1
|
Lingjuan Y, Yu H, Lei Z. Network toxicology and molecular docking techniques to explore the mechanism of bisphenol A on obesity. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2025:1-13. [PMID: 40252053 DOI: 10.1080/09603123.2025.2494735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 04/14/2025] [Indexed: 04/21/2025]
Abstract
The study investigates how bisphenol A (BPA) exposure may lead to obesity (OB) by identifying molecular mechanisms and suggests a new research approach for examining the health effects of chemical toxins. Researchers identified 237 potential targets associated with BPA exposure and OB using CTD, STITCH, DrugBank, GeneCards, and OMIM databases. Analysis with STRING and Cytoscape revealed 10 key targets, including INS, IL-6, AKT1, and PPARG. Enrichment analysis via the DAVID database indicated that these targets are primarily involved in PI3K-Akt and Insulin signaling pathways. These findings indicate that BPA may contribute to the occurrence and development of OB by influencing apoptosis, proliferation, inflammatory signaling, and insulin resistance. Molecular docking showed strong binding of BPA to INS, IL-6, AKT1, and PPARG, with molecular dynamics simulations revealing a stable complex of BPA and PPARG. This study offers insights into BPA's role in OB and supports efforts to prevent and treat OB diseases linked to exposure to BPA-containing plastic products and certain BPA-inundated environments.
Collapse
Affiliation(s)
- Yang Lingjuan
- Innovation Management and High-tech Service center, Productivity Centre of Jiangsu Province, Nanjing, Jiangsu, China
| | - Huang Yu
- Preparation Center, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Zhang Lei
- Preparation Center, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| |
Collapse
|
2
|
Halder SK, Melkani GC. The Interplay of Genetic Predisposition, Circadian Misalignment, and Metabolic Regulation in Obesity. Curr Obes Rep 2025; 14:21. [PMID: 40024983 PMCID: PMC11872776 DOI: 10.1007/s13679-025-00613-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2025] [Indexed: 03/04/2025]
Abstract
PURPOSE OF REVIEW This review explores the complex interplay between genetic predispositions to obesity, circadian rhythms, metabolic regulation, and sleep. It highlights how genetic factors underlying obesity exacerbate metabolic dysfunction through circadian misalignment and examines promising interventions to mitigate these effects. RECENT FINDINGS Genome-wide association Studies (GWAS) have identified numerous Single Nucleotide Polymorphisms (SNPs) associated with obesity traits, attributing 40-75% heritability to body mass index (BMI). These findings illuminate critical links between genetic obesity, circadian clocks, and metabolic processes. SNPs in clock-related genes influence metabolic pathways, with disruptions in circadian rhythms-driven by poor sleep hygiene or erratic eating patterns-amplifying metabolic dysfunction. Circadian clocks, synchronized with the 24-h light-dark cycle, regulate key metabolic activities, including glucose metabolism, lipid storage, and energy utilization. Genetic mutations or external disruptions, such as irregular sleep or eating habits, can destabilize circadian rhythms, promoting weight gain and metabolic disorders. Circadian misalignment in individuals with genetic predispositions to obesity disrupts the release of key metabolic hormones, such as leptin and insulin, impairing hunger regulation and fat storage. Interventions like time-restricted feeding (TRF) and structured physical activity offer promising strategies to restore circadian harmony, improve metabolic health, and mitigate obesity-related risks.
Collapse
Affiliation(s)
- Sajal Kumar Halder
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Girish C Melkani
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
- UAB Nathan Shock Center, Birmingham, AL, 35294, USA.
| |
Collapse
|
3
|
Xiong Y, Huang X, Li Y, Nie Y, Yu H, Shi Y, Xue J, Ji Z, Rong K, Zhang X. Integrating larval zebrafish model and network pharmacology for screening and identification of edible herbs with therapeutic potential for MAFLD: A promising drug Smilax glabra Roxb. Food Chem 2025; 464:141470. [PMID: 39406145 DOI: 10.1016/j.foodchem.2024.141470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/17/2024] [Accepted: 09/27/2024] [Indexed: 11/21/2024]
Abstract
Metabolic-associated fatty liver disease (MAFLD) is becoming a prevalent chronic liver disease. Many medicinal and edible herbs exhibit remarkable biological activities in ameliorating MAFLD but lack a comprehensive assessment of their therapeutic efficacy. This study determined total phenolic and flavonoid contents and in vitro antioxidant properties of 34 edible herbs. Smilax glabra Roxb. (SGR), Coreopsis tinctoria Nutt., and Smilax china L. were obtained with the best bioactivity and antioxidant capacity. The high-cholesterol diet-induced larval zebrafish model was established to compare the anti-MAFLD activity of the three herb extracts mentioned above. In vivo experiments revealed that SGR intervention significantly decreased lipid accumulation, alleviated oxidative stress, and modulated intestinal microbiota composition in zebrafish. Furthermore, three potential active components in SGR and their possible mechanisms were explored through network pharmacology and molecular docking. Our study suggested that SGR is a potential candidate for developing new drugs or dietary supplements for MAFLD therapy.
Collapse
Affiliation(s)
- Yinjuan Xiong
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Xixuan Huang
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Yuxin Li
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Yukang Nie
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Haodong Yu
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Yaqi Shi
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Jiajie Xue
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Zhehui Ji
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Keming Rong
- Research Institute of Huanong-Tianchen, Wuhan 430070, China
| | - Xuezhen Zhang
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China; Research Institute of Huanong-Tianchen, Wuhan 430070, China.
| |
Collapse
|
4
|
Liu R, Wang Z, Shi K, Shen Y, Yu X, Cheng C, Xia Y, Dai G, Zhao Z, Xiong Y, Wang D, Yang L, Yuan G, Jia J. Using Network Pharmacology and Transcriptome Sequencing to Investigate the Mechanism of Action of Luteolin and Quercetin in Treating Obesity. Chem Biol Drug Des 2025; 105:e70061. [PMID: 39909468 DOI: 10.1111/cbdd.70061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/15/2024] [Accepted: 01/22/2025] [Indexed: 02/07/2025]
Abstract
Luteolin and quercetin, which are flavonoids, are present in various traditional Chinese medicines. Although they have been shown to improve obesity, the specific mechanisms of action remain unclear. This study aimed to determine pivotal targets and major regulatory pathways involved in their mechanisms of action using network pharmacology and transcriptome sequencing. Data on luteolin/quercetin-related targets were acquired from the PharmMapper platform, and data on known obesity-related targets were collected from the OMIM and GeneCards databases. Differentially expressed genes (DEGs) involved in luteolin and quercetin action that regulate adipogenic differentiation were identified using RNA sequencing (RNA-seq). Bioinformatic analyses were performed to identify potential target genes and pathways regulated by luteolin/quercetin during adipogenesis. Finally, key genes and pathways were validated through quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting. Network pharmacology showed that luteolin/quercetin was closely associated with anti-obesity targets. The related pathways were metabolic, PI3K/AKT, and MAPK pathways. RNA-seq revealed 91 common DEGs involved in luteolin/quercetin regulation of adipogenic differentiation. Finally, nine potential target genes (including CIDEC, Mgll, Slc2a4, Pck1, and PNPLA3) were identified, and the AMPK and AKT signaling pathways were verified. The present study provides novel information regarding the molecular mechanism of luteolin and quercetin action in treating obesity and demonstrates their therapeutic effects through multiple targets and pathways.
Collapse
Affiliation(s)
- Ruoshuang Liu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zhaoxiang Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Kangru Shi
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yirong Shen
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xiawen Yu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Caiqin Cheng
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yue Xia
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Guoyu Dai
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zhicong Zhao
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yuyun Xiong
- Department of Clinical Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Dong Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ling Yang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Guoyue Yuan
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jue Jia
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
5
|
Zhang L, Song Z, He L, Zhong S, Ju X, Sha H, Xu J, Qin Q, Peng J, Liang H. Unveiling the toxicological effects and risks of prometryn on red swamp crayfish (Procambarus clarkii): Health assessments, ecological, and molecular insights. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 951:175512. [PMID: 39151629 DOI: 10.1016/j.scitotenv.2024.175512] [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: 05/31/2024] [Revised: 07/21/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
Prometryn is commonly used in agricultural and non-agricultural settings. However, possible harm to aquatic organisms remains a persistent concern. Prometryn was also the only one of the 26 triazine herbicides detected in this study. Numerous studies have assessed the harmful effects of prometryn in teleost fish and shrimp. There is a lack of information regarding the ecological and human health risks, as well as the toxic mechanisms affecting crayfish. In this study, human health risk assessment (THQ) and ecological risk assessment (RQ) were conducted on P. clarkii in the rice-crayfish co-culture (IRCC) farming model. The 96 h of exposure to 0.286 mg/L and 1.43 mg/L prometryn was conducted to investigate the potential effects and molecular mechanisms of hepatopancreatic resistance to prometryn in P. clarkii. The original sample analysis revealed that the THQ calculated from the prometryn levels in the muscle and hepatopancreas was below 0.1, suggesting no threat to human health. However, the calculated RQ values were >0.1, indicating a risk to P. clarkii. Histological analysis and biochemical index detection of the experimental samples revealed that the hepatopancreatic injury and oxidative damage in P. clarkii were caused by prometryn. Moreover, transcriptome analysis identified 2512 differentially expressed genes (DEGs) after 96 h of prometryn exposure. Prometryn exposure caused significant changes in metabolic pathways, including oxoacid metabolic processes and cytochrome P450-associated drug metabolism. Further hub gene analysis via PPI indicated that exposure to prometryn may inhibit lipid synthesis, storage, and amino acid transport and affect glucose metabolic pathways and hormone synthesis. Additionally, we hypothesized that prometryn-triggered cell death could be linked to the PI3K-Akt signaling cascade. This study's findings have significant meaning for the efficient and logical application of herbicides in IRCC, ultimately aiding in advancing a highly productive agricultural system.
Collapse
Affiliation(s)
- Lang Zhang
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China.
| | - Ziwei Song
- Department of Genetics, Wuhan University, Wuhan, Hubei 430071, China; School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Li He
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China
| | - Shan Zhong
- Department of Genetics, Wuhan University, Wuhan, Hubei 430071, China; Hubei Province Key Laboratory of Allergy and Immunology, Wuhan, Hubei 430071, China
| | - Xiaoqian Ju
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China
| | - Hang Sha
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China
| | - Jing Xu
- College of Pharmacy, South Central University for Nationalities, Wuhan 430074, China
| | - Qiuying Qin
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China; College of Pharmacy, South Central University for Nationalities, Wuhan 430074, China
| | - Jie Peng
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China.
| | - Hongwei Liang
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China.
| |
Collapse
|
6
|
Lai HH, Jeng KS, Huang CT, Chu AJ, Her GM. Heightened TPD52 linked to metabolic dysfunction and associated abnormalities in zebrafish. Arch Biochem Biophys 2024; 761:110166. [PMID: 39349129 DOI: 10.1016/j.abb.2024.110166] [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: 05/27/2024] [Revised: 09/05/2024] [Accepted: 09/25/2024] [Indexed: 10/02/2024]
Abstract
The tumor protein D52 (TPD52) gene encodes a proto-oncogene protein associated with various medical conditions, including breast and prostate cancers. It plays a role in multiple biological pathways such as cell growth, differentiation, and apoptosis. The function of TPD52 in lipid droplet biosynthesis has been investigated in vitro. However, its precise role in lipid metabolism in animal models is not fully understood. To investigate the functions of TPD52 in vivo, we performed a conditional TPD52 protein expression analysis using a Tet-off transgenic system to establish conditionally expressed Tpd52 transgenic zebrafish. The effect of Tpd52 on lipogenesis was assessed using various methods, including whole-mount Oil Red O staining, histological examination, and measurement of inflammatory markers and potential targets using real-time quantitative polymerase chain reaction and immunoblotting in Tpd52 fish. Zebrafish with increased Tpd52 levels exhibited notable weight gain and the enlargement of fat deposits, which were mainly attributed to an increase in the volume of adipocytes. Moreover, Tpd52 overexpression was correlated with the triggering of the adipocyte differentiation signaling pathway. During adipocytic differentiation in response to nutrient status, our observations revealed adipogenesis, nonalcoholic fatty liver disease, and metabolic cardiomyopathy (MCM) in Tpd52 transgenic zebrafish. To gain a deeper understanding of the contribution of these proteins to the regulation of cellular growth, we investigated the expression of their corresponding genes and proteins in zebrafish. In the present study, the activated protein kinase pathway was identified as the primary target of TPD52. Adult Tpd52 zebrafish showed increased lipid accumulation, resulting in the development of visceral obesity, nonalcoholic fatty liver disease, and MCM. These findings strongly suggest that TPD52 actively contributes to adipose tissue expansion and its subsequent effects. This investigation provides compelling evidence that Tpd52 facilitates adipocyte development and related metabolic comorbidities in zebrafish.
