1
|
Xu T, Liu F, He J, Xu P, Qu J, Wang H, Yue J, Yang Q, Wu W, Zeng G, Sun D, Chen X. Leveraging zebrafish models for advancing radiobiology: Mechanisms, applications, and future prospects in radiation exposure research. ENVIRONMENTAL RESEARCH 2024; 266:120504. [PMID: 39638026 DOI: 10.1016/j.envres.2024.120504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/12/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
Ionizing radiation (IR) represents a significant risk to human health and societal stability. To effectively analyze the mechanisms of IR and enhance protective strategies, the development of more sophisticated animal models is imperative. The zebrafish, with its high degree of genomic homology to humans and the capacity for whole-body optical visualization and high-throughput screening, represents an invaluable model for the study of IR. This review examines the benefits of utilizing zebrafish as a model organism for research on IR, emphasizing recent advancements and applications. It presents a comprehensive overview of the methodologies for establishing IR models in zebrafish, addresses current challenges, and discusses future development trends. This paper provide theoretical support for elucidating the mechanisms of IR injury and developing effective treatment strategies.
Collapse
Affiliation(s)
- Ting Xu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China; Department of Endocrinology, Yiwu Central Hospital, The Affiliated Yiwu Hospital of Wenzhou Medical University, Yiwu, 322000, China
| | - Fan Liu
- State and Local Joint Engineering Research Center for Ecological Treatment Technology of Urban Water Pollution, School of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Jiaxuan He
- State and Local Joint Engineering Research Center for Ecological Treatment Technology of Urban Water Pollution, School of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Peiye Xu
- State and Local Joint Engineering Research Center for Ecological Treatment Technology of Urban Water Pollution, School of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Junying Qu
- State and Local Joint Engineering Research Center for Ecological Treatment Technology of Urban Water Pollution, School of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China; Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, School of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Hanbing Wang
- Department of Biotechnology, The University of Hong Kong, Hong Kong SAR, 999077, China
| | - Jinghui Yue
- Nuclear Power Institute of China, Chengdu, 610200, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Wei Wu
- Bioengineering College, Chongqing University, Chongqing, 400044, China
| | - Guoming Zeng
- Intelligent Construction Technology Application Service Center, School of Architecture and Engineering, Chongqing City Vocational College, Chongqing, 402160, China
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China; Department of Endocrinology, Yiwu Central Hospital, The Affiliated Yiwu Hospital of Wenzhou Medical University, Yiwu, 322000, China; Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, School of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China.
| | - Xia Chen
- Department of Endocrinology, Yiwu Central Hospital, The Affiliated Yiwu Hospital of Wenzhou Medical University, Yiwu, 322000, China.
| |
Collapse
|
2
|
Wang H, Liu J, Qiang S, Che Y, Hu T. 4-tert-Butylphenol impairs the liver by inducing excess liver lipid accumulation via disrupting the lipid metabolism pathway in zebrafish. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 356:124385. [PMID: 38897274 DOI: 10.1016/j.envpol.2024.124385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/04/2024] [Accepted: 06/16/2024] [Indexed: 06/21/2024]
Abstract
Endocrine disrupting chemicals (EDCs) can disrupt normal endocrine function by interfering with the synthesis and release of hormones, causing adverse reactions to development, immunity, nerves, and reproduction. 4-tert-Butylphenol (4-t-BP) is disruptive to early zebrafish development, but its effects on zebrafish liver are unknown. In this study, the adverse effects of 4-t-BP on the liver were investigated using zebrafish as a model organism. 4-t-BP inhibited liver development in zebrafish embryos and induced liver damage in adult zebrafish. Even if F1 was not directly exposed to 4-t-BP, its growth and development were inhibited. 4-t-BP can lead to an increase in lipid accumulation, total cholesterol and triglycerides contents, and the activities of alanine transaminase and aspartate aminotransferase in zebrafish embryos and adult zebrafish livers, and also cause an acceleration of glucose metabolism in zebrafish embryos. In addition, qRT-PCR showed that 4-t-BP induced the changes in the expressions of liver development-, steroid and unsaturated fatty acid biosynthesis-, and glycerolipid and arachidonic acid metabolism-related genes in zebrafish embryos and inflammatory factors-, antioxidant enzymes- and lipid metabolism-related genes in adult zebrafish livers. Transcriptome sequencing of embryos showed that 4-t-BP altered the expressions of lipid metabolism pathways such as steroid and unsaturated fatty acid biosynthesis, glycerolipid, and arachidonic acid metabolism pathways. Therefore, 4-t-BP may be external stimuli that cause oxidative stress, inflammation, and lipid accumulation in zebrafish liver, resulting in tissue damage and dysfunction in zebrafish liver.
Collapse
Affiliation(s)
- Huiyun Wang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Juan Liu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Shuting Qiang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Yufeng Che
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Tingzhang Hu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China.
| |
Collapse
|
3
|
Zhang P, Cao J, Liang X, Su Z, Zhang B, Wang Z, Xie J, Chen G, Chen X, Zhang J, Feng Y, Xu Q, Song J, Hong A, Chen X, Zhang Y. Lian-Mei-Yin formula alleviates diet-induced hepatic steatosis by suppressing Yap1/FOXM1 pathway-dependent lipid synthesis. Acta Biochim Biophys Sin (Shanghai) 2024; 56:621-633. [PMID: 38516704 DOI: 10.3724/abbs.2024025] [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] [Indexed: 03/23/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, with a global prevalence of 25%. Patients with NAFLD are more likely to suffer from advanced liver disease, cardiovascular disease, or type II diabetes. However, unfortunately, there is still a shortage of FDA-approved therapeutic agents for NAFLD. Lian-Mei-Yin (LMY) is a traditional Chinese medicine formula used for decades to treat liver disorders. It has recently been applied to type II diabetes which is closely related to insulin resistance. Given that NAFLD is another disease involved in insulin resistance, we hypothesize that LMY might be a promising formula for NAFLD therapy. Herein, we verify that the LMY formula effectively reduces hepatic steatosis in diet-induced zebrafish and NAFLD model mice in a time- and dose-dependent manner. Mechanistically, LMY suppresses Yap1-mediated Foxm1 activation, which is crucial for the occurrence and development of NAFLD. Consequently, lipogenesis is ameliorated by LMY administration. In summary, the LMY formula alleviates diet-induced NAFLD in zebrafish and mice by inhibiting Yap1/Foxm1 signaling-mediated NAFLD pathology.
