodules or budding from strings of proliferative BECs in septa. These observations are not surprising as hepatocytes and BECs derive from a common fetal progenitor, the hepatoblast, and, as such, they are expected to compensate for each other's loss in adults. To investigate the cell origin of regenerated cell compartments and associated molecular mechanisms, numerous murine and zebrafish models with ability to trace cell fates have been extensively developed. This short review summarizes the clinical and preclinical studies illustrating the hepatobiliary plasticity and its potential therapeutic application.
Collapse
Affiliation(s)
- Minwook Kim
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Fatima Rizvi
- Department of Medicine, Gastroenterology Section, Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, Massachusetts
| | - Donghun Shin
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Valerie Gouon-Evans
- Department of Medicine, Gastroenterology Section, Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, Massachusetts
| |
Collapse
|
15
|
Bomkamp C, Musgrove L, Marques DMC, Fernando GF, Ferreira FC, Specht EA. Differentiation and Maturation of Muscle and Fat Cells in Cultivated Seafood: Lessons from Developmental Biology. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2023; 25:1-29. [PMID: 36374393 PMCID: PMC9931865 DOI: 10.1007/s10126-022-10174-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 10/10/2022] [Indexed: 06/16/2023]
Abstract
Cultivated meat, also known as cultured or cell-based meat, is meat produced directly from cultured animal cells rather than from a whole animal. Cultivated meat and seafood have been proposed as a means of mitigating the substantial harms associated with current production methods, including damage to the environment, antibiotic resistance, food security challenges, poor animal welfare, and-in the case of seafood-overfishing and ecological damage associated with fishing and aquaculture. Because biomedical tissue engineering research, from which cultivated meat draws a great deal of inspiration, has thus far been conducted almost exclusively in mammals, cultivated seafood suffers from a lack of established protocols for producing complex tissues in vitro. At the same time, fish such as the zebrafish Danio rerio have been widely used as model organisms in developmental biology. Therefore, many of the mechanisms and signaling pathways involved in the formation of muscle, fat, and other relevant tissue are relatively well understood for this species. The same processes are understood to a lesser degree in aquatic invertebrates. This review discusses the differentiation and maturation of meat-relevant cell types in aquatic species and makes recommendations for future research aimed at recapitulating these processes to produce cultivated fish and shellfish.
Collapse
Affiliation(s)
- Claire Bomkamp
- Department of Science & Technology, The Good Food Institute, Washington, DC USA
| | - Lisa Musgrove
- University of the Sunshine Coast, Sippy Downs, Queensland Australia
| | - Diana M. C. Marques
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Gonçalo F. Fernando
- Department of Science & Technology, The Good Food Institute, Washington, DC USA
| | - Frederico C. Ferreira
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Elizabeth A. Specht
- Department of Science & Technology, The Good Food Institute, Washington, DC USA
| |
Collapse
|
16
|
Printzi A, Mazurais D, Witten PE, Madec L, Gonzalez AA, Mialhe X, Zambonino-Infante JL, Koumoundouros G. Juvenile zebrafish (Danio rerio) are able to recover from lordosis. Sci Rep 2022; 12:21533. [PMID: 36513797 PMCID: PMC9748118 DOI: 10.1038/s41598-022-26112-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Haemal lordosis, a frequent skeletal deformity in teleost fish, has long been correlated with increased mechanical loads induced by swimming activity. In the present study, we examine whether juvenile zebrafish can recover from haemal lordosis and explore the musculoskeletal mechanisms involved. Juveniles were subjected to a swimming challenge test (SCT) that induced severe haemal lordosis in 49% of the animals and then immediately transferred them to 0.0 total body lengths (TL) per second of water velocity for a week. The recovery from lordosis was examined by means of whole mount staining, histology and gene expression analysis. Results demonstrate that 80% of the lordotic zebrafish are capable of internal and external recovery within a week after the SCT. Recovered individuals presented normal shape of the vertebral centra, maintaining though distorted internal tissue organization. Through the transcriptomic analysis of the affected haemal regions, several processes related to chromosome organization, DNA replication, circadian clock and transcription regulation were enriched within genes significantly regulated behind this musculoskeletal recovery procedure. Genes especially involved in adipogenesis, bone remodeling and muscular regeneration were regulated. A remodeling tissue-repair hypothesis behind haemal lordosis recovery is raised. Limitations and future possibilities for zebrafish as a model organism to clarify mechanically driven musculoskeletal changes are discussed.
Collapse
Affiliation(s)
- A. Printzi
- grid.8127.c0000 0004 0576 3437Biology Department, University of Crete, Crete, Greece ,grid.463763.30000 0004 0638 0577IFREMER, University of Brest, CNRS, IRD, LEMAR, 29280 Plouzané, France
| | - D. Mazurais
- grid.463763.30000 0004 0638 0577IFREMER, University of Brest, CNRS, IRD, LEMAR, 29280 Plouzané, France
| | - P. E. Witten
- grid.5342.00000 0001 2069 7798Department of Biology, Gent University, Gent, Belgium
| | - L. Madec
- grid.463763.30000 0004 0638 0577IFREMER, University of Brest, CNRS, IRD, LEMAR, 29280 Plouzané, France
| | - A.-A. Gonzalez
- grid.121334.60000 0001 2097 0141MGX-Montpellier GenomiX, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - X. Mialhe
- grid.121334.60000 0001 2097 0141MGX-Montpellier GenomiX, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - J.-L. Zambonino-Infante
- grid.463763.30000 0004 0638 0577IFREMER, University of Brest, CNRS, IRD, LEMAR, 29280 Plouzané, France
| | - G. Koumoundouros
- grid.8127.c0000 0004 0576 3437Biology Department, University of Crete, Crete, Greece
| |
Collapse
|
17
|
Li X, Liu S, Qi D, Qi H, Wang Y, Zhao K, Tian F. Genome-wide identification and expression of the peroxisome proliferator-activated receptor gene family in the Tibetan highland fish Gymnocypris przewalskii. FISH PHYSIOLOGY AND BIOCHEMISTRY 2022; 48:1685-1699. [PMID: 36469183 DOI: 10.1007/s10695-022-01152-7] [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: 01/20/2022] [Accepted: 11/27/2022] [Indexed: 06/17/2023]
Abstract
Peroxisome proliferator-activated receptor (PPAR) plays an important role in the regulation of lipid metabolism and has been widely identified in diverse species. Gymnocypris przewalskii is a native fish of the Qinghai Tibetan Plateau that survives in a chronically cold environment. In the current study, we conducted genome-wide identification of PPAR genes, revealing the existence of seven PPARs in the G. przewalskii genome. Collinearity was observed between two copies of PPARαb and PPARγ in G. przewalskii, suggesting that the additional copy might be gained through whole genome duplication. Both phylogenetic and multiple sequence alignment analyses indicated that PPARs in G. przewalskii were conserved with teleosts. The cold treatment (10 °C and 4 °C) led to the developmental delay of G. przewalskii embryos. Continuous expression of PPARs was observed during the embryonic development of G. przewalskii under normal and cold conditions, with significantly different transcriptional patterns. These results indicated that PPARs participated in the embryonic development of G. przewalskii, and were involved in the cold response during development. The current study proposed a potential role of PPARs in the cold response in the embryonic development of G. przewalskii, which shed light on understanding cold adaptation in Tibetan highland fish.
Collapse
Affiliation(s)
- Xiaohuan Li
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, No. 23 Xinning Road, Xining, 810001, Qinghai, China
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Sijia Liu
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, No. 23 Xinning Road, Xining, 810001, Qinghai, China
| | - Delin Qi
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai, China
| | - Hongfang Qi
- Qinghai Provincial Key Laboratory of Breeding and Protection of Gymnocypris Przewalskii, Xining, Qinghai, China
| | - Yang Wang
- Qinghai Provincial Key Laboratory of Breeding and Protection of Gymnocypris Przewalskii, Xining, Qinghai, China
| | - Kai Zhao
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, No. 23 Xinning Road, Xining, 810001, Qinghai, China.
| | - Fei Tian
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, No. 23 Xinning Road, Xining, 810001, Qinghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
18
|
Gómez-Abellán V, Pérez-Oliva AB, Cabas I, Hermi F, Arizcun M, García-Moreno D, Sepulcre MP, Mulero V. Peroxisome proliferator-activated receptors alpha and beta mediate the anti-inflammatory effects of the cyclopentenone prostaglandin 15-deoxy-Δ 12,14-PGJ 2 in fish granulocytes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 136:104498. [PMID: 35948178 DOI: 10.1016/j.dci.2022.104498] [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: 06/27/2022] [Revised: 07/24/2022] [Accepted: 07/25/2022] [Indexed: 06/15/2023]
Abstract
Prostaglandins (PGs) are highly reactive small lipophilic molecules derived from polyunsaturated fatty acids of the cell membrane and play a key role in the resolution of inflammation processes. 15-deoxy-Δ12,14-PGJ2 (15dPGJ2) is a cyclopentenone PG (CyPG) of the J series with anti-inflammatory, anti-proliferative and pro-apoptotic effects. This CyPG can signal through: (i) the PGD2 receptor (DP2) and peroxisome proliferator-activated receptor γ (PPARγ) or (ii) by covalent binding to protein nucleophiles, such as, thiols groups of cysteine, lysine or histidine via a Michael addition reaction, modifying its structure and function. In this work we show that acidophilic granulocytes (AGs) of gilthead seabream (Sparus aurata L.), the functional equivalent to mammalian neutrophils, constitutively expressed ppara, pparb and pparg genes, the latter showing the highest expression and up-regulation when stimulated by bacterial DNA. In addition, we tested the ability of 15dPGJ2, and its biotinylated analog, as well as several PPARγ ligands, to modulate reactive oxygen species (ROS) and/or cytokines production during a Toll like receptor (TLR)-mediated granulocyte response. Thus, 15dPGJ2 was able to significantly decrease bacterial DNA-induced ROS production and transcript levels of pparg, interleukin-1β (il1b) and prostaglandin-endoperoxide synthase 2 (ptgs2). In contrast, its biotinylated analog was less potent and a higher dose was required to elicit the same effects on ROS production and cytokine expression. In addition, different PPARγ agonists were able to mimic the effects of 15dPGJ2. Conversely, the PPARγ antagonist T007097 abolished the effect of 15dPGJ2 on DNA bacterial-induced ROS production. Surprisingly, transactivation assays revealed that both 15dPGJ2 and its biotinylated analog signaled via Pparα and Pparβ, but not by Pparγ. These results were further confirmed by HPLC/MS analysis, where Pparβ was identified as an interactor of biotin-15dPGJ2 in naïve and DNA-stimulated leukocytes. Taken together, our data show that 15dPGJ2 acts both through Ppar activation and covalent binding to proteins in fish granulocytes and identify for the first time in vertebrates a role for Pparα and Pparβ in the resolution of inflammation mediated by 15dPGJ2.