Collapse
Affiliation(s)
- Hsin-Hung Lai
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Kuo-Shyang Jeng
- Division of General Surgery, Far Eastern Memorial Hospital, New Taipei, 220, Taiwan
| | - Chung-Tsui Huang
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Far Eastern Memorial Hospital, New Taipei, 220, Taiwan
| | - An-Ju Chu
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Guor Mour Her
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
| |
Collapse
|
7
|
Chen H, Sun B, Gao W, Qiu Y, Wei W, Li Y, Ye W, Song H, Hua C, Lin X. PIK3CA mutations enhance the adipogenesis of ADSCs in facial infiltrating lipomatosis through TRPV1. iScience 2024; 27:110467. [PMID: 39104411 PMCID: PMC11298645 DOI: 10.1016/j.isci.2024.110467] [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: 10/27/2023] [Revised: 06/15/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
Facial infiltrating lipomatosis (FIL) is a congenital disorder. The pathogenesis of FIL is associated with PIK3CA mutations, but the underlying mechanisms remain undetermined. We found that the adipose tissue in FIL demonstrated adipocytes hypertrophy and increased lipid accumulation. All adipose-derived mesenchymal stem cells from FIL (FIL-ADSCs) harbored PIK3CA mutations. Moreover, FIL-ADSCs exhibited a greater capacity for adipogenesis. Knockdown of PIK3CA resulted in a reduction in the adipogenic potential of FIL-ADSCs. Furthermore, WX390, a dual-target PI3K/mTOR inhibitor, was found to impede PIK3CA-mediated adipogenesis both in vivo and in vitro. RNA sequencing (RNA-seq) revealed that the expression of transient receptor potential vanilloid subtype 1 (TRPV1) was upregulated after PI3K pathway inhibition, and overexpression or activation of TRPV1 both inhibited adipogenesis. Our study showed that PIK3CA mutations promoted adipogenesis in FIL-ADSCs and this effect was achieved by suppressing TPRV1. Pathogenesis experiments suggested that WX390 may serve as an agent for the treatment of FIL.
Collapse
Affiliation(s)
- Hongrui Chen
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Bin Sun
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Wei Gao
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yajing Qiu
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Wei Wei
- Shanghai Jiatan Pharmatech Co, LTD, Shanghai, China
| | - Yongguo Li
- Shanghai Jiatan Pharmatech Co, LTD, Shanghai, China
| | - Wei Ye
- Shanghai Jiatan Pharmatech Co, LTD, Shanghai, China
| | | | - Chen Hua
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Xiaoxi Lin
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
8
|
Pedrosa VF, Santos PADPD, Giesta LB, Zemor JC, Okamoto MH, Chagas AC, Bezerra ADC, Poersch LHDS, Romano LA. Occurrence of hepatic and splenic lipomas in Nile tilapia (Oreochromis niloticus). JOURNAL OF FISH DISEASES 2024; 47:e13869. [PMID: 37792353 DOI: 10.1111/jfd.13869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/05/2023]
Affiliation(s)
- Virgínia F Pedrosa
- Laboratory of Immunology and Pathology of Aquatic Organisms, Institute of Oceanography, Federal University of Rio Grande, Brazil
| | - Pedro A De P Dos Santos
- Laboratory of Immunology and Pathology of Aquatic Organisms, Institute of Oceanography, Federal University of Rio Grande, Brazil
| | - Luana B Giesta
- Laboratory of Immunology and Pathology of Aquatic Organisms, Institute of Oceanography, Federal University of Rio Grande, Brazil
| | - Julio C Zemor
- Institute of Oceanography, Aquaculture Marine Station, Federal University of Rio Grande, Brazil
| | - Marcelo H Okamoto
- Institute of Oceanography, Aquaculture Marine Station, Federal University of Rio Grande, Brazil
| | - Andrezza C Chagas
- Institute of Oceanography, Aquaculture Marine Station, Federal University of Rio Grande, Brazil
| | - Aline da C Bezerra
- Institute of Oceanography, Aquaculture Marine Station, Federal University of Rio Grande, Brazil
| | - Luis H da S Poersch
- Institute of Oceanography, Aquaculture Marine Station, Federal University of Rio Grande, Brazil
| | - Luis A Romano
- Laboratory of Immunology and Pathology of Aquatic Organisms, Institute of Oceanography, Federal University of Rio Grande, Brazil
| |
Collapse
|
9
|
Sanni O, Fasemore T, Nkomozepi P. Non-Genetic-Induced Zebrafish Model for Type 2 Diabetes with Emphasis on Tools in Model Validation. Int J Mol Sci 2023; 25:240. [PMID: 38203409 PMCID: PMC10778736 DOI: 10.3390/ijms25010240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/08/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
The unrelenting increase in the incidence of type 2 diabetes (T2D) necessitates the urgent need for effective animal models to mimic its pathophysiology. Zebrafish possess human-like metabolic traits and share significant genetic similarities, making them valuable candidates for studying metabolic disorders, including T2D. This review emphasizes the critical role of animal models in diabetes research, especially focusing on zebrafish as an alternative model organism. Different approaches to a non-genetic model of T2D in zebrafish, such as the glucose solution, diet-induced, chemical-induced, and combined diet-induced and glucose solution methods, with an emphasis on model validation using indicators of T2D, were highlighted. However, a significant drawback lies in the validation of these models. Some of these models have not extensively demonstrated persistent hyperglycemia or response to insulin resistance and glucose tolerance tests, depicted the morphology of the pancreatic β-cell, or showed their response to antidiabetic drugs. These tools are crucial in T2D pathology. Future research on non-genetic models of T2D in zebrafish must extensively focus on validating the metabolic deficits existing in the model with the same metabolic defects in humans and improve on the existing models for a better understanding of the molecular mechanisms underlying T2D and exploring potential therapeutic interventions.
Collapse
Affiliation(s)
- Olakunle Sanni
- Department of Human Anatomy and Physiology, University of Johannesburg, Doornfontein 2028, South Africa; (T.F.); (P.N.)
| | | | | |
Collapse
|
10
|
Ding Y, Tang Z, Zhang R, Zhang M, Guan Q, Zhang L, Wang H, Chen Y, Zhang W, Wang J. Genetic Variations of AKT1 are Associated with Risk Screening for Non-Alcoholic Fatty Liver Disease. Risk Manag Healthc Policy 2023; 16:1365-1376. [PMID: 37525829 PMCID: PMC10387243 DOI: 10.2147/rmhp.s416592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 07/12/2023] [Indexed: 08/02/2023] Open
Abstract
Purpose Protein kinase B (PKB/AKT) has shown a high profile in the research of metabolic diseases. This research sought to determine whether the AKT1 gene's single nucleotide polymorphisms (SNPs) and the risk of developing non-alcoholic fatty liver disease (NAFLD) were related. Patients and Methods Recruited in this case-control study were 2693 subjects, including 815 with NAFLD and 1878 without NAFLD. Three SNPs of AKT1 (rs2494732, rs2494752 and rs1130233) were genotyped. To examine the correlation between SNPs and NAFLD susceptibility, logistic regression was performed. Results After adjusting for sex, age, triglyceride and glucose, AKT1 rs2494732-C (all P < 0.05 in co-dominant model, dominant model and additive model) and rs2494752-G (P < 0.05 in co-dominant model) were linked to a lower risk of NAFLD. The combined effect of both SNPs on NAFLD risk was statistically significant, showing a dose dependence (Ptrend = 0.010). Sex, body mass index, hypertension, hyperglycemia, hypertriglyceridemia, high-density lipoprotein-cholesterol, alanine aminotransferase, and beneficial alleles were all significant predictors of NAFLD risk (all P < 0.05). The prediction model achieved good discrimination, with an area under the receiver operating characteristic curve of 0.779. The Hosmer-Lemeshow test suggested an inadequate calibration of the model (χ2 = 21.073, P = 0.007). Conclusion AKT1 rs2494732 and rs2494752 may be related to Chinese NAFLD susceptibility. The prediction model combining both SNPs with clinical factors displays a strong ability to discriminate NAFLD patients. Both SNPs may be exploited to design new models for early screening of NAFLD high-risk population.
Collapse
Affiliation(s)
- Yajie Ding
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Zongzhe Tang
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Ru Zhang
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Mengting Zhang
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Qing Guan
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Liuxin Zhang
- The Nethersole School of Nursing, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong (SAR), People’s Republic of China
| | - Hongliang Wang
- Department of General Practice, Ninghai Road Community Health Service Center, Nanjing, Jiangsu, People’s Republic of China
| | - Yue Chen
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Wei Zhang
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai, People’s Republic of China
| | - Jie Wang
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| |
Collapse
|
11
|
Patel P, Nandi A, Verma SK, Kaushik N, Suar M, Choi EH, Kaushik NK. Zebrafish-based platform for emerging bio-contaminants and virus inactivation research. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 872:162197. [PMID: 36781138 PMCID: PMC9922160 DOI: 10.1016/j.scitotenv.2023.162197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/23/2023] [Accepted: 02/08/2023] [Indexed: 05/27/2023]
Abstract
Emerging bio-contaminants such as viruses have affected health and environment settings of every country. Viruses are the minuscule entities resulting in severe contagious diseases like SARS, MERS, Ebola, and avian influenza. Recent epidemic like the SARS-CoV-2, the virus has undergone mutations strengthen them and allowing to escape from the remedies. Comprehensive knowledge of viruses is essential for the development of targeted therapeutic and vaccination treatments. Animal models mimicking human biology like non-human primates, rats, mice, and rabbits offer competitive advantage to assess risk of viral infections, chemical toxins, nanoparticles, and microbes. However, their economic maintenance has always been an issue. Furthermore, the redundancy of experimental results due to aforementioned aspects is also in examine. Hence, exploration for the alternative animal models is crucial for risk assessments. The current review examines zebrafish traits and explores the possibilities to monitor emerging bio-contaminants. Additionally, a comprehensive picture of the bio contaminant and virus particle invasion and abatement mechanisms in zebrafish and human cells is presented. Moreover, a zebrafish model to investigate the emerging viruses such as coronaviridae and poxviridae has been suggested.
Collapse
Affiliation(s)
- Paritosh Patel
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea
| | - Aditya Nandi
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Suresh K Verma
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India; Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden
| | - Neha Kaushik
- Department of Biotechnology, College of Engineering, The University of Suwon, 18323 Hwaseong, Republic of Korea
| | - Mrutyunjay Suar
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Eun Ha Choi
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea.
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea.
| |
Collapse
|
12
|
Ghaddar B, Diotel N. Zebrafish: A New Promise to Study the Impact of Metabolic Disorders on the Brain. Int J Mol Sci 2022; 23:ijms23105372. [PMID: 35628176 PMCID: PMC9141892 DOI: 10.3390/ijms23105372] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 02/01/2023] Open
Abstract
Zebrafish has become a popular model to study many physiological and pathophysiological processes in humans. In recent years, it has rapidly emerged in the study of metabolic disorders, namely, obesity and diabetes, as the regulatory mechanisms and metabolic pathways of glucose and lipid homeostasis are highly conserved between fish and mammals. Zebrafish is also widely used in the field of neurosciences to study brain plasticity and regenerative mechanisms due to the high maintenance and activity of neural stem cells during adulthood. Recently, a large body of evidence has established that metabolic disorders can alter brain homeostasis, leading to neuro-inflammation and oxidative stress and causing decreased neurogenesis. To date, these pathological metabolic conditions are also risk factors for the development of cognitive dysfunctions and neurodegenerative diseases. In this review, we first aim to describe the main metabolic models established in zebrafish to demonstrate their similarities with their respective mammalian/human counterparts. Then, in the second part, we report the impact of metabolic disorders (obesity and diabetes) on brain homeostasis with a particular focus on the blood-brain barrier, neuro-inflammation, oxidative stress, cognitive functions and brain plasticity. Finally, we propose interesting signaling pathways and regulatory mechanisms to be explored in order to better understand how metabolic disorders can negatively impact neural stem cell activity.