Collapse
Affiliation(s)
- Peiguang Zhang
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| | - Jieqiong Cao
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| | - Xujing Liang
- Department of Infectious Disease, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Zijian Su
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| | - Bihui Zhang
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| | - Zhenyu Wang
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| | - Junye Xie
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| | - Gengrui Chen
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Xue Chen
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| | - Jinting Zhang
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| | - Yanxian Feng
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Qin Xu
- Guangzhou University of Traditional Chinese Medicine, Guangzhou 510006, China
| | - Jianping Song
- Guangzhou University of Traditional Chinese Medicine, Guangzhou 510006, China
| | - An Hong
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| | - Xiaojia Chen
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| | - Yibo Zhang
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| |
Collapse
|
4
|
Tompach MC, Gridley CK, Li S, Clark JM, Park Y, Timme-Laragy AR. Comparing the effects of developmental exposure to alpha lipoic acid (ALA) and perfluorooctanesulfonic acid (PFOS) in zebrafish (Danio rerio). Food Chem Toxicol 2024; 186:114560. [PMID: 38432440 PMCID: PMC11034762 DOI: 10.1016/j.fct.2024.114560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 02/14/2024] [Accepted: 02/29/2024] [Indexed: 03/05/2024]
Abstract
Alpha lipoic acid (ALA) is a dietary supplement that has been used to treat a wide range of diseases, including obesity and diabetes, and have lipid-lowering effects, making it a potential candidate for mitigating dyslipidemia resulting from exposures to the per- and polyfluoroalkyl substance (PFAS) family member perfluorooctanesulfonic acid (PFOS). ALA can be considered a non-fluorinated structural analog to PFOS due to their similar 8-carbon chain and amphipathic structure, but, unlike PFOS, is rapidly metabolized. PFOS has been shown to reduce pancreatic islet area and induce β-cell lipotoxicity, indicating that changes in β-cell lipid microenvironment is a mechanism contributing to hypomorphic islets. Due to structural similarities, we hypothesized that ALA may compete with PFOS for binding to proteins and distribution throughout the body to mitigate the effects of PFOS exposure. However, ALA alone reduced islet area and fish length, with several morphological endpoints indicating additive toxicity in the co-exposures. Individually, ALA and PFOS increased fatty acid uptake from the yolk. ALA alone increased liver lipid accumulation, altered fatty acid profiling and modulated PPARɣ pathway signaling. Together, this work demonstrates that ALA and PFOS have similar effects on lipid uptake and metabolism during embryonic development in zebrafish.
Collapse
Affiliation(s)
- Madeline C Tompach
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, Amherst, MA, 01003, USA; Biotechnology Training Program, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Charlotte K Gridley
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Sida Li
- Department of Food Science, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - John M Clark
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Yeonhwa Park
- Department of Food Science, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Alicia R Timme-Laragy
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, Amherst, MA, 01003, USA; Department of Environmental Health Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA.
| |
Collapse
|
5
|
van Mever M, Mamani-Huanca M, Faught E, López-Gonzálvez Á, Hankemeier T, Barbas C, Schaaf MJM, Ramautar R. Application of a capillary electrophoresis-mass spectrometry metabolomics workflow in zebrafish larvae reveals new effects of cortisol. Electrophoresis 2024; 45:380-391. [PMID: 38072651 DOI: 10.1002/elps.202300186] [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: 08/21/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 03/20/2024]
Abstract
In contemporary biomedical research, the zebrafish (Danio rerio) is increasingly considered a model system, as zebrafish embryos and larvae can (potentially) fill the gap between cultured cells and mammalian animal models, because they can be obtained in large numbers, are small and can easily be manipulated genetically. Given that capillary electrophoresis-mass spectrometry (CE-MS) is a useful analytical separation technique for the analysis of polar ionogenic metabolites in biomass-limited samples, the aim of this study was to develop and assess a CE-MS-based analytical workflow for the profiling of (endogenous) metabolites in extracts from individual zebrafish larvae and pools of small numbers of larvae. The developed CE-MS workflow was used to profile metabolites in extracts from pools of 1, 2, 4, 8, 12, 16, 20, and 40 zebrafish larvae. For six selected endogenous metabolites, a linear response (R2 > 0.98) for peak areas was obtained in extracts from these pools. The repeatability was satisfactory, with inter-day relative standard deviation values for peak area of 9.4%-17.7% for biological replicates (n = 3 over 3 days). Furthermore, the method allowed the analysis of over 70 endogenous metabolites in a pool of 12 zebrafish larvae, and 29 endogenous metabolites in an extract from only 1 zebrafish larva. Finally, we applied the optimized CE-MS workflow to identify potential novel targets of the mineralocorticoid receptor in mediating the effects of cortisol.
Collapse
Affiliation(s)
- Marlien van Mever
- Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| | - Maricruz Mamani-Huanca
- Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Madrid, Spain
| | - Erin Faught
- Institute of Biology Leiden (IBL), Leiden University, Leiden, The Netherlands
| | - Ángeles López-Gonzálvez
- Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Madrid, Spain
| | - Thomas Hankemeier
- Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| | - Coral Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Madrid, Spain
| | - Marcel J M Schaaf
- Institute of Biology Leiden (IBL), Leiden University, Leiden, The Netherlands
| | - Rawi Ramautar
- Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| |
Collapse
|
6
|
Konadu B, Cox CK, Garrett MR, Gibert Y. Excess glucose or fat differentially affects metabolism and appetite-related gene expression during zebrafish embryogenesis. iScience 2023; 26:107063. [PMID: 37534154 PMCID: PMC10391732 DOI: 10.1016/j.isci.2023.107063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 03/28/2023] [Accepted: 06/02/2023] [Indexed: 08/04/2023] Open
Abstract
Zebrafish embryos use their yolk sac reserve as the sole nutrient source during embryogenesis. The two main forms of energy fuel can be found in the form of glucose or fat. Zebrafish embryos were exposed to glucose or injected with free fatty acid/Triacylglycerol (FFA/TAG) into the yolk sac at 24 hpf. At 72 hpf, glucose exposed or FFA/TAG injected had differential effects on gene expression in embryos, with fat activating lipolysis and β-oxidation and glucose activating the insulin pathway. Bulk RNA-seq revealed that more gene expression was affected by glucose exposure compared to FFA/TAGs injection. Appetite-controlling genes were also differently affected by glucose exposure or FFA/TAG injections. Because the embryo did not yet feed itself at the time of our analysis, gene expression changes occurred in absence of actual hunger and revealed how the embryo manages its nutrient intake before active feeding.