Collapse
Affiliation(s)
- Victoria Gómez-Abellán
- Departamento de Biología Cellular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain
| | - Ana B Pérez-Oliva
- 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
| | - Isabel Cabas
- Departamento de Biología Cellular 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
| | - Fatma Hermi
- Unit of Immunology, Environmental Microbiology and Cancerously, Faculty of Sciences of Bizerte, Jarzouna, Bizerte, 7021, University of Carthage, Tunis, Tunisia
| | - Marta Arizcun
- Oceanographic Center of Murcia, Spanish Institute of Oceanography (IEO-CSIC), 30860, Murcia, 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
| | - María P Sepulcre
- Departamento de Biología Cellular 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.
| | - Victoriano Mulero
- Departamento de Biología Cellular 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
|
19
|
Kurchaba N, Charette JM, LeMoine CMR. Metabolic consequences of PGC-1α dysregulation in adult zebrafish muscle. Am J Physiol Regul Integr Comp Physiol 2022; 323:R319-R330. [PMID: 35670765 DOI: 10.1152/ajpregu.00188.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The peroxisome proliferator activated receptor gamma co-activator 1 alpha (PGC-1α) is central to the regulation of cellular and mitochondrial energy homeostasis in mammals, but its role in other vertebrates remains unclear. Indeed, previous work suggests extensive structural and functional divergence of PGC-1α in teleosts but this remains to be directly tested. Here, we describe the initial characterization of heterozygous PGC-1α mutant zebrafish lines created by CRISPR-Cas9 disruptions of an evolutionarily conserved regulatory region of the PGC-1α proximal promoter. Using qPCR, we confirmed the disruption of PGC-1α gene expression in striated muscle, leading to a simultaneous 4-fold increase in mixed skeletal muscle PGC-1α mRNA levels and an opposite 4-fold downregulation in cardiac muscle. In mixed skeletal muscle, most downstream effector genes were largely unaffected yet two mitochondrial lipid transporters, carnitine palmitoyltransferase 1 and 2, were strongly induced. Conversely, PGC-1α depression in cardiac muscle reduced the expression of several transcriptional regulators (estrogen related receptor alpha, nuclear respiratory factor 1 and PGC-1β) without altering metabolic gene expression. Using high resolution respirometry, we determined that white muscle exhibited increased lipid oxidative capacity with little difference in markers of mitochondrial abundance. Finally, using whole animal intermittent respirometry, we show that mutant fish exhibit a 2-fold higher basal metabolism than their wildtype counterparts. Altogether, this new model confirms a central but complex role for PGC-1α in mediating energy utilization in zebrafish and we propose its use as a valuable tool to explore the intricate regulatory pathways of energy homeostasis in a popular biomedical model.
Collapse
Affiliation(s)
| | - J Michael Charette
- Department of Chemistry, Brandon University, Brandon, MB, Canada.,Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB, Canada.,CancerCare Manitoba Research Institute, Winnipeg, MB, Canada
| | | |
Collapse
|
20
|
Buerger AN, Parente CE, Harris JP, Watts EG, Wormington AM, Bisesi JH. Impacts of diethylhexyl phthalate and overfeeding on physical fitness and lipid mobilization in Danio rerio (zebrafish). CHEMOSPHERE 2022; 295:133703. [PMID: 35066078 DOI: 10.1016/j.chemosphere.2022.133703] [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: 06/30/2021] [Revised: 01/04/2022] [Accepted: 01/18/2022] [Indexed: 06/14/2023]
Abstract
As the prevalence of obesity has steadily increased on a global scale, research has shifted to explore potential contributors to this pandemic beyond overeating and lack of exercise. Environmental chemical contaminants, known as obesogens, alter metabolic processes and exacerbate the obese phenotype. Diethylhexyl phthalate (DEHP) is a common chemical plasticizer found in medical supplies, food packaging, and polyvinyl materials, and has been identified as a probable obesogen. This study investigated the hypothesis that co-exposure to DEHP and overfeeding would result in decreased lipid mobilization and physical fitness in Danio rerio (zebrafish). Four treatment groups were randomly assigned: Regular Fed (control, 10 mg/fish/day with 0 mg/kg DEHP), Overfed (20 mg/fish/day with 0 mg/kg DEHP), Regular Fed + DEHP (10 mg/fish/day with 3 mg/kg DEHP), Overfed + DEHP (20 mg/fish/day with 3 mg/kg DEHP). After 24 weeks, swim tunnel assays were conducted on half of the zebrafish from each treatment to measure critical swimming speeds (Ucrit); the other fish were euthanized without swimming. Body mass index (BMI) was measured, and tissues were collected for blood lipid characterization and gene expression analyses. Co-exposure to DEHP and overfeeding decreased swim performance as measured by Ucrit. While no differences in blood lipids were observed with DEHP exposure, differential expression of genes related to lipid metabolism and utilization in the gastrointestinal and liver tissue suggests alterations in metabolism and lipid packaging, which may impact utilization and ability to mobilize lipid reserves during physical activity following chronic exposures.
Collapse
Affiliation(s)
- Amanda N Buerger
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, USA
| | - Caitlyn E Parente
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, USA; Department of Chemistry, University of Florida, Gainesville, FL, USA
| | - Jason P Harris
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, USA; Department of Biology, University of Florida, Gainesville, FL, USA
| | - Emily G Watts
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, USA; Department of Chemistry, University of Florida, Gainesville, FL, USA
| | - Alexis M Wormington
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, USA
| | - Joseph H Bisesi
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
21
|
Zhong X, Gu J, Zhang S, Chen X, Zhang J, Miao J, Ding Z, Xu J, Cheng H. Dynamic transcriptome analysis of the muscles in high-fat diet-induced obese zebrafish (Danio rerio) under 5-HT treatment. Gene 2022; 819:146265. [PMID: 35121026 DOI: 10.1016/j.gene.2022.146265] [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: 11/16/2021] [Revised: 01/18/2022] [Accepted: 01/27/2022] [Indexed: 11/04/2022]
Abstract
Peripheral 5-hydroxytryptamine (5-HT, also called serotonin) is reportedly a potential therapeutic target in obesity-related metabolic diseases due to its regulatory role in energy homeostasis in mammals. However, information on the detailed effect of peripheral 5-HT on the energy metabolism in fishes, especially the lipid metabolism, and the underlying mechanism remains elusive. In this study, a diet-induced obesity model was developed in the zebrafish (Danio rerio), a prototypical animal model for metabolic disorders. The zebrafish were fed a high-fat diet for 8 weeks and were simultaneously injected with PBS, 0.1 mM and 10 mM 5-HT, intraperitoneally. The body weight was significantly lower in the zebrafish injected with 0.1 mM 5-HT (P < 0.05), however, there was no change in body length (P > 0.05) at the end of the 8-week treatment. The muscle tissues from the zebrafish treated with PBS and 5-HT were collected for transcriptomic analysis and the RNA-seq revealed 1134, 3713, and 2535 genes were screened out compared to the muscular DEGs among three groups. The enrichment analysis revealed DEGs to be significantly associated with multiple metabolic pathways, including ribosome, oxidative phosphorylation, proteasome, PPAR signaling pathway, and ferroptosis. Additionally, the qRT-PCR validated 12 DEGs out of which 10 genes exhibited consistent trends. Taken together, this data provided useful information on the transcriptional characteristics of the muscle tissue in the obese zebrafish exposed to 5-HT, offering important insights into the regulatory effect of peripheral 5-HT in teleosts, as well as novel approaches for preventing and treating obesity-related metabolic dysfunction.
Collapse
Affiliation(s)
- Xiangqi Zhong
- Jiangsu Key Laboratory of Marine Biotechnology/Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; Key Laboratory of Cultivation and High-value Utilization of Marine Organisms, Fisheries Research Institute of Fujian, Xiamen 361000, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
| | - Jiaze Gu
- Jiangsu Key Laboratory of Marine Biotechnology/Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| | - Siying Zhang
- Jiangsu Key Laboratory of Marine Biotechnology/Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xiangning Chen
- Jiangsu Key Laboratory of Marine Biotechnology/Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; Key Laboratory of Cultivation and High-value Utilization of Marine Organisms, Fisheries Research Institute of Fujian, Xiamen 361000, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China.
| | - Jingjing Zhang
- Jiangsu Key Laboratory of Marine Biotechnology/Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China
| | - Jintao Miao
- Jiangsu Key Laboratory of Marine Biotechnology/Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China
| | - Zhujin Ding
- Jiangsu Key Laboratory of Marine Biotechnology/Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
| | - Jianhe Xu
- Jiangsu Key Laboratory of Marine Biotechnology/Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
| | - Hanliang Cheng
- Jiangsu Key Laboratory of Marine Biotechnology/Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
| |
Collapse
|
22
|
Wang X, Ma Q, Chen L, Wu H, Chen LQ, Qiao F, Luo Y, Zhang ML, Du ZY. Peroxisome proliferator-activated receptor gamma is essential for stress adaptation by maintaining lipid homeostasis in female fish. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159162. [PMID: 35427795 DOI: 10.1016/j.bbalip.2022.159162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 03/28/2022] [Accepted: 04/07/2022] [Indexed: 01/04/2023]
Abstract
Reduction of lipid synthesis often causes free fatty acid (FFA) overload, resulting consequential oxidative stress and health damage. Environmental stresses also induce cellular oxidative stress in organisms. The functional peroxisome proliferator-activated receptor gamma (pparg) gene is essential for lipid synthesis and homeostatic lipid maintenance. However, the relationship between the pparg-mediated lipid synthesis and environmental stress adaptation awaits full elucidation. Here, we generated a pparg-knockout zebrafish model. The conversion of free fatty acids into triglycerides in the female pparg mutants was hampered by reduced esterification efficiency, thus induced lipotoxicity, as evidenced by high oxidative stress and damaged health in these mutants, which led to reduced resistance to cold, heat and ammonia nitrogen stresses. Activating pparg in the wild-type female fish via dietary supplementation with rosiglitazone (a pparg agonist), or reducing oxidative stress in the female pparg mutants via dietary supplementation with N-acetylcysteine (an antioxidant), or promoting mitochondrial fatty acid β-oxidation in the female pparg mutants via dietary supplementation with l-carnitine, resulted in significantly reduced cellular injury, and improved environmental stress resistance. Collectively, our findings reveal that the regulative function of pparg in FFA esterification is important in stress resistance in female fish, and highlight the tight correlation existing between lipotoxicity and environmental adaptation.