Collapse
|
13
|
Zhang J, Abou-Fadel J, Renteria M, Belkin O, Chen B, Zhu Y, Dammann P, Rigamonti D. Cerebral cavernous malformations do not fall in the spectrum of PIK3CA-related overgrowth. J Neurol Neurosurg Psychiatry 2022; 93:jnnp-2022-328901. [PMID: 35477890 DOI: 10.1136/jnnp-2022-328901] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/23/2022] [Indexed: 11/04/2022]
Abstract
Somatic gain-of-function (GOF) mutations in phosphatidylinositol-4, 5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), the catalytic subunit of phosphoinositide 3-kinase (PI3K), have been recently discovered in cerebral cavernous malformations (CCMs), raising the possibility that the activation of PI3K pathways is a possible universal regulator of vascular morphogenesis. However, there have been contradicting data presented among various groups and studies. To enhance the current understanding of vascular anomalies, it is essential to explore this possible relationship between altered PI3K signalling pathways and its influence on the pathogenesis of CCMs. GOF PIK3CA-mutants have been linked to overgrowth syndromes, allowing this group of disorders, resulting from somatic activating mutations in PIK3CA, to be collectively named as PIK3CA-related overgrowth spectrum disorders. This paper reviews and attempts to conceptualise the relationships and differences among clinical presentations, genotypic and phenotypic correlations and possible coexistence of PIK3CA and CCM mutations/phenotypes in CCM lesions. Finally, we present a model reflecting our hypothetical understanding of CCM pathogenesis based on a systematic review and conceptualisation of data obtained from other studies.
Collapse
Affiliation(s)
- Jun Zhang
- Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas, USA
| | - Johnathan Abou-Fadel
- Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas, USA
| | - Mellisa Renteria
- Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas, USA
| | - Ofek Belkin
- Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas, USA
| | - Bixia Chen
- Department of Neurosurgery, University of Duisburg-Essen, Essen, Germany
| | - Yuan Zhu
- Department of Neurosurgery, University of Duisburg-Essen, Essen, Germany
| | - Philipp Dammann
- Department of Neurosurgery, University of Duisburg-Essen, Essen, Germany
| | | |
Collapse
|
14
|
Obesity Affects the Proliferative Potential of Equine Endometrial Progenitor Cells and Modulates Their Molecular Phenotype Associated with Mitochondrial Metabolism. Cells 2022; 11:cells11091437. [PMID: 35563743 PMCID: PMC9100746 DOI: 10.3390/cells11091437] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/04/2022] [Accepted: 04/20/2022] [Indexed: 02/01/2023] Open
Abstract
The study aimed to investigate the influence of obesity on cellular features of equine endometrial progenitor cells (Eca EPCs), including viability, proliferation capacity, mitochondrial metabolism, and oxidative homeostasis. Eca EPCs derived from non-obese (non-OB) and obese (OB) mares were characterized by cellular phenotype and multipotency. Obesity-induced changes in the activity of Eca EPCs include the decline of their proliferative activity, clonogenic potential, mitochondrial metabolism, and enhanced oxidative stress. Eca EPCs isolated from obese mares were characterized by an increased occurrence of early apoptosis, loss of mitochondrial dynamics, and senescence-associated phenotype. Attenuated metabolism of Eca EPCs OB was related to increased expression of pro-apoptotic markers (CASP9, BAX, P53, P21), enhanced expression of OPN, PI3K, and AKT, simultaneously with decreased signaling stabilizing cellular homeostasis (including mitofusin, SIRT1, FOXP3). Obesity alters functional features and the self-renewal potential of endometrial progenitor cells. The impaired cytophysiology of progenitor cells from obese endometrium predicts lower regenerative capacity if used as autologous transplants.
Collapse
|
15
|
Jia D, Zhang R, Shao J, Zhang W, Cai L, Sun W. Exposure to trace levels of metals and fluoroquinolones increases inflammation and tumorigenesis risk of zebrafish embryos. ENVIRONMENTAL SCIENCE AND ECOTECHNOLOGY 2022; 10:100162. [PMID: 36159734 PMCID: PMC9488011 DOI: 10.1016/j.ese.2022.100162] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 05/04/2023]
Abstract
Exposure to trace-level heavy metals and antibiotics may elicit metabolic disorder, alter protein expression, and then induce pathological changes in zebrafish embryos, despite negligible physiological and developmental toxicity. This study investigated the single and combined developmental toxicity of fluoroquinolones (enrofloxacin [ENR] and ciprofloxacin [CIP]) (≤0.5 μM) and heavy metals (Cu and Cd) (≤0.5 μM) to zebrafish embryos, and molecular responses of zebrafish larvae upon exposure to the single pollutant (0.2 μM) or a binary metal-fluoroquinolone mixture (0.2 μM). In all single and mixture exposure groups, no developmental toxicity was observed, but oxidative stress, inflammation, and lipid depletion were found in zebrafish embryos, which was more severe in the mixture exposure groups than in the single exposure groups, probably due to increased metal bioaccumulation in the presence of ENR or CIP. Metabolomics analysis revealed the up-regulation of amino acids and down-regulation of fatty acids, corresponding to an active response to oxidative stress and the occurrence of inflammation. The up-regulation of antioxidase and immune proteins revealed by proteomics analysis further confirmed the occurrence of oxidative stress and inflammation. Furthermore, the KEGG pathway enrichment analysis showed a significant disturbance of pathways related to immunity and tumor, indicating the potential risk of tumorigenesis in zebrafish larvae. The findings provide molecular-level insights into the adverse effects of heavy metals and antibiotics (especially in chemical mixtures) on zebrafish embryos, and highlight the potential ecotoxicological risks of trace-level heavy metals and antibiotics in the environment.
Collapse
Affiliation(s)
- Dantong Jia
- College of Environmental Sciences and Engineering, Peking University, The Key Laboratory of Water and Sediment Sciences, Ministry of Education, Beijing, 100871, China
- State Environmental Protection Key Laboratory of All Material Fluxes in River Ecosystems, International Joint Laboratory for Regional Pollution Control, Ministry of Education, Beijing, 100871, China
| | - Ruijie Zhang
- College of Environmental Sciences and Engineering, Peking University, The Key Laboratory of Water and Sediment Sciences, Ministry of Education, Beijing, 100871, China
- State Environmental Protection Key Laboratory of All Material Fluxes in River Ecosystems, International Joint Laboratory for Regional Pollution Control, Ministry of Education, Beijing, 100871, China
| | - Jian Shao
- College of Animal Science, Guizhou University, The Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guiyang, Guizhou, 550025, China
| | - Wei Zhang
- Department of Plant, Soil and Microbial Sciences, Environmental Science and Policy Program, Michigan State University, East Lansing, MI, 48824, United States
| | - Leilei Cai
- College of Safety and Environmental Engineering, Shandong University of Science and Technology, Qingdao, Shandong, 266590, China
| | - Weiling Sun
- College of Environmental Sciences and Engineering, Peking University, The Key Laboratory of Water and Sediment Sciences, Ministry of Education, Beijing, 100871, China
- State Environmental Protection Key Laboratory of All Material Fluxes in River Ecosystems, International Joint Laboratory for Regional Pollution Control, Ministry of Education, Beijing, 100871, China
- Corresponding author. Peking University. China.
| |
Collapse
|
16
|
Tsai YW, Jeng KS, He MK, Hsieh YW, Lai HH, Lai CY, Huang CC, Chang CF, Huang CT, Her GM. MXD3 Promotes Obesity and the Androgen Receptor Signaling Pathway in Gender-Disparity Hepatocarcinogenesis. Cells 2021; 10:3434. [PMID: 34943942 PMCID: PMC8700344 DOI: 10.3390/cells10123434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/02/2021] [Accepted: 12/04/2021] [Indexed: 12/26/2022] Open
Abstract
Obesity is closely linked to metabolic diseases, particularly non-alcoholic steatohepatitis (NASH) or non-alcoholic fatty liver disease (NAFLD), ultimately leading to hepatocellular carcinoma (HCC). However, the molecular mechanisms of NASH-associated HCC (NAHCC) remain elusive. To explore the impact of Max dimerization protein 3 (MXD3), a transcription factor that regulates several cellular functions in disorders associated with metabolic diseases, we conditionally expressed Mxd3 proteins using Tet-on mxd3 transgenic zebrafish (MXs) with doxycycline (MXs + Dox) or without doxycycline (MXs - Dox) treatment. Overexpression of global MXD3 (gMX) or hepatic Mxd3 (hMX) was associated with obesity-related NAFLD pathophysiology in gMX + Dox, and liver fibrosis and HCC in hMX + Dox. Oil Red O (ORO)-stained signals were seen in intravascular blood vessels and liver buds of larval gMX + Dox, indicating that Mxd3 functionally promotes lipogenesis. The gMX + Dox-treated young adults exhibited an increase in body weight and visceral fat accumulation. The hMX + Dox-treated young adults showed normal body characteristics but exhibited liver steatosis and NASH-like phenotypes. Subsequently, steatohepatitis, liver fibrosis, and NAHCC were found in 6-month-old gMX + Dox adults compared with gMX - Dox adults at the same stage. Overexpression of Mxd3 also enhanced AR expression accompanied by the increase of AR-signaling pathways resulting in hepatocarcinogenesis in males. Our results demonstrate that global actions of Mxd3 are central to the initiation of obesity in the gMX zebrafish through their effects on adipogenesis and that MXD3 could serve as a therapeutic target for obesity-associated liver diseases.
Collapse
Affiliation(s)
- Yi-Wen Tsai
- Department of Family Medicine, Chang Gung Memorial Hospital, Keelung 204, Taiwan;
- College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan
| | - Kuo-Shyang Jeng
- Division of General Surgery, Far Eastern Memorial Hospital, New Taipei 220, Taiwan; (K.-S.J.); (C.-F.C.)
| | - Mu-Kuang He
- Taipei First Girls High School, Taipei 100, Taiwan;
| | - Yang-Wen Hsieh
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung 202, Taiwan;
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (H.-H.L.); (C.-Y.L.)
| | - Hsin-Hung Lai
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (H.-H.L.); (C.-Y.L.)
| | - Chi-Yu Lai
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (H.-H.L.); (C.-Y.L.)
| | - Chun-Chieh Huang
- Department of Radiology, Far Eastern Memorial Hospital, New Taipei 220, Taiwan;
| | - Chiung-Fang Chang
- Division of General Surgery, Far Eastern Memorial Hospital, New Taipei 220, Taiwan; (K.-S.J.); (C.-F.C.)
| | - Chung-Tsui Huang
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Far Eastern Memorial Hospital, New Taipei 220, Taiwan;
| | - Guor Mour Her
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (H.-H.L.); (C.-Y.L.)
| |
Collapse
|
17
|
Kirstein AS, Kehr S, Nebe M, Hanschkow M, Barth LAG, Lorenz J, Penke M, Breitfeld J, Le Duc D, Landgraf K, Körner A, Kovacs P, Stadler PF, Kiess W, Garten A. PTEN regulates adipose progenitor cell growth, differentiation, and replicative aging. J Biol Chem 2021; 297:100968. [PMID: 34273354 PMCID: PMC8350019 DOI: 10.1016/j.jbc.2021.100968] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/17/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022] Open
Abstract
The tumor suppressor phosphatase and tensin homolog (PTEN) negatively regulates the insulin signaling pathway. Germline PTEN pathogenic variants cause PTEN hamartoma tumor syndrome (PHTS), associated with lipoma development in children. Adipose progenitor cells (APCs) lose their capacity to differentiate into adipocytes during continuous culture, whereas APCs from lipomas of patients with PHTS retain their adipogenic potential over a prolonged period. It remains unclear which mechanisms trigger this aberrant adipose tissue growth. To investigate the role of PTEN in adipose tissue development, we performed functional assays and RNA-Seq of control and PTEN knockdown APCs. Reduction of PTEN levels using siRNA or CRISPR led to enhanced proliferation and differentiation of APCs. Forkhead box protein O1 (FOXO1) transcriptional activity is known to be regulated by insulin signaling, and FOXO1 was downregulated at the mRNA level while its inactivation through phosphorylation increased. FOXO1 phosphorylation initiates the expression of the lipogenesis-activating transcription factor sterol regulatory element-binding protein 1 (SREBP1). SREBP1 levels were higher after PTEN knockdown and may account for the observed enhanced adipogenesis. To validate this, we overexpressed constitutively active FOXO1 in PTEN CRISPR cells and found reduced adipogenesis, accompanied by SREBP1 downregulation. We observed that PTEN CRISPR cells showed less senescence compared with controls and the senescence marker CDKN1A (p21) was downregulated in PTEN knockdown cells. Cellular senescence was the most significantly enriched pathway found in RNA-Seq of PTEN knockdown versus control cells. These results provide evidence that PTEN is involved in the regulation of APC proliferation, differentiation, and senescence, thereby contributing to aberrant adipose tissue growth in patients with PHTS.