Collapse
Affiliation(s)
- Bridget Konadu
- Department of Cell and Molecular Biology, Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Carol K. Cox
- Department of Cell and Molecular Biology, Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Michael R. Garrett
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Yann Gibert
- Department of Cell and Molecular Biology, Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
7
|
Yehuda H, Madrer N, Goldberg D, Soreq H, Meerson A. Inversely Regulated Inflammation-Related Processes Mediate Anxiety-Obesity Links in Zebrafish Larvae and Adults. Cells 2023; 12:1794. [PMID: 37443828 PMCID: PMC10341043 DOI: 10.3390/cells12131794] [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: 06/14/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Anxiety and metabolic impairments are often inter-related, but the underlying mechanisms are unknown. To seek RNAs involved in the anxiety disorder-metabolic disorder link, we subjected zebrafish larvae to caffeine-induced anxiety or high-fat diet (HFD)-induced obesity followed by RNA sequencing and analyses. Notably, differentially expressed (DE) transcripts in these larval models and an adult zebrafish caffeine-induced anxiety model, as well as the transcript profiles of inherently anxious versus less anxious zebrafish strains and high-fat diet-fed versus standard diet-fed adult zebrafish, revealed inversely regulated DE transcripts. In both larval anxiety and obesity models, these included long noncoding RNAs and transfer RNA fragments, with the overrepresented immune system and inflammation pathways, e.g., the "interleukin signaling pathway" and "inflammation mediated by chemokine and cytokine signaling pathway". In adulthood, overrepresented immune system processes included "T cell activation", "leukocyte cell-cell adhesion", and "antigen processing and presentation". Furthermore, unlike adult zebrafish, obesity in larvae was not accompanied by anxiety-like behavior. Together, these results may reflect an antagonistic pleiotropic phenomenon involving a re-adjusted modulation of the anxiety-metabolic links with an occurrence of the acquired immune system. Furthermore, the HFD potential to normalize anxiety-upregulated immune-related genes may reflect the high-fat diet protection of anxiety and neurodegeneration reported by others.
Collapse
Affiliation(s)
- Hila Yehuda
- MIGAL—Galilee Research Institute, Kiryat Shmona 11016, Israel
- The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; (H.Y.); (N.M.)
| | - Nimrod Madrer
- The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; (H.Y.); (N.M.)
- The Edmond and Lily Safra Center for Brain Science, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Doron Goldberg
- MIGAL—Galilee Research Institute, Kiryat Shmona 11016, Israel
- Tel-Hai College, Upper Galilee 1220800, Israel;
| | - Hermona Soreq
- The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; (H.Y.); (N.M.)
- The Edmond and Lily Safra Center for Brain Science, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Ari Meerson
- MIGAL—Galilee Research Institute, Kiryat Shmona 11016, Israel
- Tel-Hai College, Upper Galilee 1220800, Israel;
| |
Collapse
|
8
|
Yan Z, Cao X, Sun S, Sun B, Gao J. Inhibition of GSK3B phosphorylation improves glucose and lipid metabolism disorder. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166726. [PMID: 37146915 DOI: 10.1016/j.bbadis.2023.166726] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 03/31/2023] [Accepted: 04/21/2023] [Indexed: 05/07/2023]
Abstract
Hepatic glycolipid metabolism disorder is considered as one of the key pathogenic factors for many chronic diseases. Revealing the molecular mechanism of metabolic disorder and exploring drug targets are crucial for the treatment of glucose and lipid metabolic diseases. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) has been reported to be associated with the pathogenesis of various metabolic diseases. Herein, GAPDH-knockdown ZFL cells and GAPDH-downregulation zebrafish exhibited significant lipid deposition increase and glycogen reduction, thus inducing glucose and lipid metabolism disorders. Using high-sensitivity mass spectrometry-based proteomic and phosphoproteomic analysis, we identified 6838 proteins and 3738 phosphorylated proteins in GAPDH-knockdown ZFL cells. The protein-protein interaction network and DEPPs analyses showed that gsk3baY216 were involved in lipid and glucose metabolism, which was verified by In vitro study. The enzyme activity analysis and cell staining results showed that HepG2 and NCTC-1469 cells transfected with GSK3BY216F plasmid had significantly lower glucose and insulin levels, the decreased lipid deposition, and the increased glycogen synthesis than those transfected with GSK3BY216E plasmid, suggesting that inhibition of GSK3B phosphorylation could significantly improve GSK3B hyperphosphorylation-induced glucose tolerance impairment and insulin sensitivity reduction. To our knowledge, this is the first multi-omic study of GAPDH-knockdown ZFL cells. This study provides insights into the molecular mechanism of glucose and lipid metabolic disorder, and provides potential targets (kinases) for the treatments of human glucose and lipid metabolic diseases.
Collapse
Affiliation(s)
- Ze Yan
- College of Fisheries, Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaojuan Cao
- College of Fisheries, Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China
| | - Shouxiang Sun
- College of Fisheries, Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Bing Sun
- College of Fisheries, Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Jian Gao
- College of Fisheries, Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
9
|
Kulkarni A, Muralidharan C, May SC, Tersey SA, Mirmira RG. Inside the β Cell: Molecular Stress Response Pathways in Diabetes Pathogenesis. Endocrinology 2022; 164:bqac184. [PMID: 36317483 PMCID: PMC9667558 DOI: 10.1210/endocr/bqac184] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Indexed: 11/05/2022]
Abstract
The pathogeneses of the 2 major forms of diabetes, type 1 and type 2, differ with respect to their major molecular insults (loss of immune tolerance and onset of tissue insulin resistance, respectively). However, evidence suggests that dysfunction and/or death of insulin-producing β-cells is common to virtually all forms of diabetes. Although the mechanisms underlying β-cell dysfunction remain incompletely characterized, recent years have witnessed major advances in our understanding of the molecular pathways that contribute to the demise of the β-cell. Cellular and environmental factors contribute to β-cell dysfunction/loss through the activation of molecular pathways that exacerbate endoplasmic reticulum stress, the integrated stress response, oxidative stress, and impaired autophagy. Whereas many of these stress responsive pathways are interconnected, their individual contributions to glucose homeostasis and β-cell health have been elucidated through the development and interrogation of animal models. In these studies, genetic models and pharmacological compounds have enabled the identification of genes and proteins specifically involved in β-cell dysfunction during diabetes pathogenesis. Here, we review the critical stress response pathways that are activated in β cells in the context of the animal models.