Collapse
Affiliation(s)
- Xue Wang
- LANEH, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Qiang Ma
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| | - Lingyun Chen
- LANEH, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Hongxia Wu
- LANEH, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Li-Qiao Chen
- LANEH, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Fang Qiao
- LANEH, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yuan Luo
- LANEH, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Mei-Ling Zhang
- LANEH, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Zhen-Yu Du
- LANEH, School of Life Sciences, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
23
|
Gence L, Fernezelian D, Bringart M, Veeren B, Christophe A, Brion F, Meilhac O, Bascands JL, Diotel N. Hypericum lanceolatum Lam. Medicinal Plant: Potential Toxicity and Therapeutic Effects Based on a Zebrafish Model. Front Pharmacol 2022; 13:832928. [PMID: 35359845 PMCID: PMC8963451 DOI: 10.3389/fphar.2022.832928] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/04/2022] [Indexed: 12/26/2022] Open
Abstract
Hypericum lanceolatum Lam. (H. lanceolatum) is a traditional medicinal plant from Reunion Island used for its pleiotropic effects mainly related to its antioxidant activity. The present work aimed to 1) determine the potential toxicity of the plant aqueous extract in vivo and 2) investigate its putative biological properties using several zebrafish models of oxidative stress, regeneration, estrogenicity, neurogenesis and metabolic disorders. First, we characterized the polyphenolic composition by liquid chromatography-tandem mass spectrometry (LC-MS/MS) and identified chlorogenic acid isomers, quercetin and kaempferol derivatives as the major compounds. We then evaluated for the first time the toxicity of an aqueous extract of H. lanceolatum and determined a maximum non-toxic concentration (MNTC) in zebrafish eleutheroembryos from 0 to 96 hpf following OECD (Organization for Economic Cooperation and Development) guidelines. This MNTC test was also determined on hatched eleutheroembryos after 2 days of treatment (from 3 to 5 dpf). In our study, the anti-estrogenic effects of H. lanceolatum are supported by the data from the EASZY assay. In a tail amputation model, we showed that H. lanceolatum at its MNTC displays antioxidant properties, favors immune cell recruitment and tissue regeneration. Our results also highlighted its beneficial effects in metabolic disorders. Indeed, H. lanceolatum efficiently reduces lipid accumulation and body mass index in overfed larva- and adult-models, respectively. In addition, we show that H. lanceolatum did not improve fasting blood glucose levels in a hyperglycemic zebrafish model but surprisingly inhibited neurogenesis impairment observed in diabetic conditions. In conclusion, our study highlights the antioxidant, pro-regenerative, anti-lipid accumulation and pro-neurogenic effects of H. lanceolatum in vivo and supports the use of this traditional medicinal plant as a potential alternative in the prevention and/or treatment of metabolic disorders.
Collapse
Affiliation(s)
- Laura Gence
- Université de La Réunion, INSERM, Diabéte athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
| | - Danielle Fernezelian
- Université de La Réunion, INSERM, Diabéte athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
| | - Matthieu Bringart
- Université de La Réunion, INSERM, Diabéte athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
| | - Bryan Veeren
- Université de La Réunion, INSERM, Diabéte athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
| | - Armelle Christophe
- Unité D’Écotoxicologie des Substances et des Milieux (ESMI), Institut National de L’Environnement Industriel et des Risques (INERIS), Verneuil-en-Halatte, France
| | - François Brion
- Unité D’Écotoxicologie des Substances et des Milieux (ESMI), Institut National de L’Environnement Industriel et des Risques (INERIS), Verneuil-en-Halatte, France
| | - Olivier Meilhac
- Université de La Réunion, INSERM, Diabéte athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
- CHU de La Réunion, Saint-Denis, France
| | - Jean-Loup Bascands
- Université de La Réunion, INSERM, Diabéte athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
- *Correspondence: Jean-Loup Bascands, ; Nicolas Diotel,
| | - Nicolas Diotel
- Université de La Réunion, INSERM, Diabéte athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
- *Correspondence: Jean-Loup Bascands, ; Nicolas Diotel,
| |
Collapse
|
24
|
von Hellfeld R, Pannetier P, Braunbeck T. Specificity of time- and dose-dependent morphological endpoints in the fish embryo acute toxicity (FET) test for substances with diverse modes of action: the search for a "fingerprint". ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:16176-16192. [PMID: 34643865 PMCID: PMC8827326 DOI: 10.1007/s11356-021-16354-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 09/01/2021] [Indexed: 06/13/2023]
Abstract
The fish embryo acute toxicity (FET) test with the zebrafish (Danio rerio) embryo according to OECD TG 236 was originally developed as an alternative test method for acute fish toxicity testing according to, e.g., OECD TG 203. Given the versatility of the protocol, however, the FET test has found application beyond acute toxicity testing as a common tool in environmental hazard and risk assessment. Whereas the standard OECD guideline is restricted to four core endpoints (coagulation as well as lack of somite formation, heartbeat, and tail detachment) for simple, rapid assessment of acute toxicity, further endpoints can easily be integrated into the FET test protocol. This has led to the hypothesis that an extended FET test might allow for the identification of different classes of toxicants via a "fingerprint" of morphological observations. To test this hypothesis, the present study investigated a set of 18 compounds with highly diverse modes of action with respect to acute and sublethal endpoints. Especially at higher concentrations, most observations proved toxicant-unspecific. With decreasing concentrations, however, observations declined in number, but gained in specificity. Specific observations may at best be made at test concentrations ≤ EC10. The existence of a "fingerprint" based on morphological observations in the FET is, therefore, highly unlikely in the range of acute toxicity, but cannot be excluded for experiments at sublethal concentrations.
Collapse
Affiliation(s)
- Rebecca von Hellfeld
- Center for Organismal Studies, Aquatic Ecology and Toxicology Section, University of Heidelberg, Im Neuenheimer Feld 504, 69120, Heidelberg, Germany.
- University of Aberdeen, Institute of Biological and Environmental Science, 23 St Machar Drive, AB24 3UU, Aberdeen, UK.
| | - Pauline Pannetier
- Center for Organismal Studies, Aquatic Ecology and Toxicology Section, University of Heidelberg, Im Neuenheimer Feld 504, 69120, Heidelberg, Germany
| | - Thomas Braunbeck
- Center for Organismal Studies, Aquatic Ecology and Toxicology Section, University of Heidelberg, Im Neuenheimer Feld 504, 69120, Heidelberg, Germany.
| |
Collapse
|
25
|
Kamoshita M, Kumar R, Anteghini M, Kunze M, Islinger M, Martins dos Santos V, Schrader M. Insights Into the Peroxisomal Protein Inventory of Zebrafish. Front Physiol 2022; 13:822509. [PMID: 35295584 PMCID: PMC8919083 DOI: 10.3389/fphys.2022.822509] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/07/2022] [Indexed: 12/19/2022] Open
Abstract
Peroxisomes are ubiquitous, oxidative subcellular organelles with important functions in cellular lipid metabolism and redox homeostasis. Loss of peroxisomal functions causes severe disorders with developmental and neurological abnormalities. Zebrafish are emerging as an attractive vertebrate model to study peroxisomal disorders as well as cellular lipid metabolism. Here, we combined bioinformatics analyses with molecular cell biology and reveal the first comprehensive inventory of Danio rerio peroxisomal proteins, which we systematically compared with those of human peroxisomes. Through bioinformatics analysis of all PTS1-carrying proteins, we demonstrate that D. rerio lacks two well-known mammalian peroxisomal proteins (BAAT and ZADH2/PTGR3), but possesses a putative peroxisomal malate synthase (Mlsl) and verified differences in the presence of purine degrading enzymes. Furthermore, we revealed novel candidate peroxisomal proteins in D. rerio, whose function and localisation is discussed. Our findings confirm the suitability of zebrafish as a vertebrate model for peroxisome research and open possibilities for the study of novel peroxisomal candidate proteins in zebrafish and humans.
Collapse
Affiliation(s)
- Maki Kamoshita
- College of Life and Environmental Sciences, Biosciences, University of Exeter, Exeter, United Kingdom
| | - Rechal Kumar
- College of Life and Environmental Sciences, Biosciences, University of Exeter, Exeter, United Kingdom
| | - Marco Anteghini
- LifeGlimmer GmbH, Berlin, Germany
- Systems and Synthetic Biology, Wageningen University & Research, Wageningen, Netherlands
| | - Markus Kunze
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Markus Islinger
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Vítor Martins dos Santos
- LifeGlimmer GmbH, Berlin, Germany
- Systems and Synthetic Biology, Wageningen University & Research, Wageningen, Netherlands
| | - Michael Schrader
- College of Life and Environmental Sciences, Biosciences, University of Exeter, Exeter, United Kingdom
- *Correspondence: Michael Schrader,
| |
Collapse
|
26
|
Xu M, Legradi J, Leonards P. Using comprehensive lipid profiling to study effects of PFHxS during different stages of early zebrafish development. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 808:151739. [PMID: 34848268 DOI: 10.1016/j.scitotenv.2021.151739] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 11/05/2021] [Accepted: 11/13/2021] [Indexed: 05/26/2023]
Abstract
PFHxS (Perfluorohexane sulfonic acid) is one of the short-chain perfluoroalkyl substances (PFASs) which are widely used in many industrial and consumer applications. However, limited information is available on the molecular mechanism of PFHxS toxicity (e.g. lipid metabolism). This study provides in-depth information on the lipid regulation of zebrafish embryos with and without PFHxS exposure. Lipid changes throughout zebrafish development (4 to 120 h post fertilization (hpf)) were closely associated with lipid species and lipid composition (fatty acyl chains). A comprehensive lipid analysis of four different PFHxS exposures (0, 0.3, 1, 3, and 10 μM) at different zebrafish developmental stages (24, 48, 72, and 120 hpf) was performed. Data on exposure concentration, lipids, and developmental stage showed that all PFHxS concentrations dysregulated the lipid metabolism and these were developmental-dependent. The pattern of significantly changed lipids revealed that PFHxS caused effects related to oxidative stress, inflammation, and impaired fatty acid β-oxidation. Oxidative stress and inflammation caused the remodeling of glycerophospholipid (phosphatidylcholine (PC) and phosphatidylethanolamine (PE)), with increased incorporation of omega-3 PUFA and a decreased incorporation of omega-6 PUFA.