Collapse
Affiliation(s)
- Anna S Kirstein
- University Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany.
| | - Stephanie Kehr
- Bioinformatics Group, Department of Computer Science and Interdisciplinary Center for Bioinformatics, Leipzig University, Leipzig, Germany
| | - Michèle Nebe
- University Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | - Martha Hanschkow
- University Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | - Lisa A G Barth
- University Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | - Judith Lorenz
- University Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | - Melanie Penke
- University Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | - Jana Breitfeld
- Medical Department III-Endocrinology, Nephrology, Rheumatology, Leipzig University Medical Center, Leipzig, Germany
| | - Diana Le Duc
- Institute of Human Genetics, Leipzig University Medical Center, Leipzig, Germany; Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Kathrin Landgraf
- University Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | - Antje Körner
- University Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | - Peter Kovacs
- Medical Department III-Endocrinology, Nephrology, Rheumatology, Leipzig University Medical Center, Leipzig, Germany
| | - Peter F Stadler
- Bioinformatics Group, Department of Computer Science and Interdisciplinary Center for Bioinformatics, Leipzig University, Leipzig, Germany; Max Planck Institute for Mathematics in the Sciences, Leipzig, Germany
| | - Wieland Kiess
- University Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | - Antje Garten
- University Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany; Institute for Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
18
|
Lumaquin D, Johns E, Montal E, Weiss JM, Ola D, Abuhashem A, White RM. An in vivo reporter for tracking lipid droplet dynamics in transparent zebrafish. eLife 2021; 10:e64744. [PMID: 34114952 PMCID: PMC8195600 DOI: 10.7554/elife.64744] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 05/14/2021] [Indexed: 01/22/2023] Open
Abstract
Lipid droplets are lipid storage organelles found in nearly all cell types from adipocytes to cancer cells. Although increasingly implicated in disease, current methods to study lipid droplets in vertebrate models rely on static imaging or the use of fluorescent dyes, limiting investigation of their rapid in vivo dynamics. To address this, we created a lipid droplet transgenic reporter in whole animals and cell culture by fusing tdTOMATO to Perilipin-2 (PLIN2), a lipid droplet structural protein. Expression of this transgene in transparent casper zebrafish enabled in vivo imaging of adipose depots responsive to nutrient deprivation and high-fat diet. Simultaneously, we performed a large-scale in vitro chemical screen of 1280 compounds and identified several novel regulators of lipolysis in adipocytes. Using our Tg(-3.5ubb:plin2-tdTomato) zebrafish line, we validated several of these novel regulators and revealed an unexpected role for nitric oxide in modulating adipocyte lipid droplets. Similarly, we expressed the PLIN2-tdTOMATO transgene in melanoma cells and found that the nitric oxide pathway also regulated lipid droplets in cancer. This model offers a tractable imaging platform to study lipid droplets across cell types and disease contexts using chemical, dietary, or genetic perturbations.
Collapse
Affiliation(s)
- Dianne Lumaquin
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD ProgramNew YorkUnited States
| | - Eleanor Johns
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Gerstner Sloan Kettering Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Emily Montal
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Joshua M Weiss
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD ProgramNew YorkUnited States
| | - David Ola
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Abderhman Abuhashem
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD ProgramNew YorkUnited States
- Developmental Biology Program, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Richard M White
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| |
Collapse
|
19
|
Abstract
Zebrafish are rapidly becoming a leading model organism for cancer research. The genetic pathways driving cancer are highly conserved between zebrafish and humans, and the ability to easily manipulate the zebrafish genome to rapidly generate transgenic animals makes zebrafish an excellent model organism. Transgenic zebrafish containing complex, patient-relevant genotypes have been used to model many cancer types. Here we present a comprehensive review of transgenic zebrafish cancer models as a resource to the field and highlight important areas of cancer biology that have yet to be studied in the fish. The ability to image cancer cells and niche biology in an endogenous tumor makes zebrafish an indispensable model organism in which we can further understand the mechanisms that drive tumorigenesis and screen for potential new cancer therapies.
Collapse
Affiliation(s)
- Alicia M. McConnell
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
- Harvard Stem Cell Institute, Boston, Massachusetts 02138, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Haley R. Noonan
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
- Harvard Stem Cell Institute, Boston, Massachusetts 02138, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
- Biological and Biomedical Sciences Program, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Leonard I. Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
- Harvard Stem Cell Institute, Boston, Massachusetts 02138, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
- Stem Cell and Regenerative Biology Department and Howard Hughes Medical Institute, Harvard University, Boston, Massachusetts 02138, USA
| |
Collapse
|
20
|
Wang Q, Luo C, Lu G, Chen Z. Effect of adenosine monophosphate-activated protein kinase-p53-Krüppel-like factor 2a pathway in hyperglycemia-induced cardiac remodeling in adult zebrafish. J Diabetes Investig 2020; 12:320-333. [PMID: 32881390 PMCID: PMC7926222 DOI: 10.1111/jdi.13393] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 08/07/2020] [Accepted: 08/12/2020] [Indexed: 01/10/2023] Open
Abstract
Aims/Introduction Diabetic cardiomyopathy is a type of myocardial disease. It causes left ventricular hypertrophy, followed by diastolic and systolic dysfunction, eventually leading to congestive heart failure. However, the underlying mechanism still requires further elucidation. Materials and Methods A high‐glucose zebrafish model was constructed by administering streptozocin intraperitoneally to enhance the development of cardiomyopathy and then treated with adenosine monophosphate‐activated protein kinase (AMPK) activator. Cardiac structure and function, and protein and gene expression were then analyzed. Cardiomyocytes (CMs) culture in vitro using lentivirus were used for detection of AMPK, p53 and Krüppel‐like factor 2a (klf2a) gene expression. Results In the hyperglycemia group, electrocardiogram findings showed arrhythmia, echocardiography results showed heart enlargement and dysfunction, and many differences, such as increased apoptosis and myocardial fiber loss, were observed. The phospho‐AMPK and klf2a expression were downregulated, and p53 expression was upregulated. Activation of phospho‐AMPK reduced p53 and increased klf2a expression, alleviated apoptosis in CMs and improved cardiac function in the hyperglycemic zebrafish. In vitro knockdown system of AMPK, p53 and klf2a using lentivirus illustrated an increased p53 expression and decreased klf2a expression in CMs by inhibiting AMPK. Repression of p53 and upregulation of klf2a expression were observed, but no changes in the expression of AMPK and its phosphorylated type. Conclusions In the model of streptozocin‐induced hyperglycemia zebrafish, the reduction of phosphorylated AMPK increased p53, which led to KLF2a decrease to facilitate apoptosis of CMs, inducing the cardiac remodeling and cardiac dysfunction. These results can be reversed by AMPK activator, which means the AMPK–p53–klf2a pathway might be a potential target for diabetic cardiomyopathy intervention.
Collapse
Affiliation(s)
- Qiuyun Wang
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chen Luo
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Guoping Lu
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhenyue Chen
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Knuth MM, Mahapatra D, Jima D, Wan D, Hammock BD, Law M, Kullman SW. Vitamin D deficiency serves as a precursor to stunted growth and central adiposity in zebrafish. Sci Rep 2020; 10:16032. [PMID: 32994480 PMCID: PMC7524799 DOI: 10.1038/s41598-020-72622-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 09/03/2020] [Indexed: 12/16/2022] Open
Abstract
Emerging evidence demonstrates the importance of sufficient vitamin D (1α, 25-dihydroxyvitamin D3) levels during early life stage development with deficiencies associated with long-term effects into adulthood. While vitamin D has traditionally been associated with mineral ion homeostasis, accumulating evidence suggests non-calcemic roles for vitamin D including metabolic homeostasis. In this study, we examined the hypothesis that vitamin D deficiency (VDD) during early life stage development precedes metabolic disruption. Three dietary cohorts of zebrafish were placed on engineered diets including a standard laboratory control diet, a vitamin D null diet, and a vitamin D enriched diet. Zebrafish grown on a vitamin D null diet between 2-12 months post fertilization (mpf) exhibited diminished somatic growth and enhanced central adiposity associated with accumulation and enlargement of visceral and subcutaneous adipose depots indicative of both adipocyte hypertrophy and hyperplasia. VDD zebrafish exhibited elevated hepatic triglycerides, attenuated plasma free fatty acids and attenuated lipoprotein lipase activity consistent with hallmarks of dyslipidemia. VDD induced dysregulation of gene networks associated with growth hormone and insulin signaling, including induction of suppressor of cytokine signaling. These findings indicate that early developmental VDD impacts metabolic health by disrupting the balance between somatic growth and adipose accumulation.
Collapse
Affiliation(s)
- Megan M Knuth
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Campus Box 7633, Raleigh, NC, 27695-7633, USA.
| | - Debabrata Mahapatra
- Comparative Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, 27606, USA
| | - Dereje Jima
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, 27606, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, 27606, USA
| | - Debin Wan
- Department of Entomology and Nematology and University of California Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, 95616, USA
| | - Bruce D Hammock
- Department of Entomology and Nematology and University of California Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, 95616, USA
| | - Mac Law
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, 27606, USA
| | - Seth W Kullman
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Campus Box 7633, Raleigh, NC, 27695-7633, USA.
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, 27606, USA.
| |
Collapse
|
22
|
Saggini A, Santonja C, Nájera L, Palmedo G, Kutzner H. Frequent activating PIK3CA mutations in sporadic angiolipoma. J Cutan Pathol 2020; 48:211-216. [PMID: 32662895 DOI: 10.1111/cup.13809] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/05/2020] [Accepted: 07/01/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Angiolipoma (AL) is considered as a lipoma variant that is characterized by the combination of mature adipocytes and capillary blood vessels diffusely distributed within the tumor. With the exception of recurrent PRKD2 mutations of uncertain pathogenetic significance, the genetic abnormalities of ALs are unknown, in the absence of any of the specific chromosomal aberrations described in other lipoma variants. METHODS Formalin-fixed and paraffin-embedded blocks of 13 conventional ALs and 5 cellular ALs from 17 individuals were retrieved and analyzed for mutations in exons 9 and 20 of PIK3CA by polymerase chain reaction and Sanger sequencing. RESULTS Activating PIK3CA mutations were identified in 14 tumors (78%). All PIK3CA-mutated samples carried the same exon 9 mutation, c.1634A>C (p.E545A). No mutation was detected in exon 20 of PIK3CA. No significant difference between PIK3CA-mutated and wild-type samples appeared to exist based on age, gender, and location of the tumor. All 5 cellular ALs carried the p.E545A PIK3CA mutation. CONCLUSION The high frequency of the p.E545A PIK3CA mutation in both conventional and cellular ALs suggests that activation of the PI3K/AKT pathway plays a key role in AL pathogenesis and reinforces the concept that cellular AL should be regarded as a variant of AL.
Collapse
Affiliation(s)
| | - Carlos Santonja
- Department of Pathology, Jiménez Díaz Foundation University Hospital, Madrid, Spain
| | - Laura Nájera
- Department of Pathology, Puerta de Hierro University Hospital, Madrid, Spain
| | | | | |
Collapse
|
23
|
Raby L, Völkel P, Le Bourhis X, Angrand PO. Genetic Engineering of Zebrafish in Cancer Research. Cancers (Basel) 2020; 12:E2168. [PMID: 32759814 PMCID: PMC7464884 DOI: 10.3390/cancers12082168] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/19/2022] Open
Abstract
Zebrafish (Danio rerio) is an excellent model to study a wide diversity of human cancers. In this review, we provide an overview of the genetic and reverse genetic toolbox allowing the generation of zebrafish lines that develop tumors. The large spectrum of genetic tools enables the engineering of zebrafish lines harboring precise genetic alterations found in human patients, the generation of zebrafish carrying somatic or germline inheritable mutations or zebrafish showing conditional expression of the oncogenic mutations. Comparative transcriptomics demonstrate that many of the zebrafish tumors share molecular signatures similar to those found in human cancers. Thus, zebrafish cancer models provide a unique in vivo platform to investigate cancer initiation and progression at the molecular and cellular levels, to identify novel genes involved in tumorigenesis as well as to contemplate new therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | - Pierre-Olivier Angrand
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277–CANTHER–Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (L.R.); (P.V.); (X.L.B.)
| |
Collapse
|
24
|
Kaur N, Chugh H, Tomar V, Sakharkar MK, Dass SK, Chandra R. Cinnamon attenuates adiposity and affects the expression of metabolic genes in Diet-Induced obesity model of zebrafish. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2930-2939. [PMID: 31317780 DOI: 10.1080/21691401.2019.1641509] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The prevalence of obesity is increasing at an alarming rate worldwide with about 30% of the world population classified as obese. Obese body structure results when energy intake exceeds energy expenditure in an individual. Increase in the consumption of high-energy eatables, in the context of portion and energy provided, has resulted in obese populations which is becoming the leading cause of metabolic disorders related to morbidity. The obesity-related comorbidity is an enormous liability on health services and will affect measures taken in tackling the increasing obesity rate. Prevention of an obese phenotype is the most suitable long-term strategy. Another approach towards the treatment of obesity is weight management through phytotherapeutics. In this study, we explored the anti-obesity effects of Cinnamon (Cinnamomum zeylanicum) in adult male zebrafish. Through BMI measurements, blood glucose level analyses, serum triglyceride analyses, Oil Red O staining as well as quantitative Real Time-PCR, the ability of cinnamon to reduce metabolic disorders associated with obesity is investigated for the first time in a zebrafish model. Our studies indicate that cinnamon ameliorates the genotypic and phonotypic characteristics associated with obesity through lowering of BMI, blood glucose, triglyceride levels, lipid levels in the liver and through gene modulation.