Collapse
Affiliation(s)
- Abhishek Kulkarni
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Charanya Muralidharan
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Sarah C May
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Sarah A Tersey
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Raghavendra G Mirmira
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| |
Collapse
|
10
|
Chowdhury S, Saikia SK. Use of Zebrafish as a Model Organism to Study Oxidative Stress: A Review. Zebrafish 2022; 19:165-176. [PMID: 36049069 DOI: 10.1089/zeb.2021.0083] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Abstract
Dioxygen is an integral part of every living organism, but its concentration varies from organ to organ. Production of metabolites from dioxygen may result in oxidative stress. Since oxidative stress has the potential to damage various biomolecules in the cell, therefore, it has presently become an active field of research. Oxidative stress has been studied in a wide range of model organisms from vertebrates to invertebrates, from rodents to piscine organisms, and from in vivo to in vitro models. But zebrafish (adults, larvae, or embryonic stage) emerged out to be the most promising vertebrate model organism to study oxidative stress because of its vast advantages (transparent embryo, cost-effectiveness, similarity to human genome, easy developmental processes, numerous offspring per spawning, and many more). This is evidenced by voluminous number of researches on oxidative stress in zebrafish exposed to chemicals, radiations, nanoparticles, pesticides, heavy metals, etc. On these backgrounds, this review attempts to highlight the potentiality of zebrafish as model of oxidative stress compared with other companion models. Several areas, from biomedical to environmental research, have been covered to explain it as a more convenient and reliable animal model for experimental research on oxidative mechanisms.
Collapse
Affiliation(s)
- Sabarna Chowdhury
- Aquatic Ecology and Fish Biology Laboratory, Department of Zoology, Visva-Bharati University, Santiniketan, West Bengal, India
| | - Surjya Kumar Saikia
- Aquatic Ecology and Fish Biology Laboratory, Department of Zoology, Visva-Bharati University, Santiniketan, West Bengal, India
| |
Collapse
|
11
|
An Association between Insulin Resistance and Neurodegeneration in Zebrafish Larval Model ( Danio rerio). Int J Mol Sci 2022; 23:ijms23158290. [PMID: 35955446 PMCID: PMC9368350 DOI: 10.3390/ijms23158290] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/19/2022] [Accepted: 07/22/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Type 2 diabetes mellitus has recently been identified as a mediator of neurodegeneration. However, the molecular mechanisms have not been clearly elucidated. We aimed to investigate insulin resistance associated with neurodegenerative events in zebrafish larvae. Methods: Larvae aged 72 h-post-fertilization (hpf) were induced to insulin resistance by immersion in 250 nM insulin and were then reinduced with 100 nM insulin at 96 hpf. This model was validated by a glucose levels assay, qPCR analysis of selected genes (akt, pepck, zglut3 and claudin-5a) and Oil Red-O (ORO) staining of the yolk sac for lipid distribution. The association of insulin resistance and neurodegeneration was validated by malondialdehyde (MDA), glutathione (GSH) assays, and by integrating next-generation sequencing with database for annotation, visualization and integrated discovery (DAVID). Results: There was a significant increase in glucose levels at 180 min in the insulin-resistant group. However, it decreased at 400 min after the re-challenge. Insulin-signaling mediators, akt and pepck, were showed significantly downregulated up to 400 min after insulin immersion (p < 0.05). Meanwhile, claudin-5a assessed blood−brain barrier (BBB) integrity and showed significant deterioration after 400 min of post-insulin immersion. ORO staining remarked the increase in yolk sac size in the insulin-resistant group. After the confirmation of insulin resistance, MDA levels increased significantly in the insulin-resistant group compared to the control group in the following parameters. Furthermore, dysregulated MAPK- and Wnt/Ca2+-signaling pathways were observed in the insulin-resistant group, disrupting energy metabolism and causing BBB injury. Conclusions: We conclude that the insulin-resistant zebrafish larvae alter the metabolic physiology associated with neurodegeneration.
Collapse
|
12
|
Zhong X, Gu J, Zhang S, Chen X, Zhang J, Miao J, Ding Z, Xu J, Cheng H. Dynamic transcriptome analysis of the muscles in high-fat diet-induced obese zebrafish (Danio rerio) under 5-HT treatment. Gene 2022; 819:146265. [PMID: 35121026 DOI: 10.1016/j.gene.2022.146265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/18/2022] [Accepted: 01/27/2022] [Indexed: 11/04/2022]
Abstract
Peripheral 5-hydroxytryptamine (5-HT, also called serotonin) is reportedly a potential therapeutic target in obesity-related metabolic diseases due to its regulatory role in energy homeostasis in mammals. However, information on the detailed effect of peripheral 5-HT on the energy metabolism in fishes, especially the lipid metabolism, and the underlying mechanism remains elusive. In this study, a diet-induced obesity model was developed in the zebrafish (Danio rerio), a prototypical animal model for metabolic disorders. The zebrafish were fed a high-fat diet for 8 weeks and were simultaneously injected with PBS, 0.1 mM and 10 mM 5-HT, intraperitoneally. The body weight was significantly lower in the zebrafish injected with 0.1 mM 5-HT (P < 0.05), however, there was no change in body length (P > 0.05) at the end of the 8-week treatment. The muscle tissues from the zebrafish treated with PBS and 5-HT were collected for transcriptomic analysis and the RNA-seq revealed 1134, 3713, and 2535 genes were screened out compared to the muscular DEGs among three groups. The enrichment analysis revealed DEGs to be significantly associated with multiple metabolic pathways, including ribosome, oxidative phosphorylation, proteasome, PPAR signaling pathway, and ferroptosis. Additionally, the qRT-PCR validated 12 DEGs out of which 10 genes exhibited consistent trends. Taken together, this data provided useful information on the transcriptional characteristics of the muscle tissue in the obese zebrafish exposed to 5-HT, offering important insights into the regulatory effect of peripheral 5-HT in teleosts, as well as novel approaches for preventing and treating obesity-related metabolic dysfunction.
Collapse
Affiliation(s)
- Xiangqi Zhong
- Jiangsu Key Laboratory of Marine Biotechnology/Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; Key Laboratory of Cultivation and High-value Utilization of Marine Organisms, Fisheries Research Institute of Fujian, Xiamen 361000, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
| | - Jiaze Gu
- Jiangsu Key Laboratory of Marine Biotechnology/Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| | - Siying Zhang
- Jiangsu Key Laboratory of Marine Biotechnology/Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xiangning Chen
- Jiangsu Key Laboratory of Marine Biotechnology/Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; Key Laboratory of Cultivation and High-value Utilization of Marine Organisms, Fisheries Research Institute of Fujian, Xiamen 361000, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China.