Collapse
Affiliation(s)
- Mengmeng Xu
- Department of Environment and Health, Vrije Universiteit, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands.
| | - Jessica Legradi
- Department of Environment and Health, Vrije Universiteit, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| | - Pim Leonards
- Department of Environment and Health, Vrije Universiteit, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| |
Collapse
|
27
|
Identification of Scd5 as a functional regulator of visceral fat deposition and distribution. iScience 2022; 25:103916. [PMID: 35252813 PMCID: PMC8889148 DOI: 10.1016/j.isci.2022.103916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/24/2021] [Accepted: 02/09/2022] [Indexed: 11/20/2022] Open
Abstract
Ectopic deposition of visceral adipose tissue (VAT) in abdomen is usually accompanied with systematic chaos of energy metabolism, a higher risk of cardiovascular diseases and type II diabetes. Here, we identified a previously unexplored gene Scd5 as a master regulator of fat distribution, which alone plays a significant role in determining the VAT accumulation. Firstly, zebrafish scd5 had the highest homology with human SCD5 compared to other SCDs in mouse and rat. We then observed that scd5-homozygous mutant zebrafish displayed a puffy, short and rounded apple-shaped figure. Whole-mount micro-CT scan showed that excessive VAT deposition and short spine are responsible for the abnormal body ratio. And the supplementation of ω3-polyunsaturated fatty acid (ω3-PUFA) in dietary significantly decreased VAT accumulation in scd5−/− zebrafish. Lastly, transcriptional analyses revealed that the Wnt, PPAR, C/EBP, and fat synthesis signaling pathways are significantly affected in the VAT of scd5−/− mutant and restored by ω3-PUFA. Zebrafish scd5 is a better match of homolog to human SCD5 scd5 deficiency induced significant VAT depositions in zebrafish Supplementation of ω3-PUFA significantly reduced the VAT deposition in scd5 mutants
Collapse
|
28
|
The Preservation of PPARγ Genome Duplicates in Some Teleost Lineages: Insights into Lipid Metabolism and Xenobiotic Exploitation. Genes (Basel) 2022; 13:genes13010107. [PMID: 35052447 PMCID: PMC8774674 DOI: 10.3390/genes13010107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/28/2021] [Accepted: 12/31/2021] [Indexed: 01/10/2023] Open
Abstract
Three peroxisome proliferator-activated receptor paralogues (PPARα, -β and -γ) are currently recognized in vertebrate genomes. PPARγ is known to modulate nutrition, adipogenesis and immunity in vertebrates. Natural ligands of PPARγ have been proposed; however, the receptor also binds synthetic ligands such as endocrine disruptors. Two paralogues of PPARα and PPARβ have been documented in teleost species, a consequence of the 3R WGD. Recently, two PPARγ paralogue genes were also identified in Astyanax mexicanus. We aimed to determine whether the presence of two PPARγ paralogues is prevalent in other teleost genomes, through genomic and phylogenetic analysis. Our results showed that besides Characiformes, two PPARγ paralogous genes were also identified in other teleost taxa, coinciding with the teleost-specific, whole-genome duplication and with the retention of both genes prior to the separation of the Clupeocephala. To functionally characterize these genes, we used the European sardine (Sardina pilchardus) as a model. PPARγA and PPARγB display a different tissue distribution, despite the similarity of their functional profiles: they are unresponsive to tested fatty acids and other human PPARγ ligands yet yield a transcriptional response in the presence of tributyltin (TBT). This observation puts forward the relevance of comparative analysis to decipher alternative binding architectures and broadens the disruptive potential of man-made chemicals for aquatic species.
Collapse
|
29
|
Garoche C, Boulahtouf A, Grimaldi M, Chiavarina B, Toporova L, den Broeder MJ, Legler J, Bourguet W, Balaguer P. Interspecies Differences in Activation of Peroxisome Proliferator-Activated Receptor γ by Pharmaceutical and Environmental Chemicals. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2021; 55:16489-16501. [PMID: 34843233 DOI: 10.1021/acs.est.1c04318] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Endocrine disrupting chemicals (EDCs) are able to deregulate the hormone system, notably through interactions with nuclear receptors (NRs). The mechanisms of action and biological effects of many EDCs have mainly been tested on human and mouse but other species such as zebrafish and xenopus are increasingly used as a model to study the effects of EDCs. Among NRs, peroxisome proliferator-activated receptor γ (PPARγ) is a main target of EDCs, for which most experimental data have been obtained from human and mouse models. To assess interspecies differences, we tested known human PPARγ ligands on reporter cell lines expressing either human, mouse, zebrafish, or xenopus PPARγ. Using these cell lines, we were able to highlight major interspecies differences. Known hPPARγ pharmaceutical ligands modulated hPPARγ and mPPARγ activities in a similar manner, while xPPARγ was less responsive and zfPPARγ was not modulated at all by these compounds. On the contrary, human liver X receptor (hLXR) ligands GW 3965 and WAY-252623 were only active on zfPPARγ. Among environmental compounds, several molecules activated the PPARγ of the four species similarly, e.g., phthalates (MEHP), perfluorinated compounds (PFOA, PFOS), and halogenated derivatives of BPA (TBBPA, TCBPA), but some of them like diclofenac and the organophosphorus compounds tri-o-tolyl phosphate and triphenyl phosphate were most active on zfPPARγ. This study confirms or shows for the first time the h, m, x, and zfPPARγ activities of several chemicals and demonstrates the importance of the use of species-specific models to study endocrine and metabolism disruption by environmental chemicals.
Collapse
Affiliation(s)
- Clémentine Garoche
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université Montpellier, Institut régional du Cancer de Montpellier (ICM), 34290 Montpellier, France
| | - Abdelhay Boulahtouf
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université Montpellier, Institut régional du Cancer de Montpellier (ICM), 34290 Montpellier, France
| | - Marina Grimaldi
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université Montpellier, Institut régional du Cancer de Montpellier (ICM), 34290 Montpellier, France
| | - Barbara Chiavarina
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université Montpellier, Institut régional du Cancer de Montpellier (ICM), 34290 Montpellier, France
| | - Lucia Toporova
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université Montpellier, Institut régional du Cancer de Montpellier (ICM), 34290 Montpellier, France
| | - Marjo J den Broeder
- Institute for Risk Assessment Sciences, Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, The Netherlands
| | - Juliette Legler
- Institute for Risk Assessment Sciences, Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, The Netherlands
| | - William Bourguet
- Centre de Biologie Structurale (CBS), Inserm U1053, CNRS, Université Montpellier, 34290 Montpellier, France
| | - Patrick Balaguer
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université Montpellier, Institut régional du Cancer de Montpellier (ICM), 34290 Montpellier, France
| |
Collapse
|
30
|
Obesity Animal Models for Acupuncture and Related Therapy Research Studies. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6663397. [PMID: 34630614 PMCID: PMC8497105 DOI: 10.1155/2021/6663397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 09/02/2021] [Indexed: 11/17/2022]
Abstract
Obesity and related diseases are considered as pandemic representing a worldwide threat for health. Animal models are critical to validate the effects and understand the mechanisms related to classical or innovative preventive and therapeutic strategies. It is, therefore, important to identify the best animal models for translational research, using different evaluation criteria such as the face, construct, and predictive validity. Because the pharmacological treatments and surgical interventions currently used for treating obesity often present many undesirable side effects, relatively high relapse probabilities, acupuncture, electroacupuncture (EA), and related therapies have gained more popularity and attention. Many kinds of experimental animal models have been used for obesity research studies, but in the context of acupuncture, most of the studies were performed in rodent obesity models. Though, are these obesity rodent models really the best for acupuncture or related therapies research studies? In this study, we review different obesity animal models that have been used over the past 10 years for acupuncture and EA research studies. We present their respective advantages, disadvantages, and specific constraints. With the development of research on acupuncture and EA and the increasing interest regarding these approaches, proper animal models are critical for preclinical studies aiming at developing future clinical trials in the human. The aim of the present study is to provide researchers with information and guidance related to the preclinical models that are currently available to investigate the outcomes of acupuncture and related therapies.
Collapse
|
31
|
Experimental animal models for diabetes and its related complications-a review. Lab Anim Res 2021; 37:23. [PMID: 34429169 PMCID: PMC8385906 DOI: 10.1186/s42826-021-00101-4] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/13/2021] [Indexed: 12/16/2022] Open
Abstract
Diabetes mellitus, a very common and multifaceted metabolic disorder is considered as one of the fastest growing public health problems in the world. It is characterized by hyperglycemia, a condition with high glucose level in the blood plasma resulting from defects in insulin secretion or its action and in some cases both the impairment in secretion and also action of insulin coexist. Historically, animal models have played a critical role in exploring and describing malady pathophysiology and recognizable proof of targets and surveying new remedial specialists and in vivo medicines. In the present study, we reviewed the experimental models employed for diabetes and for its related complications. This paper reviews briefly the broad chemical induction of alloxan and streptozotocin and its mechanisms associated with type 1 and type 2 diabetes. Also we highlighted the different models in other species and other animals.