Collapse
Affiliation(s)
- Navrinder Kaur
- a Department of Chemistry, University of Delhi , Delhi , India.,b Department of Research and Education, Artemis Hospitals , Gurgaon , India
| | - Heerak Chugh
- a Department of Chemistry, University of Delhi , Delhi , India
| | - Vartika Tomar
- a Department of Chemistry, University of Delhi , Delhi , India
| | | | - Sujata K Dass
- d Department of Neurology, B.L. Kapur Hospital , New Delhi , India
| | - Ramesh Chandra
- a Department of Chemistry, University of Delhi , Delhi , India.,e Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi , Delhi , India
| |
Collapse
|
25
|
Meguro S, Hosoi S, Hasumura T. High-fat diet impairs cognitive function of zebrafish. Sci Rep 2019; 9:17063. [PMID: 31745184 PMCID: PMC6863811 DOI: 10.1038/s41598-019-53634-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 11/03/2019] [Indexed: 01/14/2023] Open
Abstract
An unhealthy diet with excessive fat intake has often been claimed to induce not only obesity but also cognitive dysfunction in mammals; however, it is not known whether this is the case in zebrafish. Here, we investigated the effect of excessive fat in the diet on cognitive function and on gene expression in the telencephalon of zebrafish. Cognitive function, as measured by active avoidance test, was impaired by feeding of a high-fat diet compared with a control diet. In RNA sequencing analysis of the telencephalon, 97 genes were identified with a fold change in expression greater than 2 and a p-value less than 0.05 between the two diets. In quantitative real-time PCR analysis of the telencephalon, genes related to neuronal activity, anti-oxidative stress, blood–brain barrier function and amyloid-β degradation were found to be downregulated, whereas genes related to apoptosis and amyloid-β production were found to be upregulated, in the high-fat diet group, which are changes known to occur in mammals fed a high-fat diet. Collectively, these results are similar to those found in mammals, suggesting that zebrafish can serve as a suitable animal model in research into cognitive impairment induced by excessive fat in the diet.
Collapse
Affiliation(s)
- Shinichi Meguro
- Biological Science Research, Kao Corporation, 2606 Akabane, Ichikai-machi, Haga-gun, Tochigi, 321-3497, Japan.
| | - Sayaka Hosoi
- Biological Science Research, Kao Corporation, 1334 Minato, Wakayama, Wakayama, 640-8580, Japan
| | - Takahiro Hasumura
- Biological Science Research, Kao Corporation, 2606 Akabane, Ichikai-machi, Haga-gun, Tochigi, 321-3497, Japan
| |
Collapse
|
26
|
Khor ES, Noor SM, Wong PF. Understanding the Role of ztor in Aging-related Diseases Using the Zebrafish Model. In Vivo 2019; 33:1713-1720. [PMID: 31662495 DOI: 10.21873/invivo.11661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/15/2019] [Accepted: 08/16/2019] [Indexed: 12/14/2022]
Abstract
The mammalian target of rapamycin (mTOR), a 289 kDa serine/threonine protein kinase of the phosphoinositide 3-kinase (PI3K)-related family is known for its role in regulating lifespan and the aging process in humans and rodents. Aging in zebrafish very much resembles aging in humans. Aged zebrafish often manifest with spinal curvature, cataracts and cognitive frailty, akin to human age-related phenotypical effects such as osteoarthritis, dwindling vision and cognitive dysfunction. However, the role of the zebrafish orthologue of mTOR, ztor, is less defined in these areas. This review paper discusses the tale of growing old in the zebrafish, the physiological roles of ztor in normal developmental processes and its involvement in the pathogenesis of aging-related diseases such as metabolic disorders and cancers.
Collapse
Affiliation(s)
- Eng-Soon Khor
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Suzita Mohd Noor
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Pooi-Fong Wong
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
27
|
Yen CC, Chen SC, Hung GY, Wu PK, Chua WY, Lin YC, Yen CH, Chen YC, Wang JY, Yang MH, Chao Y, Chang MC, Chen WM. Expression profile‑driven discovery of AURKA as a treatment target for liposarcoma. Int J Oncol 2019; 55:938-948. [PMID: 31485600 DOI: 10.3892/ijo.2019.4861] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 07/23/2019] [Indexed: 11/06/2022] Open
Abstract
Liposarcoma (LPS) is one of the most frequently reported type of soft‑tissue sarcoma (STS). Well‑differentiated (WD) LPS and dedifferentiated (DD) LPS are the two most common subtypes. Chemotherapy has been considered to be ineffective in LPS, and novel treatment agents are thus necessary. In this study, we reanalyzed two published microarray data sets of LPS. By comparing the top 50 upregulated genes in DD LPS in both sets of data, we identified 12 overlapping genes. Of note, the top five gene sets enriched in DD LPS in both sets of data were involved in cell cycle regulation. Among the 12 overlapping genes, aurora kinase A (AURKA) is a well‑known gene involved in cell cycle regulation; we thus further investigated this gene. AURKA was significantly upregulated in DD LPS, compared with WD LPS. Among 40 cases of DD LPS in GSE30929, patients with high AURKA expression in tumors had significantly worse distant recurrence‑free survival than those with low expression. In an in vitro model, MLN8237, an AURKA inhibitor, could inhibit AURKA in LPS cell lines with a resultant G2/M arrest. MLN8237 was also reported to exert a cytotoxic effect by inducing apoptosis in LPS cell lines. Furthermore, except for cisplatin, MLN8237 had a significantly synergistic effect with chemotherapy agents against LPS. MLN8237 induced cellular senescence in LPS cell lines with increased expression of DcR2, a senescence biomarker, and upregulated expression of cytokines associated with the senescence‑associated secretory phenotype, including interleukin (IL)‑1α, IL‑6 and IL‑8. Our study identified AURKA as a potential biomarker for predicting poor prognosis in LPS. The findings of the present study suggested the potential of AURKA as a therapeutic target in LPS cell line models, while the novel combination of AURKA inhibitors and chemotherapy requires further investigation.
Collapse
Affiliation(s)
- Chueh-Chuan Yen
- Division of Medical Oncology, Center for Immuno‑Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 11217, Taiwan, R.O.C
| | - San-Chi Chen
- Division of Medical Oncology, Center for Immuno‑Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 11217, Taiwan, R.O.C
| | - Giun-Yi Hung
- Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 11217, Taiwan, R.O.C
| | - Po-Kuei Wu
- Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 11217, Taiwan, R.O.C
| | - Wei-Yang Chua
- Division of Medical Oncology, Center for Immuno‑Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 11217, Taiwan, R.O.C
| | - Yung-Chan Lin
- Division of Medical Oncology, Center for Immuno‑Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 11217, Taiwan, R.O.C
| | - Chiao-Han Yen
- Division of Medical Oncology, Center for Immuno‑Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 11217, Taiwan, R.O.C
| | - Yen-Chun Chen
- Division of Medical Oncology, Center for Immuno‑Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 11217, Taiwan, R.O.C
| | - Jir-You Wang
- Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 11217, Taiwan, R.O.C
| | - Muh-Hwa Yang
- Division of Medical Oncology, Center for Immuno‑Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 11217, Taiwan, R.O.C
| | - Yee Chao
- Division of Medical Oncology, Center for Immuno‑Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 11217, Taiwan, R.O.C
| | - Ming-Chau Chang
- Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 11217, Taiwan, R.O.C
| | - Wei-Ming Chen
- Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 11217, Taiwan, R.O.C
| |
Collapse
|
28
|
Tran VC, Cho SY, Kwon J, Kim D. Alginate oligosaccharide (AOS) improves immuno-metabolic systems by inhibiting STOML2 overexpression in high-fat-diet-induced obese zebrafish. Food Funct 2019; 10:4636-4648. [DOI: 10.1039/c9fo00982e] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AOS improves immuno-metabolism systems in high-fat-died-induced obese zebrafish by regulating STOML2.
Collapse
Affiliation(s)
- Van Cuong Tran
- Department of Food Science and Technology
- Chonnam National University
- Gwangju
- Republic of Korea
- Department of Food Science and Post-harvest Technology
| | - Se-Young Cho
- Biological Disaster Analysis Group
- Korea Basic Science Institute
- Daejeon
- Republic of Korea
| | - Joseph Kwon
- Biological Disaster Analysis Group
- Korea Basic Science Institute
- Daejeon
- Republic of Korea
| | - Duwoon Kim
- Department of Food Science and Technology
- Chonnam National University
- Gwangju
- Republic of Korea
- Foodborne Virus Research Center
| |
Collapse
|
29
|
Faillaci F, Milosa F, Critelli RM, Turola E, Schepis F, Villa E. Obese zebrafish: A small fish for a major human health condition. Animal Model Exp Med 2018; 1:255-265. [PMID: 30891575 PMCID: PMC6388073 DOI: 10.1002/ame2.12042] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/11/2018] [Accepted: 10/18/2018] [Indexed: 12/14/2022] Open
Abstract
Obesity is becoming a silent worldwide epidemic, with a steady increase in both adults and children. To date, even though several drugs have been licensed for long-term obesity treatment, none of them are yet used in routine clinical practice. So far the only successful intervention has been behavioral therapy. A suitable and economic experimental model mimicking the human condition would therefore be extremely useful to evaluate preventive measures and novel treatments. Zebrafish are emerging as an important model system to study obesity and related metabolic disease. Remarkable similarities have been reported in lipid metabolism and the adipogenic pathway between zebrafish and mammals. Moreover, the zebrafish possesses a number of features-the relative inexpensiveness of animal husbandry, its optical transparency and the ability to produce a large number of offspring at low cost-that make it ideal for large-scale screening and for testing drugs and intervention. In this review, we summarize recent progress in using zebrafish as a model system to study obesity and obesity-related metabolic disorders. We describe several zebrafish models (in both larvae and adult animals) that develop obesity and non-alcoholic fatty liver disease (NAFLD) using different approaches, including gene manipulation, diet manipulation and modification of microbiota composition. For these models, we have outlined the specific aspects related to obesity and its development and we have summarized their advantages and limitations.
Collapse
Affiliation(s)
- Francesca Faillaci
- Department of Internal MedicineGastroenterology UnitUniversity of Modena and Reggio EmiliaModenaItaly
- Women in Hepatology GroupModenaItaly
| | - Fabiola Milosa
- Women in Hepatology GroupModenaItaly
- National Institute of Gastroenterology“S. de Bellis” Research HospitalCastellana GrotteItaly
| | - Rosina Maria Critelli
- Department of Internal MedicineGastroenterology UnitUniversity of Modena and Reggio EmiliaModenaItaly
- Women in Hepatology GroupModenaItaly
| | - Elena Turola
- Department of Internal MedicineEndocrinology UnitAOU of ParmaParmaItaly
| | - Filippo Schepis
- Department of Internal MedicineGastroenterology UnitUniversity of Modena and Reggio EmiliaModenaItaly
| | - Erica Villa
- Department of Internal MedicineGastroenterology UnitUniversity of Modena and Reggio EmiliaModenaItaly
- Women in Hepatology GroupModenaItaly
| |
Collapse
|
30
|
Howe DG, Blake JA, Bradford YM, Bult CJ, Calvi BR, Engel SR, Kadin JA, Kaufman TC, Kishore R, Laulederkind SJF, Lewis SE, Moxon SAT, Richardson JE, Smith C. Model organism data evolving in support of translational medicine. Lab Anim (NY) 2018; 47:277-289. [PMID: 30224793 PMCID: PMC6322546 DOI: 10.1038/s41684-018-0150-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 08/13/2018] [Indexed: 02/07/2023]
Abstract
Model organism databases (MODs) have been collecting and integrating biomedical research data for 30 years and were designed to meet specific needs of each model organism research community. The contributions of model organism research to understanding biological systems would be hard to overstate. Modern molecular biology methods and cost reductions in nucleotide sequencing have opened avenues for direct application of model organism research to elucidating mechanisms of human diseases. Thus, the mandate for model organism research and databases has now grown to include facilitating use of these data in translational applications. Challenges in meeting this opportunity include the distribution of research data across many databases and websites, a lack of data format standards for some data types, and sustainability of scale and cost for genomic database resources like MODs. The issues of widely distributed data and application of data standards are some of the challenges addressed by FAIR (Findable, Accessible, Interoperable, and Re-usable) data principles. The Alliance of Genome Resources is now moving to address these challenges by bringing together expertly curated research data from fly, mouse, rat, worm, yeast, zebrafish, and the Gene Ontology consortium. Centralized multi-species data access, integration, and format standardization will lower the data utilization barrier in comparative genomics and translational applications and will provide a framework in which sustainable scale and cost can be addressed. This article presents a brief historical perspective on how the Alliance model organisms are complementary and how they have already contributed to understanding the etiology of human diseases. In addition, we discuss four challenges for using data from MODs in translational applications and how the Alliance is working to address them, in part by applying FAIR data principles. Ultimately, combined data from these animal models are more powerful than the sum of the parts.