| | - Jingjing Zhang
- Jiangsu Key Laboratory of Marine Biotechnology/Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China
| | - Jintao Miao
- Jiangsu Key Laboratory of Marine Biotechnology/Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China
| | - Zhujin Ding
- Jiangsu Key Laboratory of Marine Biotechnology/Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
| | - Jianhe Xu
- Jiangsu Key Laboratory of Marine Biotechnology/Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
| | - Hanliang Cheng
- Jiangsu Key Laboratory of Marine Biotechnology/Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
| |
Collapse
|
13
|
Vasyutina M, Alieva A, Reutova O, Bakaleiko V, Murashova L, Dyachuk V, Catapano AL, Baragetti A, Magni P. The zebrafish model system for dyslipidemia and atherosclerosis research: Focus on environmental/exposome factors and genetic mechanisms. Metabolism 2022; 129:155138. [PMID: 35051509 DOI: 10.1016/j.metabol.2022.155138] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/15/2021] [Accepted: 01/13/2022] [Indexed: 12/13/2022]
Abstract
Dyslipidemias and atherosclerosis play a pivotal role in cardiovascular risk and disease. Although some pathophysiological mechanisms underlying these conditions have been unveiled, several knowledge gaps still remain. Experimental models, both in vitro and in vivo, have been instrumental to our better understanding of such complex processes. The latter have often been based on rodent species, either wild-type or, in several instances, genetically modified. In this context, the zebrafish may represent an additional very useful in vivo experimental model for dyslipidemia and atherosclerosis. Interestingly, the lipid metabolism of zebrafish shares several features with that present in humans, recapitulating some molecular features and pathophysiological aspects in a better way than that of rodents. The zebrafish model may be of help to address questions related to exposome factors as well as to genetic features, aiming to dissect selected aspects of the more complex scenario observed in humans. Indeed, exposome-related dyslipidemia/atherosclerosis research in zebrafish may target different scientific questions, related to nutrition, microbiota, temperature, light exposure at the larval stage, exposure to chemicals and epigenetic consequences of such external factors. Addressing genetic features related to dyslipidemia/atherosclerosis using the zebrafish model is already a reality and active research is now ongoing in this promising area. Novel technologies (gene and genome editing) may help to identify new candidate genes involved in dyslipidemia and dyslipidemia-related diseases. Based on these considerations, the zebrafish experimental model appears highly suitable for the study of exposome factors, genes and molecules involved in the development of atherosclerosis-related disease as well as for the validation of novel potential treatment options.
Collapse
Affiliation(s)
- Marina Vasyutina
- Almazov Federal Medical Research Centre, Saint Petersburg, Russia.
| | - Asiiat Alieva
- Almazov Federal Medical Research Centre, Saint Petersburg, Russia
| | - Olga Reutova
- Almazov Federal Medical Research Centre, Saint Petersburg, Russia
| | | | - Lada Murashova
- Almazov Federal Medical Research Centre, Saint Petersburg, Russia
| | | | - Alberico L Catapano
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy; IRCCS MultiMedica, Milan, Italy
| | - Andrea Baragetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy; IRCCS MultiMedica, Milan, Italy
| | - Paolo Magni
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy; IRCCS MultiMedica, Milan, Italy.
| |
Collapse
|
14
|
Facchinello N, Astone M, Audano M, Oberkersch RE, Spizzotin M, Calura E, Marques M, Crisan M, Mitro N, Santoro MM. Oxidative pentose phosphate pathway controls vascular mural cell coverage by regulating extracellular matrix composition. Nat Metab 2022; 4:123-140. [PMID: 35102339 PMCID: PMC7612297 DOI: 10.1038/s42255-021-00514-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 12/06/2021] [Indexed: 12/11/2022]
Abstract
Vascular mural cells (vMCs) play an essential role in the development and maturation of the vasculature by promoting vessel stabilization through their interactions with endothelial cells. Whether endothelial metabolism influences mural cell recruitment and differentiation is unknown. Here, we show that the oxidative pentose phosphate pathway (oxPPP) in endothelial cells is required for establishing vMC coverage of the dorsal aorta during early vertebrate development in zebrafish and mice. We demonstrate that laminar shear stress and blood flow maintain oxPPP activity, which in turn, promotes elastin expression in blood vessels through production of ribose-5-phosphate. Elastin is both necessary and sufficient to drive vMC recruitment and maintenance when the oxPPP is active. In summary, our work demonstrates that endothelial cell metabolism regulates blood vessel maturation by controlling vascular matrix composition and vMC recruitment.
Collapse
Affiliation(s)
- Nicola Facchinello
- Laboratory of Angiogenesis and Cancer Metabolism, DiBio, University of Padua, Padua, Italy
| | - Matteo Astone
- Laboratory of Angiogenesis and Cancer Metabolism, DiBio, University of Padua, Padua, Italy
| | - Matteo Audano
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, Milan, Italy
| | - Roxana E Oberkersch
- Laboratory of Angiogenesis and Cancer Metabolism, DiBio, University of Padua, Padua, Italy
| | - Marianna Spizzotin
- Laboratory of Angiogenesis and Cancer Metabolism, DiBio, University of Padua, Padua, Italy
| | - Enrica Calura
- Department of Biology, University of Padua, Padua, Italy
| | - Madalena Marques
- Centre for Cardiovascular Science and Centre for Regenerative Medicine/Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Mihaela Crisan
- Centre for Cardiovascular Science and Centre for Regenerative Medicine/Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Nico Mitro
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, Milan, Italy
| | - Massimo M Santoro
- Laboratory of Angiogenesis and Cancer Metabolism, DiBio, University of Padua, Padua, Italy.
| |
Collapse
|
15
|
Cheung PY, Harrison PT, Davidson AJ, Hollywood JA. In Vitro and In Vivo Models to Study Nephropathic Cystinosis. Cells 2021; 11:6. [PMID: 35011573 PMCID: PMC8750259 DOI: 10.3390/cells11010006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/17/2021] [Accepted: 12/19/2021] [Indexed: 12/18/2022] Open
Abstract
The development over the past 50 years of a variety of cell lines and animal models has provided valuable tools to understand the pathophysiology of nephropathic cystinosis. Primary cultures from patient biopsies have been instrumental in determining the primary cause of cystine accumulation in the lysosomes. Immortalised cell lines have been established using different gene constructs and have revealed a wealth of knowledge concerning the molecular mechanisms that underlie cystinosis. More recently, the generation of induced pluripotent stem cells, kidney organoids and tubuloids have helped bridge the gap between in vitro and in vivo model systems. The development of genetically modified mice and rats have made it possible to explore the cystinotic phenotype in an in vivo setting. All of these models have helped shape our understanding of cystinosis and have led to the conclusion that cystine accumulation is not the only pathology that needs targeting in this multisystemic disease. This review provides an overview of the in vitro and in vivo models available to study cystinosis, how well they recapitulate the disease phenotype, and their limitations.