Collapse
|
32
|
Venezia O, Islam S, Cho C, Timme-Laragy AR, Sant KE. Modulation of PPAR signaling disrupts pancreas development in the zebrafish, Danio rerio. Toxicol Appl Pharmacol 2021; 426:115653. [PMID: 34302850 DOI: 10.1016/j.taap.2021.115653] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 11/15/2022]
Abstract
Peroxisome Proliferator Activated Receptors (PPARs) are transcription factors that regulate processes such as lipid and glucose metabolism. Synthetic PPAR ligands, designed as therapeutics for metabolic disease, provide a tool to assess the relationship between PPAR activity and pancreas development in vivo, an area that remains poorly characterized. Here, we aim to assess the effects of PPAR agonists and antagonists on gene expression, embryonic morphology and pancreas development in transgenic zebrafish embryos. To evaluate developmental perturbations, we assessed gross body and pancreas morphology at 4 days post fertilization (dpf) in response to developmental exposures with PPARα, PPARγ, and PPARβ/δ agonists and antagonists at 0, 0.01, 0.1, 1, and 10 μM concentrations. All ligand exposures, with the exception of the PPARα agonist, resulted in significantly altered fish length and yolk sac area. PPARγ agonist and antagonist had higher incidence of darkened yolk sac and craniofacial deformities, whereas PPARα antagonist had higher incidence of pericardial edema and death. Significantly reduced endocrine pancreas area was observed in both PPARγ ligands and PPARα agonist exposed embryos, some of which also exhibited aberrant endocrine pancreas morphology. Both PPARβ/δ ligands caused reduced exocrine pancreas length and novel aberrant phenotype, and disrupted gene expression of pancreatic targets pdx1, gcga, and try. Lipid staining was performed at 8 dpf and revealed altered lipid accumulation consistent with isoform function. These data indicate chronic exposure to synthetic ligands may induce morphological and pancreatic defects in zebrafish embryos.
Collapse
Affiliation(s)
- Olivia Venezia
- Department of Environmental Health Sciences, University of Massachusetts-Amherst, Amherst, MA, United States of America
| | - Sadia Islam
- Department of Environmental Health Sciences, University of Massachusetts-Amherst, Amherst, MA, United States of America
| | - Christine Cho
- School of Public Health, San Diego State University, San Diego, CA, United States of America
| | - Alicia R Timme-Laragy
- Department of Environmental Health Sciences, University of Massachusetts-Amherst, Amherst, MA, United States of America
| | - Karilyn E Sant
- Department of Environmental Health Sciences, University of Massachusetts-Amherst, Amherst, MA, United States of America; School of Public Health, San Diego State University, San Diego, CA, United States of America.
| |
Collapse
|
33
|
Pandelides Z, Aluru N, Thornton C, Watts HE, Willett KL. Transcriptomic Changes and the Roles of Cannabinoid Receptors and PPARγ in Developmental Toxicities Following Exposure to Δ9-Tetrahydrocannabinol and Cannabidiol. Toxicol Sci 2021; 182:44-59. [PMID: 33892503 PMCID: PMC8285010 DOI: 10.1093/toxsci/kfab046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Human consumption of cannabinoid-containing products during early life or pregnancy is rising. However, information about the molecular mechanisms involved in early life stage Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD) toxicities is critically lacking. Here, larval zebrafish (Danio rerio) were used to measure THC- and CBD-mediated changes on transcriptome and the roles of cannabinoid receptors (Cnr) 1 and 2 and peroxisome proliferator activator receptor γ (PPARγ) in developmental toxicities. Transcriptomic profiling of 96-h postfertilization (hpf) cnr+/+ embryos exposed (6 - 96 hpf) to 4 μM THC or 0.5 μM CBD showed differential expression of 904 and 1095 genes for THC and CBD, respectively, with 360 in common. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways enriched in the THC and CBD datasets included those related to drug, retinol, and steroid metabolism and PPAR signaling. The THC exposure caused increased mortality and deformities (pericardial and yolk sac edemas, reduction in length) in cnr1-/- and cnr2-/- fish compared with cnr+/+ suggesting Cnr receptors are involved in protective pathways. Conversely, the cnr1-/- larvae were more resistant to CBD-induced malformations, mortality, and behavioral alteration implicating Cnr1 in CBD-mediated toxicity. Behavior (decreased distance travelled) was the most sensitive endpoint to THC and CBD exposure. Coexposure to the PPARγ inhibitor GW9662 and CBD in cnr+/+ and cnr2-/- strains caused more adverse outcomes compared with CBD alone, but not in the cnr1-/- fish, suggesting that PPARγ plays a role in CBD metabolism downstream of Cnr1. Collectively, PPARγ, Cnr1, and Cnr2 play important roles in the developmental toxicity of cannabinoids with Cnr1 being the most critical.
Collapse
Affiliation(s)
- Zacharias Pandelides
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi 38677, USA
| | - Neelakanteswar Aluru
- Biology Department, Woods Hole Oceanographic Institution and Woods Hole Center for Oceans and Human Health, Woods Hole, Massachusetts 02543, USA
| | - Cammi Thornton
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi 38677, USA
| | - Haley E Watts
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi 38677, USA
| | - Kristine L Willett
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi 38677, USA
| |
Collapse
|
34
|
Zebrafish and Flavonoids: Adjuvants against Obesity. Molecules 2021; 26:molecules26103014. [PMID: 34069388 PMCID: PMC8158719 DOI: 10.3390/molecules26103014] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 12/16/2022] Open
Abstract
Obesity is a pathological condition, defined as an excessive accumulation of fat, primarily caused by an energy imbalance. The storage of excess energy in the form of triglycerides within the adipocyte leads to lipotoxicity and promotes the phenotypic switch in the M1/M2 macrophage. These changes induce the development of a chronic state of low-grade inflammation, subsequently generating obesity-related complications, commonly known as metabolic syndromes. Over the past decade, obesity has been studied in many animal models. However, due to its competitive aspects and unique characteristics, the use of zebrafish has begun to gain traction in experimental obesity research. To counteract obesity and its related comorbidities, several natural substances have been studied. One of those natural substances reported to have substantial biological effects on obesity are flavonoids. This review summarizes the results of studies that examined the effects of flavonoids on obesity and related diseases and the emergence of zebrafish as a model of diet-induced obesity.
Collapse
|
35
|
Generation of a Novel Transgenic Zebrafish for Studying Adipocyte Development and Metabolic Control. Int J Mol Sci 2021; 22:ijms22083994. [PMID: 33924375 PMCID: PMC8069137 DOI: 10.3390/ijms22083994] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/07/2021] [Accepted: 04/10/2021] [Indexed: 01/10/2023] Open
Abstract
Zebrafish have become a popular animal model for studying various biological processes and human diseases. The metabolic pathways and players conserved among zebrafish and mammals facilitate the use of zebrafish to understand the pathological mechanisms underlying various metabolic disorders in humans. Adipocytes play an important role in metabolic homeostasis, and zebrafish adipocytes have been characterized. However, a versatile and reliable zebrafish model for long-term monitoring of adipose tissues has not been reported. In this study, we generated stable transgenic zebrafish expressing enhanced green fluorescent protein (EGFP) in adipocytes. The transgenic zebrafish harbored adipose tissues that could be detected using GFP fluorescence and the morphology of single adipocyte could be investigated in vivo. In addition, we demonstrated the applicability of this model to the long-term in vivo imaging of adipose tissue development and regulation based on nutrition. The transgenic zebrafish established in this study may serve as an excellent tool to advance the characterization of white adipose tissue in zebrafish, thereby aiding the development of therapeutic interventions to treat metabolic diseases in humans.
Collapse
|
36
|
Jersin RÅ, Tallapragada DSP, Madsen A, Skartveit L, Fjære E, McCann A, Lawrence-Archer L, Willems A, Bjune JI, Bjune MS, Våge V, Nielsen HJ, Thorsen HL, Nedrebø BG, Busch C, Steen VM, Blüher M, Jacobson P, Svensson PA, Fernø J, Rydén M, Arner P, Nygård O, Claussnitzer M, Ellingsen S, Madsen L, Sagen JV, Mellgren G, Dankel SN. Role of the Neutral Amino Acid Transporter SLC7A10 in Adipocyte Lipid Storage, Obesity, and Insulin Resistance. Diabetes 2021; 70:680-695. [PMID: 33408126 DOI: 10.2337/db20-0096] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 12/14/2020] [Indexed: 11/13/2022]
Abstract
Elucidation of mechanisms that govern lipid storage, oxidative stress, and insulin resistance may lead to improved therapeutic options for type 2 diabetes and other obesity-related diseases. Here, we find that adipose expression of the small neutral amino acid transporter SLC7A10, also known as alanine-serine-cysteine transporter-1 (ASC-1), shows strong inverse correlates with visceral adiposity, insulin resistance, and adipocyte hypertrophy across multiple cohorts. Concordantly, loss of Slc7a10 function in zebrafish in vivo accelerates diet-induced body weight gain and adipocyte enlargement. Mechanistically, SLC7A10 inhibition in human and murine adipocytes decreases adipocyte serine uptake and total glutathione levels and promotes reactive oxygen species (ROS) generation. Conversely, SLC7A10 overexpression decreases ROS generation and increases mitochondrial respiratory capacity. RNA sequencing revealed consistent changes in gene expression between human adipocytes and zebrafish visceral adipose tissue following loss of SLC7A10, e.g., upregulation of SCD (lipid storage) and downregulation of CPT1A (lipid oxidation). Interestingly, ROS scavenger reduced lipid accumulation and attenuated the lipid-storing effect of SLC7A10 inhibition. These data uncover adipocyte SLC7A10 as a novel important regulator of adipocyte resilience to nutrient and oxidative stress, in part by enhancing glutathione levels and mitochondrial respiration, conducive to decreased ROS generation, lipid accumulation, adipocyte hypertrophy, insulin resistance, and type 2 diabetes.