Collapse
Affiliation(s)
- Douglas G Howe
- The Institute of Neuroscience, University of Oregon, Eugene, OR, USA.
| | | | - Yvonne M Bradford
- The Institute of Neuroscience, University of Oregon, Eugene, OR, USA
| | | | - Brian R Calvi
- Department of Biology, Indiana University, Bloomington, IN, USA
| | - Stacia R Engel
- Department of Genetics, Stanford University, Palo Alto, CA, USA
| | | | | | - Ranjana Kishore
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Stanley J F Laulederkind
- Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, WI, USA
| | - Suzanna E Lewis
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Sierra A T Moxon
- The Institute of Neuroscience, University of Oregon, Eugene, OR, USA
| | | | | |
Collapse
|
31
|
Zang L, Maddison LA, Chen W. Zebrafish as a Model for Obesity and Diabetes. Front Cell Dev Biol 2018; 6:91. [PMID: 30177968 PMCID: PMC6110173 DOI: 10.3389/fcell.2018.00091] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 07/25/2018] [Indexed: 12/13/2022] Open
Abstract
Obesity and diabetes now considered global epidemics. The prevalence rates of diabetes are increasing in parallel with the rates of obesity and the strong connection between these two diseases has been coined as “diabesity.” The health risks of overweight or obesity include Type 2 diabetes mellitus (T2DM), coronary heart disease and cancer of numerous organs. Both obesity and diabetes are complex diseases that involve the interaction of genetics and environmental factors. The underlying pathogenesis of obesity and diabetes are not well understood and further research is needed for pharmacological and surgical management. Consequently, the use of animal models of obesity and/or diabetes is important for both improving the understanding of these diseases and to identify and develop effective treatments. Zebrafish is an attractive model system for studying metabolic diseases because of the functional conservation in lipid metabolism, adipose biology, pancreas structure, and glucose homeostasis. It is also suited for identification of novel targets associated with the risk and treatment of obesity and diabetes in humans. In this review, we highlight studies using zebrafish to model metabolic diseases, and discuss the advantages and disadvantages of studying pathologies associated with obesity and diabetes in zebrafish.
Collapse
Affiliation(s)
- Liqing Zang
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, United States.,Graduate School of Regional Innovation Studies, Mie University, Tsu, Japan
| | - Lisette A Maddison
- Center for Reproductive Biology, Washington State University, Pullman, WA, United States
| | - Wenbiao Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, United States
| |
Collapse
|
32
|
Völkel P, Dupret B, Le Bourhis X, Angrand PO. [The zebrafish model in oncology]. Med Sci (Paris) 2018; 34:345-353. [PMID: 29658479 DOI: 10.1051/medsci/20183404016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Although cell culture and mouse models will remain a cornerstone of cancer research, the unique capabilities of the zebrafish outline the potential of this model for shedding light on cancer biology in vivo. Zebrafish develops cancers spontaneously, after chemical mutagenesis or through genetic manipulations. Furthermore, zebrafish cancers are similar to human tumors at the histological and molecular levels allowing the study of tumor initiation, progression and heterogeneity. Xenotransplantation of human cancer cells in embryos or adult zebrafish presents the advantage of following cancer cell behavior in vivo. Finally, zebrafish embryos are used in molecule screens and contribute to the identification of novel anti-cancer therapeutic strategies. Here, we review different involvements of the zebrafish model in cancer research.
Collapse
Affiliation(s)
- Pamela Völkel
- CNRS Lille, Inserm U908, Université de Lille, Bâtiment SN3, Cité Scientifique, 59655 Villeneuve d'Ascq, France
| | - Babara Dupret
- Inserm U908, Université de Lille, Bâtiment SN3, Cité Scientifique, 59655 Villeneuve d'Ascq, France
| | - Xuefen Le Bourhis
- Inserm U908, Université de Lille, Bâtiment SN3, Cité Scientifique, 59655 Villeneuve d'Ascq, France
| | - Pierre-Olivier Angrand
- Inserm U908, Université de Lille, Bâtiment SN3, Cité Scientifique, 59655 Villeneuve d'Ascq, France
| |
Collapse
|
33
|
Jang YJ, Son HJ, Ahn J, Jung CH, Ha T. Coumestrol modulates Akt and Wnt/β-catenin signaling during the attenuation of adipogenesis. Food Funct 2018; 7:4984-4991. [PMID: 27868125 DOI: 10.1039/c6fo01127f] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Coumestrol is a natural phytochemical present in plants such as red clover and soy, and has been reported to stimulate the estrogen receptor as a major phytoestrogen. While the molecular mechanisms responsible for the anti-adipogenic effects of phytoestrogens such as genistein and daidzein have been previously investigated, the effects of coumestrol on adipogenesis remain to be elucidated. We observed that coumestrol dose-dependently attenuates MDI (mixture of 3-isobutyl-1-methylxanthine, dexamethasone, and insulin)-induced lipid accumulation, consistent with an earlier study, while significantly inhibiting MDI-induced adipogenesis in the first 48 hours of differentiation, a critical time window for anti-adipogenic effects. Coumestrol treatment suppressed MDI-induced protein expression of PPARγ and C/EBPα in adipocytes, leading to the subsequent downregulation of FAS and aP2 expression. Akt and GSK3β were phosphorylated shortly after MDI stimulation, and these responses were inhibited by coumestrol treatment. Coumestrol also increased LRP6 protein expression, resulting in the recovery of β-catenin downregulation by MDI, while attenuating MDI-induced downregulation of Wnt10b. In addition, mRNA and protein expression of c-Myc and cyclin D1, target genes of β-catenin, were both recovered by coumestrol treatment. These results suggest that coumestrol inhibits adipocyte differentiation via regulation of Akt and Wnt/β-catenin signaling and may have potential for development as an agent to prevent adipogenesis.
Collapse
Affiliation(s)
- Young Jin Jang
- Metabolic Mechanism Research Group, Korea Food Research Institute, Seongnam, Republic of Korea.
| | - Hyo Jeong Son
- Metabolic Mechanism Research Group, Korea Food Research Institute, Seongnam, Republic of Korea.
| | - Jiyun Ahn
- Metabolic Mechanism Research Group, Korea Food Research Institute, Seongnam, Republic of Korea. and Division of Food Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| | - Chang Hwa Jung
- Metabolic Mechanism Research Group, Korea Food Research Institute, Seongnam, Republic of Korea. and Division of Food Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| | - Taeyoul Ha
- Metabolic Mechanism Research Group, Korea Food Research Institute, Seongnam, Republic of Korea. and Division of Food Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
34
|
Kamel M, Ninov N. Catching new targets in metabolic disease with a zebrafish. Curr Opin Pharmacol 2017; 37:41-50. [DOI: 10.1016/j.coph.2017.08.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 08/04/2017] [Accepted: 08/11/2017] [Indexed: 12/12/2022]
|
35
|
Abstract
Zebrafish is an often used model of vertebrate lipid metabolism. In this article, we examined the effects of diets rich in fish oil, a dietary fat that has been shown to have antiobesity effects in mammals, or lard on body fat accumulation in zebrafish. Adult female zebrafish were fed a high-fat diet containing 20% (w/w) fish oil or lard for 4 weeks. Fish in the fish oil diet group had less body fat accumulation compared with those in the lard diet group. In the intestine, expression of genes for the alpha (hadhaa) and beta (hadhb) subunits of the beta-oxidation enzyme hydroxyacyl-Coenzyme A dehydrogenase/3-ketoacyl-Coenzyme A thiolase/enoyl-Coenzyme A hydratase was significantly increased in the fish oil diet group compared with the lard diet group (p < 0.05). In the liver, expression of the gene for fatty acid synthase (fasn) was significantly decreased in the fish oil diet group compared with the lard diet group (p < 0.05). These results suggest that the mechanisms underlying the antiobesity effect of fish oil are similar in zebrafish and mammals.
Collapse
Affiliation(s)
- Shinichi Meguro
- Biological Science Research , Kao Corporation, Tochigi, Japan
| | | |
Collapse
|
36
|
Gut P, Reischauer S, Stainier DYR, Arnaout R. LITTLE FISH, BIG DATA: ZEBRAFISH AS A MODEL FOR CARDIOVASCULAR AND METABOLIC DISEASE. Physiol Rev 2017; 97:889-938. [PMID: 28468832 PMCID: PMC5817164 DOI: 10.1152/physrev.00038.2016] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/09/2017] [Accepted: 01/10/2017] [Indexed: 12/17/2022] Open
Abstract
The burden of cardiovascular and metabolic diseases worldwide is staggering. The emergence of systems approaches in biology promises new therapies, faster and cheaper diagnostics, and personalized medicine. However, a profound understanding of pathogenic mechanisms at the cellular and molecular levels remains a fundamental requirement for discovery and therapeutics. Animal models of human disease are cornerstones of drug discovery as they allow identification of novel pharmacological targets by linking gene function with pathogenesis. The zebrafish model has been used for decades to study development and pathophysiology. More than ever, the specific strengths of the zebrafish model make it a prime partner in an age of discovery transformed by big-data approaches to genomics and disease. Zebrafish share a largely conserved physiology and anatomy with mammals. They allow a wide range of genetic manipulations, including the latest genome engineering approaches. They can be bred and studied with remarkable speed, enabling a range of large-scale phenotypic screens. Finally, zebrafish demonstrate an impressive regenerative capacity scientists hope to unlock in humans. Here, we provide a comprehensive guide on applications of zebrafish to investigate cardiovascular and metabolic diseases. We delineate advantages and limitations of zebrafish models of human disease and summarize their most significant contributions to understanding disease progression to date.
Collapse
Affiliation(s)
- Philipp Gut
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Sven Reischauer
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Didier Y R Stainier
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Rima Arnaout
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| |
Collapse
|
37
|
Hayes MN, Langenau DM. Discovering novel oncogenic pathways and new therapies using zebrafish models of sarcoma. Methods Cell Biol 2017; 138:525-561. [PMID: 28129857 DOI: 10.1016/bs.mcb.2016.11.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Sarcoma is a type of cancer affecting connective, supportive, or soft tissue of mesenchymal origin. Despite rare incidence in adults (<1%), over 15% of pediatric cancers are sarcoma. Sadly, both adults and children with relapsed or metastatic disease have devastatingly high rates of mortality. Current treatment options for sarcoma include surgery, radiation, and/or chemotherapy; however, significant limitations exist with respect to the efficacy of these strategies. Strong impetus has been placed on the development of novel therapies and preclinical models for uncovering mechanisms involved in the development, progression, and therapy resistance of sarcoma. Over the past 15 years, the zebrafish has emerged as a powerful genetic model of human cancer. High genetic conservation when combined with a unique susceptibility to develop sarcoma has made the zebrafish an effective tool for studying these diseases. Transgenic and gene-activation strategies have been employed to develop zebrafish models of rhabdomyosarcoma, malignant peripheral nerve sheath tumors, Ewing's sarcoma, chordoma, hemangiosarcoma, and liposarcoma. These models all display remarkable molecular and histopathological conservation with their human cancer counterparts and have offered excellent platforms for understanding disease progression in vivo. Short tumor latency and the amenability of zebrafish for ex vivo manipulation, live imaging studies, and tumor cell transplantation have allowed for efficient study of sarcoma initiation, growth, self-renewal, and maintenance. When coupled with facile chemical genetic approaches, zebrafish models of sarcoma have provided a strong translational tool to uncover novel drug pathways and new therapeutic strategies.