Collapse
Affiliation(s)
- Pang Yuk Cheung
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland 1142, New Zealand; (P.Y.C.); (A.J.D.)
| | - Patrick T. Harrison
- Department of Physiology, BioSciences Institute, University College Cork, T12 XF62 Cork, Ireland;
| | - Alan J. Davidson
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland 1142, New Zealand; (P.Y.C.); (A.J.D.)
| | - Jennifer A. Hollywood
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland 1142, New Zealand; (P.Y.C.); (A.J.D.)
| |
Collapse
|
16
|
A network-based approach to identify protein kinases critical for regulating srebf1 in lipid deposition causing obesity. Funct Integr Genomics 2021; 21:557-570. [PMID: 34327622 DOI: 10.1007/s10142-021-00798-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/09/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022]
Abstract
Obesity is a rapidly growing health pandemic, underlying a wide variety of disease conditions leading to increases in global mortality. It is known that the phosphorylation of various proteins regulates sterol regulatory element-binding transcription factors 1 (srebf1), a key lipogenic transcription factor, to cause the development of obesity. To detect the key protein kinases for regulating srebf1 in lipid deposition, we established the srebf1 knockout model in zebrafish (KO, srebf1-/-) by CRISPR/Cas9. The KO zebrafish exhibited a significant reduction of total free fatty acid content (fell 60.5%) and lipid deposition decrease compared with wild-type (WT) zebrafish. Meanwhile, srebf1 deletion in zebrafish eliminated lipid deposition induced by high-fat diet feeding. Compared with WT zebrafish, a total of 697 differentially expressed proteins and 316 differentially expressed phosphoproteins with 439 sites were identified in KO by differential proteomic and phosphoproteomic analyses. A significant number of proteins identified were involved in lipid and glucose metabolism. Moreover, some protein kinases critical for regulating srebf1 in lipid deposition, including Cdk2, Pkc, Prkceb, mTORC1, Mapk12, and Wnk1, were determined by network analyses. An in vitro study was performed to verify the network analysis results. Our findings provide potential targets (kinases) for human obesity treatments.
Collapse
|
17
|
Roles of Estrogens in the Healthy and Diseased Oviparous Vertebrate Liver. Metabolites 2021; 11:metabo11080502. [PMID: 34436443 PMCID: PMC8398935 DOI: 10.3390/metabo11080502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 12/12/2022] Open
Abstract
The liver is a vital organ that sustains multiple functions beneficial for the whole organism. It is sexually dimorphic, presenting sex-biased gene expression with implications for the phenotypic differences between males and females. Estrogens are involved in this sex dimorphism and their actions in the liver of several reptiles, fishes, amphibians, and birds are discussed. The liver participates in reproduction by producing vitellogenins (yolk proteins) and eggshell proteins under the control of estrogens that act via two types of receptors active either mainly in the cell nucleus (ESR) or the cell membrane (GPER1). Estrogens also control hepatic lipid and lipoprotein metabolisms, with a triglyceride carrier role for VLDL from the liver to the ovaries during oogenesis. Moreover, the activation of the vitellogenin genes is used as a robust biomarker for exposure to xenoestrogens. In the context of liver diseases, high plasma estrogen levels are observed in fatty liver hemorrhagic syndrome (FLHS) in chicken implicating estrogens in the disease progression. Fishes are also used to investigate liver diseases, including models generated by mutation and transgenesis. In conclusion, studies on the roles of estrogens in the non-mammalian oviparous vertebrate liver have contributed enormously to unveil hormone-dependent physiological and physiopathological processes.
Collapse
|
18
|
Zebrafish as an animal model for biomedical research. Exp Mol Med 2021; 53:310-317. [PMID: 33649498 PMCID: PMC8080808 DOI: 10.1038/s12276-021-00571-5] [Citation(s) in RCA: 187] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 12/15/2022] Open
Abstract
Zebrafish have several advantages compared to other vertebrate models used in modeling human diseases, particularly for large-scale genetic mutant and therapeutic compound screenings, and other biomedical research applications. With the impactful developments of CRISPR and next-generation sequencing technology, disease modeling in zebrafish is accelerating the understanding of the molecular mechanisms of human genetic diseases. These efforts are fundamental for the future of precision medicine because they provide new diagnostic and therapeutic solutions. This review focuses on zebrafish disease models for biomedical research, mainly in developmental disorders, mental disorders, and metabolic diseases.
Collapse
|
19
|
van de Venter M, Didloff J, Reddy S, Swanepoel B, Govender S, Dambuza NS, Williams S, Koekemoer TC, Venables L. Wild-Type Zebrafish ( Danio rerio) Larvae as a Vertebrate Model for Diabetes and Comorbidities: A Review. Animals (Basel) 2020; 11:E54. [PMID: 33396883 PMCID: PMC7824285 DOI: 10.3390/ani11010054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 12/12/2022] Open
Abstract
Zebrafish have become a popular alternative to higher animals in biomedical and pharmaceutical research. The development of stable mutant lines to model target specific aspects of many diseases, including diabetes, is well reported. However, these mutant lines are much more costly and challenging to maintain than wild-type zebrafish and are simply not an option for many research facilities. As an alternative to address the disadvantages of advanced mutant lines, wild-type larvae may represent a suitable option. In this review, we evaluate organ development in zebrafish larvae and discuss established methods that use wild-type zebrafish larvae up to seven days post fertilization to test for potential drug candidates for diabetes and its commonly associated conditions of oxidative stress and inflammation. This provides an up to date overview of the relevance of wild-type zebrafish larvae as a vertebrate antidiabetic model and confidence as an alternative tool for preclinical studies. We highlight the advantages and disadvantages of established methods and suggest recommendations for future developments to promote the use of zebrafish, specifically larvae, rather than higher animals in the early phase of antidiabetic drug discovery.