Collapse
Affiliation(s)
- Regine Å Jersin
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Divya Sri Priyanka Tallapragada
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - André Madsen
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Linn Skartveit
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Even Fjære
- Institute of Marine Research, Bergen, Norway
| | | | - Laurence Lawrence-Archer
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Aron Willems
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Jan-Inge Bjune
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Mona S Bjune
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Villy Våge
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
- Center of Health Research, Førde Hospital Trust, Førde, Norway
| | | | | | - Bjørn Gunnar Nedrebø
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haugesund Hospital, Haugesund, Norway
| | | | - Vidar M Steen
- NORMENT, K.G. Jebsen Center for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr. E. Martens Research Group for Biological Psychiatry, Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Matthias Blüher
- Clinic for Endocrinology and Nephrology, Medical Research Center, Leipzig, Germany
| | - Peter Jacobson
- Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Per-Arne Svensson
- Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Johan Fernø
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Peter Arner
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Ottar Nygård
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Melina Claussnitzer
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Ståle Ellingsen
- Institute of Marine Research, Bergen, Norway
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Lise Madsen
- Institute of Marine Research, Bergen, Norway
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Jørn V Sagen
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
- Bergen Stem Cell Consortium, Haukeland University Hospital, Bergen, Norway
| | - Gunnar Mellgren
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Simon N Dankel
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
37
|
Brandts I, Barría C, Martins MA, Franco-Martínez L, Barreto A, Tvarijonaviciute A, Tort L, Oliveira M, Teles M. Waterborne exposure of gilthead seabream (Sparus aurata) to polymethylmethacrylate nanoplastics causes effects at cellular and molecular levels. JOURNAL OF HAZARDOUS MATERIALS 2021; 403:123590. [PMID: 32795822 DOI: 10.1016/j.jhazmat.2020.123590] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 07/22/2020] [Accepted: 07/26/2020] [Indexed: 06/11/2023]
Abstract
This study evaluated the effect of a short-term exposure to 45 nm polymethylmethacrylate nanoplastics (PMMA-NPs) on the gilthead seabream (Sparus aurata), by assessing biomarkers at different levels of biological organization in liver and plasma. Fish were exposed via water to PMMA-NPs (0, 0.001, 0.01, 0.1, 1 and 10 mg L-1) and sampled after 24 and 96 h. Results showed a general up-regulation of mRNA levels of key genes associated with lipid metabolism (e.g. apolipoprotein A1 and retinoid X receptor). Together with the modulation of the lipid pathway genes we also found a global increase in cholesterol and triglycerides in plasma. Antioxidant-related genes (e.g. glutathione peroxidase 1) were also up-regulated after 24 h of exposure, but their expression levels returned to control afterwards. Total antioxidant capacity (TAC) was increased throughout the experiment, however at 96 h the antioxidant capacity became less efficient, reflected by an increase in the total oxidative status (TOS). Concomitantly, we found an increase in the erythrocytic nuclear abnormalities (ENAs) throughout the trial. Altogether, PMMA-NPs activated the organism's antioxidant defenses and induced alterations in lipid metabolism pathways and genotoxicity in the blood cells of gilthead seabream.
Collapse
Affiliation(s)
- I Brandts
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - C Barría
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - M A Martins
- Department of Physics & CICECO, University of Aveiro, 3810-193 Aveiro, Portugal
| | - L Franco-Martínez
- Interdisciplinary Laboratory of Clinical Analysis INTERLAB-UMU, Regional Campus of International Excellence Mare Nostrum, University of Murcia, Espinardo, Murcia 30100, Spain
| | - A Barreto
- Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal
| | - A Tvarijonaviciute
- Interdisciplinary Laboratory of Clinical Analysis INTERLAB-UMU, Regional Campus of International Excellence Mare Nostrum, University of Murcia, Espinardo, Murcia 30100, Spain
| | - L Tort
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - M Oliveira
- Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal
| | - M Teles
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain.
| |
Collapse
|
38
|
Caioni G, Viscido A, d’Angelo M, Panella G, Castelli V, Merola C, Frieri G, Latella G, Cimini A, Benedetti E. Inflammatory Bowel Disease: New Insights into the Interplay between Environmental Factors and PPARγ. Int J Mol Sci 2021; 22:985. [PMID: 33498177 PMCID: PMC7863964 DOI: 10.3390/ijms22030985] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/14/2021] [Accepted: 01/18/2021] [Indexed: 12/19/2022] Open
Abstract
The pathophysiological processes of inflammatory bowel diseases (IBDs), i.e., Crohn's disease (CD) and ulcerative colitis (UC), are still not completely understood. The exact etiology remains unknown, but it is well established that the pathogenesis of the inflammatory lesions is due to a dysregulation of the gut immune system resulting in over-production of pro-inflammatory cytokines. Increasing evidence underlines the involvement of both environmental and genetic factors. Regarding the environment, the microbiota seems to play a crucial role. Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors that exert pleiotropic effects on glucose homeostasis, lipid metabolism, inflammatory/immune processes, cell proliferation, and fibrosis. Furthermore, PPARs modulate interactions with several environmental factors, including microbiota. A significantly impaired PPARγ expression was observed in UC patients' colonic epithelial cells, suggesting that the disruption of PPARγ signaling may represent a critical step of the IBD pathogenesis. This paper will focus on the role of PPARγ in the interaction between environmental factors and IBD, and it will analyze the most suitable in vitro and in vivo models available to better study these relationships.
Collapse
Affiliation(s)
- Giulia Caioni
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.V.); (M.d.); (G.P.); (V.C.); (G.F.); (G.L.); (A.C.)
| | - Angelo Viscido
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.V.); (M.d.); (G.P.); (V.C.); (G.F.); (G.L.); (A.C.)
| | - Michele d’Angelo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.V.); (M.d.); (G.P.); (V.C.); (G.F.); (G.L.); (A.C.)
| | - Gloria Panella
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.V.); (M.d.); (G.P.); (V.C.); (G.F.); (G.L.); (A.C.)
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy;
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.V.); (M.d.); (G.P.); (V.C.); (G.F.); (G.L.); (A.C.)
| | - Carmine Merola
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy;
| | - Giuseppe Frieri
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.V.); (M.d.); (G.P.); (V.C.); (G.F.); (G.L.); (A.C.)
| | - Giovanni Latella
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.V.); (M.d.); (G.P.); (V.C.); (G.F.); (G.L.); (A.C.)
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.V.); (M.d.); (G.P.); (V.C.); (G.F.); (G.L.); (A.C.)
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, PA 19122, USA
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.V.); (M.d.); (G.P.); (V.C.); (G.F.); (G.L.); (A.C.)
| |
Collapse
|
39
|
Faheem M, Bhandari RK. Detrimental Effects of Bisphenol Compounds on Physiology and Reproduction in Fish: A Literature Review. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 81:103497. [PMID: 32950715 PMCID: PMC11491272 DOI: 10.1016/j.etap.2020.103497] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/24/2020] [Accepted: 09/11/2020] [Indexed: 06/11/2023]
Abstract
Bisphenol-A is one of the most studied endocrine-chemicals, which is widely used all over the world in plastic manufacture. Because of its extensive use, it has become one of the most abundant chemical environmental pollutants, especially in aquatic environments. BPA is known to affect fish reproduction via estrogen receptors but many studies advocate that BPA affects almost all aspects of fish physiology. The possible modes of action include genomic, as well as and non-genomic mechanisms, estrogen, androgen, and thyroid receptor-mediated effects. Due to the high detrimental effects of BPA, various analogs of BPA are being used as alternatives. Recent evidence suggests that the analogs of BPA have similar modes of action, with accompanying effects on fish physiology and reproduction. In this review, a detailed comparison of effects produced by BPA and analogs and their mode of action is discussed.
Collapse
|
40
|
Ka J, Jin SW. Zebrafish as an Emerging Model for Dyslipidemia and Associated Diseases. J Lipid Atheroscler 2020; 10:42-56. [PMID: 33537252 PMCID: PMC7838516 DOI: 10.12997/jla.2021.10.1.42] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 11/06/2020] [Accepted: 11/30/2020] [Indexed: 01/03/2023] Open
Abstract
Dyslipidemia related diseases such as hyperlipidemia and atherosclerosis are the leading cause of death in humans. While cellular and molecular basis on the pathophysiology of dyslipidemia has been extensively investigated over decades, we still lack comprehensive understanding on the etiology of dyslipidemia due to the complexity and the innate multimodality of the diseases. While mouse has been the model organism of choice to investigate the pathophysiology of human dyslipidemia, zebrafish, a small freshwater fish which has traditionally used to study vertebrate development, has recently emerged as an alternative model organism. In this review, we will provide comprehensive perspective on zebrafish as a model organism for human dyslipidemia; we will discuss the attributes of zebrafish as a model, and compare the lipid metabolism in zebrafish and humans. In addition, we will summarize current landscape of zebrafish-based dyslipidemia research.
Collapse
Affiliation(s)
- Jun Ka
- Cell Logistics Research Center and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Suk-Won Jin
- Cell Logistics Research Center and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea.,Yale Cardiovascular Research Center and Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
41
|
van der Ven LTM, Schoonen WG, Groot RM, den Ouden F, Heusinkveld HJ, Zwart EP, Hodemaekers HM, Rorije E, de Knecht J. The effects of aliphatic alcohols and related acid metabolites in zebrafish embryos - correlations with rat developmental toxicity and with effects in advanced life stages in fish. Toxicol Appl Pharmacol 2020; 407:115249. [PMID: 32979392 DOI: 10.1016/j.taap.2020.115249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/02/2020] [Accepted: 09/21/2020] [Indexed: 01/05/2023]
Abstract
The zebrafish embryo toxicity test (ZFET) is a simple medium-throughput test to inform about (sub)acute lethal effects in embryos. Enhanced analysis through morphological and teratological scoring, and through gene expression analysis, detects developmental effects and the underlying toxicological pathways. Altogether, the ZFET may inform about hazard of chemical exposure for embryonal development in humans, as well as for lethal effects in juvenile and adult fish. In this study, we compared the effects within a series of 12 aliphatic alcohols and related carboxylic acid derivatives (ethanol, acetic acid, 2-methoxyethanol, 2-methoxyacetic acid, 2-butoxyethanol, 2-butoxyacetic acid, 2-hydroxyacetic acid, 2-ethylhexan-1-ol, 2-ethylhexanoic acid, valproic acid, 2-aminoethanol, 2-(2-hydroxyethylamino)ethanol) in ZFET and early life stage (ELS, 28d) exposures, and compared ZFET results with existing results of rat developmental studies and LC50s in adult fish. High correlation scores were observed between compound potencies in ZFET with either ELS, LC50 in fish and developmental toxicity in rats, indicating similar potency ranking among the models. Compounds could be mapped to specific pathways in an adverse outcome pathway (AOP) network through morphological scoring and gene expression analysis in ZFET. Similarity of morphological effects and gene expression profiles in pairs of alcohols with their acid metabolites suggested metabolic activation of the parent alcohols, although with additional, metabolite-independent activity independent for ethanol and 2-ethylhexanol. Overall, phenotypical and gene expression analysis with these compounds indicates that the ZFET can potentially contribute to the AOP for developmental effects in rodents, and to predict toxicity of acute and chronic exposure in advanced life stages in fish.