Collapse
Affiliation(s)
- M N Hayes
- Massachusetts General Hospital, Boston, MA, United States; Massachusetts General Hospital, Charlestown, MA, United States; Harvard Stem Cell Institute, Boston, MA, United States
| | - D M Langenau
- Massachusetts General Hospital, Boston, MA, United States; Massachusetts General Hospital, Charlestown, MA, United States; Harvard Stem Cell Institute, Boston, MA, United States
| |
Collapse
|
38
|
Newman T, Jhinku N, Meier M, Horsfield J. Dietary Intake Influences Adult Fertility and Offspring Fitness in Zebrafish. PLoS One 2016; 11:e0166394. [PMID: 27870856 PMCID: PMC5117665 DOI: 10.1371/journal.pone.0166394] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 10/27/2016] [Indexed: 01/30/2023] Open
Abstract
The burden of malnutrition, including both over- and undernutrition, is a major public health concern. Here we used a zebrafish model of diet-induced obesity to analyze the impact of dietary intake on fertility and the phenotype of the next generation. Over an eight-week period, one group received 60 mg of food each day (60 mg arm), while another received 5 mg (5 mg arm). At the end of the diet, the body mass index of the 60 mg arm was 1.5 fold greater than the 5 mg arm. The intervention also had a marked impact on fertility; breeding success and egg production in the 60 mg arm were increased 2.1- and 6.2-fold compared to the 5 mg arm, respectively. Transcriptome analysis of eggs revealed that transcripts involved in metabolic biological processes differed according to dietary intake. The progeny from the differentially fed fish were more likely to survive when the parents had access to more food. An intergenerational crossover study revealed that while parental diet did not influence weight gain in the offspring, the progeny of well-fed parents had increased levels of physical activity when exposed again to high nutrient availability. We conclude that dietary intake has an important influence on fertility and the subsequent fitness of offspring, even prior to breeding.
Collapse
Affiliation(s)
- Trent Newman
- Department of Pathology, Dunedin School of Medicine, PO Box 913, University of Otago, Dunedin, 9054, New Zealand
| | - Noel Jhinku
- Department of Pathology, Dunedin School of Medicine, PO Box 913, University of Otago, Dunedin, 9054, New Zealand
| | - Michael Meier
- Department of Pathology, Dunedin School of Medicine, PO Box 913, University of Otago, Dunedin, 9054, New Zealand
- Gravida: National Centre for Growth and Development, University of Auckland, Auckland, New Zealand
| | - Julia Horsfield
- Department of Pathology, Dunedin School of Medicine, PO Box 913, University of Otago, Dunedin, 9054, New Zealand
- Gravida: National Centre for Growth and Development, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| |
Collapse
|
39
|
Otis JP, Farber SA. High-fat Feeding Paradigm for Larval Zebrafish: Feeding, Live Imaging, and Quantification of Food Intake. J Vis Exp 2016. [PMID: 27842350 DOI: 10.3791/54735] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Zebrafish are emerging as a model of dietary lipid processing and metabolic disease. This protocol describes how to feed larval zebrafish a lipid-rich meal, which consists of an emulsion of chicken egg yolk liposomes created by sonicating egg yolk in embryo media. Detailed instructions are provided to screen larvae for egg yolk consumption so that larvae that fail to feed will not confound experimental results. The chicken egg yolk liposomes can be spiked with fluorescent lipid analogs, including fatty acids and cholesterol, enabling both systemic and subcellular visualization of dietary lipid processing. Several methods are described to mount larvae that are conducive to short- and long-term live imaging with both upright and inverted objectives at high and low magnification. Additionally presented is an assay to quantify larval food intake by extracting the lipids of larvae fed fluorescent lipid analogs, spotting the lipids on a thin layer chromatography plate, and quantifying the fluorescence. Finally, critical aspects of the procedures, important controls, options for modifying the protocols to address specific experimental questions, and potential limitations are discussed. These techniques can be applied not only to focused, hypothesis driven inquiries, but also to a variety of screens and live imaging techniques to study dietary lipid metabolism and the control of food intake.
Collapse
Affiliation(s)
- Jessica P Otis
- Department of Embryology, Carnegie Institution for Science
| | - Steven A Farber
- Department of Embryology, Carnegie Institution for Science; Department of Biology, Johns Hopkins University;
| |
Collapse
|
40
|
Morales Fénero CI, Colombo Flores AA, Câmara NOS. Inflammatory diseases modelling in zebrafish. World J Exp Med 2016; 6:9-20. [PMID: 26929916 PMCID: PMC4759353 DOI: 10.5493/wjem.v6.i1.9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 10/20/2015] [Accepted: 12/18/2015] [Indexed: 02/06/2023] Open
Abstract
The ingest of diets with high content of fats and carbohydrates, low or no physical exercise and a stressful routine are part of the everyday lifestyle of most people in the western world. These conditions are triggers for different diseases with complex interactions between the host genetics, the metabolism, the immune system and the microbiota, including inflammatory bowel diseases (IBD), obesity and diabetes. The incidence of these disorders is growing worldwide; therefore, new strategies for its study are needed. Nowadays, the majority of researches are in use of murine models for understand the genetics, physiopathology and interaction between cells and signaling pathways to find therapeutic solutions to these diseases. The zebrafish, a little tropical water fish, shares 70% of our genes and conserves anatomic and physiological characteristics, as well as metabolical pathways, with mammals, and is rising as a new complementary model for the study of metabolic and inflammatory diseases. Its high fecundity, fast development, transparency, versatility and low cost of maintenance makes the zebrafish an interesting option for new researches. In this review, we offer a discussion of the existing genetic and induced zebrafish models of two important Western diseases that have a strong inflammatory component, the IBD and the obesity.
Collapse
|
41
|
Ouadah-Boussouf N, Babin PJ. Pharmacological evaluation of the mechanisms involved in increased adiposity in zebrafish triggered by the environmental contaminant tributyltin. Toxicol Appl Pharmacol 2016; 294:32-42. [PMID: 26812627 DOI: 10.1016/j.taap.2016.01.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 01/09/2016] [Accepted: 01/09/2016] [Indexed: 01/06/2023]
Abstract
One proposed contributing factor to the rise in overweight and obesity is exposure to endocrine disrupting chemicals. Tributyltin chloride (TBT), an organotin, induces adipogenesis in cell culture models and may increases adipose mass in vivo in vertebrate model organisms. It has been hypothesized that TBT acts via the peroxisome proliferator activated receptor (PPAR)γ-dependent pathway. However, the mechanisms involved in the effects of TBT exposure on in vivo adipose tissue metabolism remain unexplored. Semitransparent zebrafish larvae, with their well-developed white adipose tissue, offer a unique opportunity for studying the effects of toxicant chemicals and pharmaceuticals on adipocyte biology and whole-organism adiposity in a vertebrate model. Within hours, zebrafish larvae, treated at environmentally-relevant nanomolar concentrations of TBT, exhibited a remarkable increase in adiposity linked to adipocyte hypertrophy. Under the experimental conditions used, we also demonstrated that zebrafish larvae adipose tissue proved to be highly responsive to selected human nuclear receptor agonists and antagonists. Retinoid X receptor (RXR) homodimers and RXR/liver X receptor heterodimers were suggested to be in vivo effectors of the obesogenic effect of TBT on zebrafish white adipose tissue. RXR/PPARγ heterodimers may be recruited to modulate adiposity in zebrafish but were not a necessary requirement for the short term in vivo TBT obesogenic effect. Together, the present results suggest that TBT may induce the promotion of triacylglycerol storage in adipocytes via RXR-dependent pathways without necessary using PPAR isoforms.
Collapse
Affiliation(s)
- Nafia Ouadah-Boussouf
- Maladies Rares: Génétique et Métabolisme (MRGM), Univ. Bordeaux, INSERM, U1211, F-33615 Pessac, France
| | - Patrick J Babin
- Maladies Rares: Génétique et Métabolisme (MRGM), Univ. Bordeaux, INSERM, U1211, F-33615 Pessac, France.
| |
Collapse
|
42
|
Ceol CJ, Houvras Y. Uncharted Waters: Zebrafish Cancer Models Navigate a Course for Oncogene Discovery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 916:3-19. [PMID: 27165347 DOI: 10.1007/978-3-319-30654-4_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Over a decade has elapsed since the first genetically-engineered zebrafish cancer model was described. During this time remarkable progress has been made. Sophisticated genetic tools have been built to generate oncogene expressing cancers and characterize multiple models of solid and blood tumors. These models have led to unique insights into mechanisms of tumor initiation and progression. New drug targets have been identified, particularly through the functional analysis of cancer genomes. Now in the second decade, zebrafish cancer models are poised for even faster growth as they are used in high-throughput genetic analyses to elucidate key mechanisms underlying critical cancer phenotypes.
Collapse
Affiliation(s)
- Craig J Ceol
- Program in Molecular Medicine and Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA.
| | - Yariv Houvras
- Departments of Surgery and Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
43
|
Montalbano G, Mania M, Guerrera MC, Abbate F, Laurà R, Navarra M, Vega JA, Ciriaco E, Germanà A. Morphological differences in adipose tissue and changes in BDNF/Trkb expression in brain and gut of a diet induced obese zebrafish model. Ann Anat 2015; 204:36-44. [PMID: 26617157 DOI: 10.1016/j.aanat.2015.11.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 10/14/2015] [Accepted: 11/04/2015] [Indexed: 12/15/2022]
Abstract
Obesity is a multifactorial disease generated by an alteration in balance between energy intake and expenditure, also dependent on genetic and non-genetic factors. Moreover, various nuclei of the hypothalamus receive and process peripheral stimuli from the gastrointestinal tract, controlling food intake and therefore energy balance. Among anorexigenic molecules, brain-derived neurotrophic factor (BDNF) acts through the tyrosine-kinase receptor TrkB. Numerous data demonstrate that the BDNF/TrkB system has a fundamental role in the control of food intake and body weight. Quantitative PCR and immunohistochemistry for both BDNF and TrkB were used to determine changes in levels in the brain and gastro-intestinal tract of an experimental zebrafish model of diet-induced obesity. Overfed animals showed increased weight and body mass index as well as accumulation of adipose tissue in the visceral, subcutaneous and hepatic areas. These changes were concomitant with decreased levels of BDNF mRNA in the gastro-intestinal tract and increased expression of TrkB mRNA in the brain. Overfeeding did not change the density of cells displaying immunoreactivity for BDNF or TrkB in the brain although both were significantly diminished in the gastro-intestinal tract. These results suggest an involvement of the BDNF/TrkB system in the regulation of food intake and energy balance in zebrafish, as in mammals.
Collapse
Affiliation(s)
- Giuseppe Montalbano
- Dipartimento di Scienze Veterinarie, 98168 Messina, Italy; Zebrafish Neuromorphology Lab, Università di Messina, Polo Universitario SS. Annunziata, 98168 Messina, Italy.
| | - Manuela Mania
- Dipartimento di Scienze Veterinarie, 98168 Messina, Italy; Zebrafish Neuromorphology Lab, Università di Messina, Polo Universitario SS. Annunziata, 98168 Messina, Italy
| | - Maria Cristina Guerrera
- Dipartimento di Scienze Veterinarie, 98168 Messina, Italy; Zebrafish Neuromorphology Lab, Università di Messina, Polo Universitario SS. Annunziata, 98168 Messina, Italy
| | - Francesco Abbate
- Dipartimento di Scienze Veterinarie, 98168 Messina, Italy; Zebrafish Neuromorphology Lab, Università di Messina, Polo Universitario SS. Annunziata, 98168 Messina, Italy
| | - Rosaria Laurà
- Dipartimento di Scienze Veterinarie, 98168 Messina, Italy; Zebrafish Neuromorphology Lab, Università di Messina, Polo Universitario SS. Annunziata, 98168 Messina, Italy
| | - Michele Navarra
- Dipartimento di Scienza del Farmaco e Prodotti per la Salute, Università di Messina, Polo Universitario SS. Annunziata, 98168 Messina, Italy
| | - Jose A Vega
- Departamento de Morfologia y Biologia Celular, Facultad de Medicina, Universidad de Oviedo, Av de Julián Clavería 6, 33006 Oviedo, España; Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, 5 Poniente, 1670 Talca, Chile
| | - Emilia Ciriaco
- Dipartimento di Scienze Veterinarie, 98168 Messina, Italy; Zebrafish Neuromorphology Lab, Università di Messina, Polo Universitario SS. Annunziata, 98168 Messina, Italy
| | - Antonino Germanà
- Dipartimento di Scienze Veterinarie, 98168 Messina, Italy; Zebrafish Neuromorphology Lab, Università di Messina, Polo Universitario SS. Annunziata, 98168 Messina, Italy
| |
Collapse
|
44
|
Zang L, Shimada Y, Nishimura Y, Tanaka T, Nishimura N. Repeated Blood Collection for Blood Tests in Adult Zebrafish. J Vis Exp 2015:e53272. [PMID: 26383512 DOI: 10.3791/53272] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Repeated blood collection is one of the most common techniques performed on laboratory animals. However, a non-lethal protocol for blood collection from zebrafish has not been established. The previous methods for blood collection from zebrafish are lethal, such as lateral incision, decapitation and tail ablation. Thus we have developed a novel "repeated" blood collection method, and present here a detailed protocol outlining this procedure. This method is minimally invasive and results in a very low mortality rate (2.3%) for zebrafish, thus enabling repeated blood sampling from the same individual. The maximum volume of blood sampling is dependent on body weight of the fish. The volume for repeated blood sampling at intervals should be ≤0.4% of body weight every week or ≤1% every 2 weeks, which were evaluated by measurements of blood hemoglobin. Additionally, hemoglobin, fasting blood glucose, plasma triacylglycerol (TG) and total cholesterol levels in male and female adult zebrafish were measured. We also applied this method to investigate the dysregulation of glucose metabolism in diet-induced obesity. This blood collection method will allow many applications, including glucose and lipid metabolism and hematological studies, which will increase the use of zebrafish as a human disease model organism.