Collapse
Affiliation(s)
- Maryna van de Venter
- Department of Biochemistry and Microbiology, Nelson Mandela University, PO Box 77000, Port Elizabeth 6031, South Africa; (J.D.); (S.R.); (B.S.); (S.G.); (S.W.); (T.C.K.); (L.V.)
| | - Jenske Didloff
- Department of Biochemistry and Microbiology, Nelson Mandela University, PO Box 77000, Port Elizabeth 6031, South Africa; (J.D.); (S.R.); (B.S.); (S.G.); (S.W.); (T.C.K.); (L.V.)
| | - Shanika Reddy
- Department of Biochemistry and Microbiology, Nelson Mandela University, PO Box 77000, Port Elizabeth 6031, South Africa; (J.D.); (S.R.); (B.S.); (S.G.); (S.W.); (T.C.K.); (L.V.)
| | - Bresler Swanepoel
- Department of Biochemistry and Microbiology, Nelson Mandela University, PO Box 77000, Port Elizabeth 6031, South Africa; (J.D.); (S.R.); (B.S.); (S.G.); (S.W.); (T.C.K.); (L.V.)
| | - Sharlene Govender
- Department of Biochemistry and Microbiology, Nelson Mandela University, PO Box 77000, Port Elizabeth 6031, South Africa; (J.D.); (S.R.); (B.S.); (S.G.); (S.W.); (T.C.K.); (L.V.)
| | - Ntokozo Shirley Dambuza
- Department of Pharmacy, Nelson Mandela University, PO Box 77000, Port Elizabeth 6031, South Africa;
| | - Saralene Williams
- Department of Biochemistry and Microbiology, Nelson Mandela University, PO Box 77000, Port Elizabeth 6031, South Africa; (J.D.); (S.R.); (B.S.); (S.G.); (S.W.); (T.C.K.); (L.V.)
| | - Trevor Craig Koekemoer
- Department of Biochemistry and Microbiology, Nelson Mandela University, PO Box 77000, Port Elizabeth 6031, South Africa; (J.D.); (S.R.); (B.S.); (S.G.); (S.W.); (T.C.K.); (L.V.)
| | - Luanne Venables
- Department of Biochemistry and Microbiology, Nelson Mandela University, PO Box 77000, Port Elizabeth 6031, South Africa; (J.D.); (S.R.); (B.S.); (S.G.); (S.W.); (T.C.K.); (L.V.)
| |
Collapse
|
20
|
Landgraf K, Klöting N, Gericke M, Maixner N, Guiu-Jurado E, Scholz M, Witte AV, Beyer F, Schwartze JT, Lacher M, Villringer A, Kovacs P, Rudich A, Blüher M, Kiess W, Körner A. The Obesity-Susceptibility Gene TMEM18 Promotes Adipogenesis through Activation of PPARG. Cell Rep 2020; 33:108295. [PMID: 33086065 DOI: 10.1016/j.celrep.2020.108295] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 08/25/2020] [Accepted: 09/30/2020] [Indexed: 01/14/2023] Open
Abstract
TMEM18 is the strongest candidate for childhood obesity identified from GWASs, yet as for most GWAS-derived obesity-susceptibility genes, the functional mechanism remains elusive. We here investigate the relevance of TMEM18 for adipose tissue development and obesity. We demonstrate that adipocyte TMEM18 expression is downregulated in children with obesity. Functionally, downregulation of TMEM18 impairs adipocyte formation in zebrafish and in human preadipocytes, indicating that TMEM18 is important for adipocyte differentiation in vivo and in vitro. On the molecular level, TMEM18 activates PPARG, particularly upregulating PPARG1 promoter activity, and this activation is repressed by inflammatory stimuli. The relationship between TMEM18 and PPARG1 is also evident in adipocytes of children and is clinically associated with obesity and adipocyte hypertrophy, inflammation, and insulin resistance. Our findings indicate a role of TMEM18 as an upstream regulator of PPARG signaling driving healthy adipogenesis, which is dysregulated with adipose tissue dysfunction and obesity.
Collapse
Affiliation(s)
- Kathrin Landgraf
- Center for Pediatric Research Leipzig (CPL), Hospital for Children & Adolescents, University of Leipzig, Leipzig 04103, Germany.
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig 04103, Germany; Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig, Leipzig 04103, Germany
| | - Martin Gericke
- Institute of Anatomy, University of Leipzig, Leipzig 04103, Germany
| | - Nitzan Maixner
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Esther Guiu-Jurado
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig, Leipzig 04103, Germany
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig 04103, Germany; LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig 04103, Germany
| | - A Veronica Witte
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig 04103, Germany
| | - Frauke Beyer
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig 04103, Germany
| | - Julian T Schwartze
- Center for Pediatric Research Leipzig (CPL), Hospital for Children & Adolescents, University of Leipzig, Leipzig 04103, Germany
| | - Martin Lacher
- Department of Pediatric Surgery, University of Leipzig, Leipzig 04103, Germany
| | - Arno Villringer
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig 04103, Germany
| | - Peter Kovacs
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig, Leipzig 04103, Germany
| | - Assaf Rudich
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig 04103, Germany; Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig, Leipzig 04103, Germany
| | - Wieland Kiess
- Center for Pediatric Research Leipzig (CPL), Hospital for Children & Adolescents, University of Leipzig, Leipzig 04103, Germany
| | - Antje Körner
- Center for Pediatric Research Leipzig (CPL), Hospital for Children & Adolescents, University of Leipzig, Leipzig 04103, Germany.
| |
Collapse
|
21
|
A Great Catch for Investigating Inborn Errors of Metabolism-Insights Obtained from Zebrafish. Biomolecules 2020; 10:biom10091352. [PMID: 32971894 PMCID: PMC7564250 DOI: 10.3390/biom10091352] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/18/2020] [Accepted: 09/19/2020] [Indexed: 12/14/2022] Open
Abstract
Inborn errors of metabolism cause abnormal synthesis, recycling, or breakdown of amino acids, neurotransmitters, and other various metabolites. This aberrant homeostasis commonly causes the accumulation of toxic compounds or depletion of vital metabolites, which has detrimental consequences for the patients. Efficient and rapid intervention is often key to survival. Therefore, it requires useful animal models to understand the pathomechanisms and identify promising therapeutic drug targets. Zebrafish are an effective tool to investigate developmental mechanisms and understanding the pathophysiology of disorders. In the past decades, zebrafish have proven their efficiency for studying genetic disorders owing to the high degree of conservation between human and zebrafish genes. Subsequently, several rare inherited metabolic disorders have been successfully investigated in zebrafish revealing underlying mechanisms and identifying novel therapeutic targets, including methylmalonic acidemia, Gaucher’s disease, maple urine disorder, hyperammonemia, TRAPPC11-CDGs, and others. This review summarizes the recent impact zebrafish have made in the field of inborn errors of metabolism.