Collapse
Affiliation(s)
- Leo T M van der Ven
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands.
| | - Willem G Schoonen
- Centre for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Renate M Groot
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Fatima den Ouden
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Harm J Heusinkveld
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Edwin P Zwart
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Hennie M Hodemaekers
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Emiel Rorije
- Centre for Safety of Substances and Products, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Joop de Knecht
- Centre for Safety of Substances and Products, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| |
Collapse
|
42
|
Dong YZ, Li L, Espe M, Lu KL, Rahimnejad S. Hydroxytyrosol Attenuates Hepatic Fat Accumulation via Activating Mitochondrial Biogenesis and Autophagy through the AMPK Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:9377-9386. [PMID: 32786840 DOI: 10.1021/acs.jafc.0c03310] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Two experiments were carried out to examine the impacts of hydroxytyrosol (HT) on lipid metabolism and mitochondrial function in Megalobrama amblycephala. Triplicate groups of fish were fed four test diets: (1) low-fat diet (LFD, 5% fat), (2) high-fat diet (HFD, 15% fat), (3) LFD + 100 mg/kg HT (LFD + HT), and (4) HFD + 100 mg/kg HT (HFD + HT) (in vivo). Hepatocytes from the same batch were exposed to three media including L-15 medium (L15), oleic acid (OA) medium [L15 + 400 μM OA], and OA + HT medium [L15 + 400 μM OA + 10 μM HT] to explore the roles of HT in mitochondrial function (in vitro). Fish fed HFD had excessive fat deposition in the liver, and HT inclusion in the HFD decreased hepatic fat deposition. Transmission electron microscopy revealed that the HFD triggers loss of cristae and metrical density and hydropic changes in mitochondria and that HT supplementation attenuates the ultrastructural alterations of mitochondria. The in vitro test showed that HT decreases fat deposition in hepatocytes, suppresses the reactive oxygen species formation, and facilitates the expression of phospho-AMPK protein and the genes involved in mitochondria biogenesis (PGC-1, NRF-1, TFAM) and autophagy (PINK1, Mul1, Atg5). These findings suggest the lipid-lowering effect of HT mediated by activation of mitochondrial biogenesis and autophagy through the AMPK pathway.
Collapse
Affiliation(s)
- Yan-Zou Dong
- Key Laboratory for Feed Quality Testing and Safety, Fisheries College, Jimei University, Xiamen 361021, China
| | - Lei Li
- Key Laboratory for Feed Quality Testing and Safety, Fisheries College, Jimei University, Xiamen 361021, China
| | - Marit Espe
- Institute of Marine Research (IMR), Bergen NO-5817, Norway
| | - Kang-Le Lu
- Key Laboratory for Feed Quality Testing and Safety, Fisheries College, Jimei University, Xiamen 361021, China
| | - Samad Rahimnejad
- University of South Bohemia in Ceske Budejovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zatisi 728/ II, Vodnany 389 25, Czech Republic
| |
Collapse
|
43
|
Li L, Lv H, Jiang Z, Qiao F, Chen L, Zhang M, Du Z. Peroxisomal proliferator‐activated receptor α‐b deficiency induces the reprogramming of nutrient metabolism in zebrafish. J Physiol 2020; 598:4537-4553. [DOI: 10.1113/jp279814] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 07/10/2020] [Indexed: 12/17/2022] Open
Affiliation(s)
- Ling‐Yu Li
- LANEH School of Life Sciences East China Normal University Shanghai China
| | - Hong‐Bo Lv
- LANEH School of Life Sciences East China Normal University Shanghai China
| | - Zhe‐Yue Jiang
- LANEH School of Life Sciences East China Normal University Shanghai China
| | - Fang Qiao
- LANEH School of Life Sciences East China Normal University Shanghai China
| | - Li‐Qiao Chen
- LANEH School of Life Sciences East China Normal University Shanghai China
| | - Mei‐Ling Zhang
- LANEH School of Life Sciences East China Normal University Shanghai China
| | - Zhen‐Yu Du
- LANEH School of Life Sciences East China Normal University Shanghai China
| |
Collapse
|
44
|
Stanford BC, Clake DJ, Morris MR, Rogers SM. The power and limitations of gene expression pathway analyses toward predicting population response to environmental stressors. Evol Appl 2020; 13:1166-1182. [PMID: 32684953 PMCID: PMC7359838 DOI: 10.1111/eva.12935] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 12/16/2022] Open
Abstract
Rapid environmental changes impact the global distribution and abundance of species, highlighting the urgency to understand and predict how populations will respond. The analysis of differentially expressed genes has elucidated areas of the genome involved in adaptive divergence to past and present environmental change. Such studies however have been hampered by large numbers of differentially expressed genes and limited knowledge of how these genes work in conjunction with each other. Recent methods (broadly termed "pathway analyses") have emerged that aim to group genes that behave in a coordinated fashion to a factor of interest. These methods aid in functional annotation and uncovering biological pathways, thereby collapsing complex datasets into more manageable units, providing more nuanced understandings of both the organism-level effects of modified gene expression, and the targets of adaptive divergence. Here, we reanalyze a dataset that investigated temperature-induced changes in gene expression in marine-adapted and freshwater-adapted threespine stickleback (Gasterosteus aculeatus), using Weighted Gene Co-expression Network Analysis (WGCNA) with PANTHER Gene Ontology (GO)-Slim overrepresentation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. Six modules exhibited a conserved response and six a divergent response between marine and freshwater stickleback when acclimated to 7°C or 22°C. One divergent module showed freshwater-specific response to temperature, and the remaining divergent modules showed differences in height of reaction norms. PPARAa, a transcription factor that regulates fatty acid metabolism and has been implicated in adaptive divergence, was located in a module that had higher expression at 7°C and in freshwater stickleback. This updated methodology revealed patterns that were not found in the original publication. Although such methods hold promise toward predicting population response to environmental stressors, many limitations remain, particularly with regard to module expression representation, database resources, and cross-database integration.
Collapse
Affiliation(s)
| | - Danielle J. Clake
- Department of Biological SciencesUniversity of CalgaryCalgaryABCanada
| | | | - Sean M. Rogers
- Department of Biological SciencesUniversity of CalgaryCalgaryABCanada
- Bamfield Marine Sciences CentreBamfieldBCCanada
| |
Collapse
|
45
|
Williams MB, Watts SA. Current basis and future directions of zebrafish nutrigenomics. GENES AND NUTRITION 2019; 14:34. [PMID: 31890052 PMCID: PMC6935144 DOI: 10.1186/s12263-019-0658-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/16/2019] [Indexed: 12/19/2022]
Abstract
This review investigates the current state of nutrigenomics in the zebrafish animal models. The zebrafish animal model has been used extensively in the study of disease onset and progression and associated molecular changes. In this review, we provide a synopsis of nutrigenomics using the zebrafish animal model. Obesity and dyslipidemia studies describe the genomics of dietary-induced obesity in relation to high-fat/high-calorie diets. Inflammation and cardiovascular studies describe dietary effects on the expression of acute inflammatory markers and resulting chronic inflammatory issues including atherosclerosis. We also evaluated the genomic response to bioactive dietary compounds associated with metabolic disorders. Carbohydrate metabolism and β-cell function studies describe the impacts of high-carbohydrate dietary challenges on nutritional programming. We also report tumorigenesis in relation to dietary carcinogen exposure studies that can result in permanent genomic changes. Vitamin and mineral deficiency studies demonstrate transgenerational genomic impacts of micronutrients in the diet and temporal expression changes. Circadian rhythm studies describe the relation between metabolism and natural temporal cycles of gene expression that impacts health. Bone formation studies describe the role of dietary composition that influences bone reabsorption regulation. Finally, this review provides future directions in the use of the zebrafish model for nutrigenomic and nutrigenetic research.
Collapse
Affiliation(s)
- Michael B Williams
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Stephen A Watts
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| |
Collapse
|
46
|
Jiang YP, Tang YL, Wang SS, Wu JS, Zhang M, Pang X, Wu JB, Chen Y, Tang YJ, Liang XH. PRRX1-induced epithelial-to-mesenchymal transition in salivary adenoid cystic carcinoma activates the metabolic reprogramming of free fatty acids to promote invasion and metastasis. Cell Prolif 2019; 53:e12705. [PMID: 31657086 PMCID: PMC6985691 DOI: 10.1111/cpr.12705] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/04/2019] [Accepted: 09/14/2019] [Indexed: 02/05/2023] Open
Abstract
Objectives Increasing evidences demonstrate a close correlation between epithelial‐to‐mesenchymal transition (EMT) induction and cancer lipid metabolism. However, the molecular mechanisms have not been clarified. Materials and methods In our study, the relative expression level of PRRX1 was detected, its relationship with free fatty acid (FFA) and PPARG2 was analysed in 85 SACC tissues and 15 salivary glands from the benign salivary tumours. We also compared the FFAs composition and levels in these SACC cells. PPARG2 was detected in PRRX1‐induced FFAs treatment as well as Src and MMP‐9 were detected in FFAs treatment–induced invasion and migration of SACC cells, and ChIP test was performed to identify the target interactions. Results Our data showed that overexpression of PRRX1 induced EMT and facilitated the invasion and migration of SACC cells, and PRRX1 expression was closely associated with high FFAs level and poor prognosis of SACC patients. Furthermore, PRRX1 silence led to the increase of PPARG2 and the reduction of FFAs level and the migration and invasion of SACC cells. And inhibition of PPARG2 rescued FFAs level and migration and invasion capabilities of SACC cells. Free fatty acids treatment induced an increase of Stat5‐DNA binding activity via Src‐ and MMP‐9‐dependent pathway. Conclusions Collectively, our findings showed that the PRRX1/PPARG2/FFAs signalling in SACC was important for accelerating tumour metastasis through the induction of EMT and the metabolic reprogramming of FFAs.