Collapse
Affiliation(s)
- Liqing Zang
- Department of Translational Medical Science, Graduate School of Medicine, Mie University;
| | - Yasuhito Shimada
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics, and Pharmacoinformatics, Graduate School of Medicine, Mie University
| | - Yuhei Nishimura
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics, and Pharmacoinformatics, Graduate School of Medicine, Mie University
| | - Toshio Tanaka
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics, and Pharmacoinformatics, Graduate School of Medicine, Mie University
| | - Norihiro Nishimura
- Department of Translational Medical Science, Graduate School of Medicine, Mie University
| |
Collapse
|
45
|
Zhao S, Huang J, Ye J. A fresh look at zebrafish from the perspective of cancer research. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:80. [PMID: 26260237 PMCID: PMC4531851 DOI: 10.1186/s13046-015-0196-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 07/28/2015] [Indexed: 12/31/2022]
Abstract
Zebrafish represent a vertebrate model organism that has been widely, and increasingly, employed over the last decade in the study of developmental processes, wound healing, microbe-host interactions, and drug screening. With the increase in the laboratory use of zebrafish, several advantages, such as a high genetic homology to humans and transparent embryos, which allow clear disease evaluation, have greatly widened its use as a model for studying tumor development in vivo. The use of zebrafish has been applied in several areas of cancer research, mainly in the following domains: (1) establishing cancer models by carcinogenic chemical, genetic technology, and xenotransplantation; (2) evaluating tumor angiogenesis; (3) studying tumor metastasis; and (4) anti-tumor drug screening and drug toxicity evaluation. In this study, we provide a comprehensive overview of the role of zebrafish in order to underline the advantages of using them as a model organism in cancer research. Several related successful events are also reviewed.
Collapse
Affiliation(s)
- Shuai Zhao
- Department of Surgical oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jian Huang
- Department of Surgical oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China.
| | - Jun Ye
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
46
|
The promise of zebrafish as a chemical screening tool in cancer therapy. Future Med Chem 2015; 7:1395-405. [DOI: 10.4155/fmc.15.73] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Cancer progression in zebrafish recapitulates many aspects of human cancer and as a result, zebrafish have been gaining popularity for their potential use in basic and translational cancer research. Human cancer can be modeled in zebrafish by induction using chemical mutagens, xenotransplantation or by genetic manipulation. Chemical screens based on zebrafish cancer models offer a rapid, powerful and inexpensive means of evaluating the potential of suppression or prevention on cancer. The identification of small molecules through such screens will serve as ideal entry points for novel chemical therapies for cancer treatment. This article outlines advances that have been made within the growing field of zebrafish cancer models and presents their advantages for chemical drug screening.
Collapse
|
47
|
Cross-species oncogenomics using zebrafish models of cancer. Curr Opin Genet Dev 2015; 30:73-9. [PMID: 26070506 DOI: 10.1016/j.gde.2015.04.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 04/23/2015] [Indexed: 12/11/2022]
Abstract
The zebrafish is a relatively recent addition to cancer modeling. These models have now been extensively used in cross-species oncogenomic analyses at both the DNA and RNA levels. The goal of such studies is to identify conserved events that occur in both human and fish tumors which may act as central drivers of tumor phenotypes. Numerous comparisons of somatic DNA changes, using array CGH and exome sequencing, have demonstrated a relatively small set of conserved changes across species. In contrast, striking conservation of RNA expression patterns have been observed between the two species in models such as melanoma, leukemia, and rhabdomyosarcoma. In the future, the zebrafish will increasingly be used to model epigenetic and noncoding aspects of cancer biology.
Collapse
|
48
|
Long-term hyperphagia and caloric restriction caused by low- or high-density husbandry have differential effects on zebrafish postembryonic development, somatic growth, fat accumulation and reproduction. PLoS One 2015; 10:e0120776. [PMID: 25799180 PMCID: PMC4370574 DOI: 10.1371/journal.pone.0120776] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 01/29/2015] [Indexed: 12/19/2022] Open
Abstract
In recent years, the zebrafish (Danio rerio) has emerged as an alternative vertebrate model for energy homeostasis and metabolic diseases, including obesity and anorexia. It has been shown that diet-induced obesity (DIO) in zebrafish shares multiple pathophysiological features with obesity in mammals. However, a systematic and comprehensive analysis of the different pathways of energy expenditure in obese and starved fish had been missing thus far. Here, we carry out long-term ad libitum feeding (hyperphagia) and caloric restriction studies induced by low- or high-density husbandry, respectively, to investigate the impact of caloric intake on the timing of scale formation, a crucial step of postembryonic development and metamorphosis, and on somatic growth, body weight, fat storage and female reproduction. We show that all of them are positively affected by increased caloric intake, that middle-aged fish develop severe DIO, and that the body mass index (BMI) displays a strict linear correlation with whole-body triglyceride levels in adult zebrafish. Interestingly, juvenile fish are largely resistant to DIO, while BMI and triglyceride values drop in aged fish, pointing to aging-associated anorexic effects. Histological analyses further indicate that increased fat storage in white adipose tissue involves both hyperplasia and hypertrophy of adipocytes. Furthermore, in ovaries, caloric intake primarily affects the rate of oocyte growth, rather than total oocyte numbers. Finally, comparing the different pathways of energy expenditure with each other, we demonstrate that they are differentially affected by caloric restriction / high-density husbandry. In juvenile fish, scale formation is prioritized over somatic growth, while in sexually mature adults, female reproduction is prioritized over somatic growth, and somatic growth over fat storage. Our data will serve as a template for future functional studies to dissect the neuroendocrine regulators of energy homeostasis mediating differential energy allocation.
Collapse
|
49
|
Meguro S, Hasumura T, Hase T. Body fat accumulation in zebrafish is induced by a diet rich in fat and reduced by supplementation with green tea extract. PLoS One 2015; 10:e0120142. [PMID: 25785691 PMCID: PMC4364667 DOI: 10.1371/journal.pone.0120142] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 01/19/2015] [Indexed: 01/21/2023] Open
Abstract
Fat-rich diets not only induce obesity in humans but also make animals obese. Therefore, animals that accumulate body fat in response to a high-fat diet (especially rodents) are commonly used in obesity research. The effect of dietary fat on body fat accumulation is not fully understood in zebrafish, an excellent model of vertebrate lipid metabolism. Here, we explored the effects of dietary fat and green tea extract, which has anti-obesity properties, on body fat accumulation in zebrafish. Adult zebrafish were allocated to four diet groups and over 6 weeks were fed a high-fat diet containing basal diet plus two types of fat or a low-fat diet containing basal diet plus carbohydrate or protein. Another group of adult zebrafish was fed a high-fat diet with or without 5% green tea extract supplementation. Zebrafish fed the high-fat diets had nearly twice the body fat (visceral, subcutaneous, and total fat) volume and body fat volume ratio (body fat volume/body weight) of those fed low-fat diets. There were no differences in body fat accumulation between the two high-fat groups, nor were there any differences between the two low-fat groups. Adding green tea extract to the high-fat diet significantly suppressed body weight, body fat volume, and body fat volume ratio compared with the same diet lacking green tea extract. 3-Hydroxyacyl-coenzyme A dehydrogenase and citrate synthase activity in the liver and skeletal muscle were significantly higher in fish fed the diet supplemented with green tea extract than in those fed the unsupplemented diet. Our results suggest that a diet rich in fat, instead of protein or carbohydrate, induced body fat accumulation in zebrafish with mechanisms that might be similar to those in mammals. Consequently, zebrafish might serve as a good animal model for research into obesity induced by high-fat diets.
Collapse
Affiliation(s)
- Shinichi Meguro
- Biological Science Research, Kao Corporation, Haga-gun, Tochigi, Japan
- * E-mail:
| | - Takahiro Hasumura
- Biological Science Research, Kao Corporation, Haga-gun, Tochigi, Japan
| | - Tadashi Hase
- Biological Science Research, Kao Corporation, Haga-gun, Tochigi, Japan
| |
Collapse
|
50
|
Otis JP, Zeituni EM, Thierer JH, Anderson JL, Brown AC, Boehm ED, Cerchione DM, Ceasrine AM, Avraham-Davidi I, Tempelhof H, Yaniv K, Farber SA. Zebrafish as a model for apolipoprotein biology: comprehensive expression analysis and a role for ApoA-IV in regulating food intake. Dis Model Mech 2015; 8:295-309. [PMID: 25633982 PMCID: PMC4348566 DOI: 10.1242/dmm.018754] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 01/22/2015] [Indexed: 12/31/2022] Open
Abstract
Improved understanding of lipoproteins, particles that transport lipids throughout the circulation, is vital to developing new treatments for the dyslipidemias associated with metabolic syndrome. Apolipoproteins are a key component of lipoproteins. Apolipoproteins are proteins that structure lipoproteins and regulate lipid metabolism through control of cellular lipid exchange. Constraints of cell culture and mouse models mean that there is a need for a complementary model that can replicate the complex in vivo milieu that regulates apolipoprotein and lipoprotein biology. Here, we further establish the utility of the genetically tractable and optically clear larval zebrafish as a model of apolipoprotein biology. Gene ancestry analyses were implemented to determine the closest human orthologs of the zebrafish apolipoprotein A-I (apoA-I), apoB, apoE and apoA-IV genes and therefore ensure that they have been correctly named. Their expression patterns throughout development were also analyzed, by whole-mount mRNA in situ hybridization (ISH). The ISH results emphasized the importance of apolipoproteins in transporting yolk and dietary lipids: mRNA expression of all apolipoproteins was observed in the yolk syncytial layer, and intestinal and liver expression was observed from 4-6 days post-fertilization (dpf). Furthermore, real-time PCR confirmed that transcription of three of the four zebrafish apoA-IV genes was increased 4 hours after the onset of a 1-hour high-fat feed. Therefore, we tested the hypothesis that zebrafish ApoA-IV performs a conserved role to that in rat in the regulation of food intake by transiently overexpressing ApoA-IVb.1 in transgenic larvae and quantifying ingestion of co-fed fluorescently labeled fatty acid during a high-fat meal as an indicator of food intake. Indeed, ApoA-IVb.1 overexpression decreased food intake by approximately one-third. This study comprehensively describes the expression and function of eleven zebrafish apolipoproteins and serves as a springboard for future investigations to elucidate their roles in development and disease in the larval zebrafish model.
Collapse
Affiliation(s)
- Jessica P Otis
- Carnegie Institution for Science, Department of Embryology, Baltimore, MD 21218, USA
| | - Erin M Zeituni
- Carnegie Institution for Science, Department of Embryology, Baltimore, MD 21218, USA
| | - James H Thierer
- Carnegie Institution for Science, Department of Embryology, Baltimore, MD 21218, USA Johns Hopkins University, Department of Biology, Baltimore, MD 21218, USA
| | - Jennifer L Anderson
- Carnegie Institution for Science, Department of Embryology, Baltimore, MD 21218, USA
| | - Alexandria C Brown
- Carnegie Institution for Science, Department of Embryology, Baltimore, MD 21218, USA
| | - Erica D Boehm
- Carnegie Institution for Science, Department of Embryology, Baltimore, MD 21218, USA Johns Hopkins University, Department of Biology, Baltimore, MD 21218, USA
| | - Derek M Cerchione
- Carnegie Institution for Science, Department of Embryology, Baltimore, MD 21218, USA Johns Hopkins University, Department of Biology, Baltimore, MD 21218, USA
| | - Alexis M Ceasrine
- Carnegie Institution for Science, Department of Embryology, Baltimore, MD 21218, USA Johns Hopkins University, Department of Biology, Baltimore, MD 21218, USA
| | - Inbal Avraham-Davidi
- Weizmann Institute of Science, Department of Biological Regulation, Rehovot 7610001, Israel
| | - Hanoch Tempelhof
- Weizmann Institute of Science, Department of Biological Regulation, Rehovot 7610001, Israel
| | - Karina Yaniv
- Weizmann Institute of Science, Department of Biological Regulation, Rehovot 7610001, Israel
| | - Steven A Farber
- Carnegie Institution for Science, Department of Embryology, Baltimore, MD 21218, USA Johns Hopkins University, Department of Biology, Baltimore, MD 21218, USA
| |
Collapse
|