Collapse
Affiliation(s)
- Ya-Ping Jiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), Chengdu, China.,Department of Implant, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Sha-Sha Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Jia-Shun Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Mei Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Xin Pang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Jing-Biao Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Yu Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Ya-Jie Tang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China.,Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| |
Collapse
|
47
|
Jin M, Zhang B, Sun Y, Zhang S, Li X, Sik A, Bai Y, Zheng X, Liu K. Involvement of peroxisome proliferator-activated receptor γ in anticonvulsant activity of α-asaronol against pentylenetetrazole-induced seizures in zebrafish. Neuropharmacology 2019; 162:107760. [PMID: 31493468 DOI: 10.1016/j.neuropharm.2019.107760] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 07/06/2019] [Accepted: 09/02/2019] [Indexed: 02/06/2023]
Abstract
In mammals, peroxisome proliferators activated receptors (PPARs), the nuclear hormone receptors, have been reported to be involved in seizure control. Selective agonists and antagonists of PPARs raise seizure thresholds and suppress seizures, respectively. In this study, we evaluated the anticonvulsant effects of α-asaronol, a metabolic product of α-asarone, on pentylenetetrazole (PTZ)-induced seizures in zebrafish and investigated the underlying mechanisms. As a result, α-asaronol ameliorated seizures with increase of seizure latency, as well as decrease of seizure-like behavior, c-fos expression, and abnormal neuronal discharge in a concentration dependent manner. By comparing gene expression profiles of zebrafish undergoing seizures and α-asaronol pretreated zebrafish, we found that α-asaronol attenuate seizures through increase of PPAR γ expression, while PPAR γ antagonist GW9662 inhibit the anti-seizures actions of α-asaronol. Moreover, molecular docking simulation implied the physical interaction between α-asaronol and PPAR γ. The overall results indicated that the anticonvulsant effects of α-asaronol are regulated through PPAR γ-mediated pathway, which shed light on development of α-asaronol as a potential antiepileptic drug. In addition, it is for first time to report that PPAR γ is associated with seizures in zebrafish, supporting previous evidence that zebrafish is a suitable alternative for studying seizures.
Collapse
Affiliation(s)
- Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789, East Jingshi Road, Ji'nan, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Biosensor of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China.
| | - Baoyue Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789, East Jingshi Road, Ji'nan, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Biosensor of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Ying Sun
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, The College of Life Sciences, Northwest University, Xi'an, 710069, Shanxi Province, PR China; Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, Shanxi Province, 710069, PR China
| | - Shanshan Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789, East Jingshi Road, Ji'nan, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Biosensor of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Xiang Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology of Natural Products (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, NO.44 West Culture Road, Ji'nan, 250012, Shandong Province, PR China
| | - Attila Sik
- Institute of Physiology, Medical School, University of Pecs, Pecs, H-7624, Hungary; Szentagothai Research Centre, University of Pecs, Pecs, H-7624, Hungary; Institute of Clinical Sciences, Medical School, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Yajun Bai
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, The College of Life Sciences, Northwest University, Xi'an, 710069, Shanxi Province, PR China; Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, Shanxi Province, 710069, PR China.
| | - Xiaohui Zheng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, The College of Life Sciences, Northwest University, Xi'an, 710069, Shanxi Province, PR China; Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, Shanxi Province, 710069, PR China.
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789, East Jingshi Road, Ji'nan, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Biosensor of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China.
| |
Collapse
|
48
|
Sant KE, Timme-Laragy AR. Zebrafish as a Model for Toxicological Perturbation of Yolk and Nutrition in the Early Embryo. Curr Environ Health Rep 2019; 5:125-133. [PMID: 29417450 DOI: 10.1007/s40572-018-0183-2] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Developmental toxicity assessments often focus on structural outcomes and overlook subtle metabolic differences which occur during the early embryonic period. Deviant embryonic nutrition can result in later-life disease, including diabetes, obesity, and cardiovascular disease. Prior to placenta-mediated nutrient exchange, the human embryo requires maternally supplied nutritional substrates for growth, called yolk. Here, we compare the biology of the human and zebrafish yolk and review examples of toxicant-mediated perturbation of yolk defects, composition, and utilization. RECENT FINDINGS Zebrafish embryos, like human embryos, have a protruding yolk sac that serves as a nutritional cache. Aberrant yolk morphology is a common qualitative finding in fish embryotoxicity studies, but quantitative assessment and characterization provides an opportunity to uncover mechanistic targets of toxicant effects on embryonic nutrition. The zebrafish and the study of its yolk sac is an excellent model for uncovering toxicant disruptions to early embryonic nutrition and has potential to discover mechanistic insights into the developmental origins of health and disease.
Collapse
Affiliation(s)
- Karilyn E Sant
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Goessman 171, 686 N Pleasant St, Amherst, MA, 01003, USA
| | - Alicia R Timme-Laragy
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Goessman 171, 686 N Pleasant St, Amherst, MA, 01003, USA.
| |
Collapse
|
49
|
Kaitetzidou E, Katsiadaki I, Lagnel J, Antonopoulou E, Sarropoulou E. Unravelling paralogous gene expression dynamics during three-spined stickleback embryogenesis. Sci Rep 2019; 9:3752. [PMID: 30842559 PMCID: PMC6403355 DOI: 10.1038/s41598-019-40127-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 02/08/2019] [Indexed: 12/24/2022] Open
Abstract
Development requires the implementation of a plethora of molecular mechanisms, involving a large set of genes to ensure proper cell differentiation, morphogenesis of tissues and organs as well as the growth of the organism. Genome duplication and resulting paralogs are considered to provide the raw genetic materials important for new adaptation opportunities and boosting evolutionary innovation. The present study investigated paralogous genes, involved in three-spined stickleback (Gasterosteus aculeatus) development. Therefore, the transcriptomes of five early stages comprising developmental leaps were explored. Obtained expression profiles reflected the embryo's needs at different stages. Early stages, such as the morula stage comprised transcripts mainly involved in energy requirements while later stages were mostly associated with GO terms relevant to organ development and morphogenesis. The generated transcriptome profiles were further explored for differential expression of known and new paralogous genes. Special attention was given to hox genes, with hoxa13a being of particular interest and to pigmentation genes where itgb1, involved in the melanophore development, displayed a complementary expression pattern throughout studied stages. Knowledge obtained by untangling specific paralogous gene functions during development might not only significantly contribute to the understanding of teleost ontogenesis but might also shed light on paralogous gene evolution.
Collapse
Affiliation(s)
- Elisavet Kaitetzidou
- Department of Zoology, School of Biology, Faculty of Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece.,Institute for Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece
| | - Ioanna Katsiadaki
- Centre for Environment Fisheries and Aquaculture Science, (Cefas), Weymouth, UK
| | - Jacques Lagnel
- Institute for Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece.,Institut National de la Recherche Agronomique (INRA), Génétique et Amélioration des Fruits et Légumes (GALF), Montfavet Cedex, France
| | - Efthimia Antonopoulou
- Department of Zoology, School of Biology, Faculty of Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Elena Sarropoulou
- Institute for Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece.
| |
Collapse
|
50
|
Buerger AN, Schmidt J, Chase A, Paixao C, Patel TN, Brumback BA, Kane AS, Martyniuk CJ, Bisesi JH. Examining the responses of the zebrafish (Danio rerio) gastrointestinal system to the suspected obesogen diethylhexyl phthalate. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 245:1086-1094. [PMID: 30682743 DOI: 10.1016/j.envpol.2018.11.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/26/2018] [Accepted: 11/09/2018] [Indexed: 06/09/2023]
Abstract
Epidemiological evidence suggests that phthalate plasticizers may act as "obesogens", which are chemicals that exacerbate obesity. The gastrointestinal (GI) system is the primary exposure route for phthalates, however, the relationship between phthalate-driven perturbations of GI system functions that can influence obesity has yet to be examined. To address this knowledge gap, we exposed Danio rerio (zebrafish) for 60 days to either (1) Control feeding (5 mg/fish/day), (2) Overfeeding (20 mg/fish/day) or (3) Overfeeding with diethyl-hexyl phthalate (DEHP) (20 mg/fish/day with 3 mg/kg DEHP). After 60 days, Overfed and Overfed + DEHP zebrafish had elevated body mass, and hepatosomatic and gonadosomatic indices. RNAseq analysis of the GI revealed enrichment of gene networks related to lipid metabolism in the Overfed + DEHP group. Many of the enriched networks were under transcriptional control of peroxisome proliferator activated receptor alpha (pparα), a known modulator of lipid metabolism, immune function, and GI function. Real-time PCR confirmed that pparα was overexpressed in the Overfed + DEHP zebrafish, further revealing a pathway by which DEHP may influence lipid metabolism via the GI. These data increase our understanding of phthalate-driven effects on GI function and lipid metabolism, identifying gut-specific gene networks that may drive phthalate-exacerbated obesity.
Collapse
Affiliation(s)
- Amanda N Buerger
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, 32611, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, 32611, USA
| | - Jordan Schmidt
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, 32611, USA; Department of Physiological Sciences, UF Genetics Institute, University of Florida, Gainesville, FL, 32611, USA
| | - Amanda Chase
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, 32611, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, 32611, USA
| | - Carla Paixao
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, 32611, USA; Department of Physiological Sciences, UF Genetics Institute, University of Florida, Gainesville, FL, 32611, USA
| | - Tejas N Patel
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, 32611, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, 32611, USA
| | - Babette A Brumback
- Department of Biostatistics, University of Florida, Gainesville, FL, 32611, USA
| | - Andrew S Kane
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, 32611, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, 32611, USA; Emerging Pathogens Institute, University of Florida, Gainesville, FL, 32611, USA
| | - Christopher J Martyniuk
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, 32611, USA; Department of Physiological Sciences, UF Genetics Institute, University of Florida, Gainesville, FL, 32611, USA
| | - Joseph H Bisesi
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, 32611, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|