Collapse
|
22
|
Katoch S, Patial V. Zebrafish: An emerging model system to study liver diseases and related drug discovery. J Appl Toxicol 2020; 41:33-51. [PMID: 32656821 DOI: 10.1002/jat.4031] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/31/2020] [Accepted: 06/11/2020] [Indexed: 01/03/2023]
Abstract
The zebrafish has emerged as a powerful vertebrate model for studying liver-associated disorders. Liver damage is a crucial problem in the process of drug development and zebrafish have proven to be an important tool for the high-throughput screening of drugs for hepatotoxicity. Although the structure of the zebrafish liver differs to that of mammals, the fundamental physiologic processes, genetic mutations and manifestations of pathogenic responses to environmental insults exhibit much similarity. The larval transparency of the zebrafish is a great advantage for real-time imaging in hepatic studies. The zebrafish has a broad spectrum of cytochrome P450 enzymes, which enable the biotransformation of drugs via similar pathways as mammals, including oxidation, reduction and hydrolysis reactions. In the present review, we appraise the various drugs, chemicals and toxins used to study liver toxicity in zebrafish and their similarities to the rodent models for liver-related studies. Interestingly, the zebrafish has also been effectively used to study the pathophysiology of nonalcoholic and alcoholic fatty liver disease. The genetic models of liver disorders and their easy manipulation provide great opportunity in the area of drug development. The zebrafish has proven to be an influential model for the hepatic system due to its invertebrate-like advantages coupled with its vertebrate biology. The present review highlights the pivotal role of zebrafish in bridging the gap between cell-based and mammalian models.
Collapse
Affiliation(s)
- Swati Katoch
- Pharmacology and Toxicology Laboratory, Food and Nutraceuticals Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, India
| | - Vikram Patial
- Pharmacology and Toxicology Laboratory, Food and Nutraceuticals Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR- Institute of Himalayan Bioresource Technology, Palampur, India
| |
Collapse
|
23
|
Wiggenhauser LM, Kroll J. Vascular Damage in Obesity and Diabetes: Highlighting Links Between Endothelial Dysfunction and Metabolic Disease in Zebrafish and Man. Curr Vasc Pharmacol 2020; 17:476-490. [PMID: 30378499 DOI: 10.2174/1570161116666181031101413] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 09/11/2018] [Accepted: 09/25/2018] [Indexed: 02/08/2023]
Abstract
Endothelial dysfunction is an initial pathophysiological mechanism of vascular damage and is further recognized as an independent predictor of negative prognosis in diabetes-induced micro- and macrovascular complications. Insight into the capability of zebrafish to model metabolic disease like obesity and type II diabetes has increased and new evidence on the induction of vascular pathologies in zebrafish through metabolic disease is available. Here, we raise the question, if zebrafish can be utilized to study the initial impairments of vascular complications in metabolic disorders. In this review, we focus on the advances made to develop models of obesity and type II diabetes in zebrafish, discuss the key points and characteristics of these models, while highlighting the available information linked to the development of endothelial dysfunction in zebrafish and man. We show that larval and adult zebrafish develop metabolic dysregulation in the settings of obesity and diabetes, exhibiting pathophysiological mechanisms, which mimic the human condition. The most important genes related to endothelial dysfunction are present in zebrafish and further display similar functions as in mammals. Several suggested contributors to endothelial dysfunction found in these models, namely hyperinsulinaemia, hyperglycaemia, hyperlipidaemia and hyperleptinaemia are highlighted and the available data from zebrafish are summarised. Many underlying processes of endothelial dysfunction in obesity and diabetes are fundamentally present in zebrafish and provide ground for the assumption, that zebrafish can develop endothelial dysfunction. Conservation of basic biological mechanisms is established for zebrafish, but focused investigation on the subject is now needed as validation and particularly more research is necessary to understand the differences between zebrafish and man. The available data demonstrate the relevance of zebrafish as a model for metabolic disease and their ability to become a proponent for the investigation of vascular damage in the settings of obesity and diabetes.
Collapse
Affiliation(s)
- Lucas Moritz Wiggenhauser
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Jens Kroll
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| |
Collapse
|
24
|
Dolgova NV, Nehzati S, MacDonald TC, Summers KL, Crawford AM, Krone PH, George GN, Pickering IJ. Disruption of selenium transport and function is a major contributor to mercury toxicity in zebrafish larvae. Metallomics 2020; 11:621-631. [PMID: 30688331 DOI: 10.1039/c8mt00315g] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Mercury is one of the most toxic elements threatening the biosphere, with levels steadily rising due to both natural and human activities. Selenium is an essential micronutrient, required for normal development and functioning of many organisms. While selenium is known to counteract mercury's toxicity under some conditions, to date information about the mercury-selenium relationship is fragmented and often controversial. As part of a systematic study of mercury and selenium interactions, zebrafish (Danio rerio) larvae (a model verterbrate) were exposed to methylmercury chloride or mercuric chloride. The influence of pre- and post-treatment of selenomethionine on the level and distribution of mercury and selenium in the brain and eye sections, as well as on toxicity, were examined. Selenomethionine treatment decreased the amount of maternally transfered mercury in the larval brain. Selenomethionine treatment prior to exposure to mercuric chloride increased both mercury and selenium levels in the brain but decreased their toxic effects. Conversely, methylmercury levels were not changed as a result of selenium pre-treatment, while toxicity was increased. Strikingly, both forms of mercury severely disrupted selenium metabolism, not only by depleting selenium levels due to formation of Hg-Se complexes, but also by blocking selenium transport into and out of tissues, suggesting that restoring normal selenium levels by treating the organism with selenium after mercury exposure may not be possible. Disruption of selenium metabolism by mercury may lead to disruption in function of selenoproteins. Indeed, the production of thyroid hormones by selenoprotein deiodinases was found to be severely impaired as a result of mercury exposure, with selenomethionine not always being a suitable source of selenium to restore thyroid hormone levels.
Collapse
Affiliation(s)
- Natalia V Dolgova
- Molecular and Environment Sciences Group, Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Wang J, Li Y, Lai K, Zhong Q, Demin KA, Kalueff AV, Song C. High-glucose/high-cholesterol diet in zebrafish evokes diabetic and affective pathogenesis: The role of peripheral and central inflammation, microglia and apoptosis. Prog Neuropsychopharmacol Biol Psychiatry 2020; 96:109752. [PMID: 31446160 DOI: 10.1016/j.pnpbp.2019.109752] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/18/2019] [Accepted: 08/21/2019] [Indexed: 01/07/2023]
Abstract
Neuroinflammation and metabolic deficits contribute to the etiology of human affective disorders, such as anxiety and depression. The zebrafish (Danio rerio) has recently emerged as a powerful new model organism in CNS disease modeling. Here, we exposed zebrafish to 2% glucose and 10% cholesterol for 19 days to experimentally induce type 2 diabetes (DM) and to assess stress responses, microglia, inflammation and apoptosis. We analyzed zebrafish anxiety-like behavior in the novel tank and light-dark box (Days 15-16) tests, as well as examined their biochemical and genomic biomarkers (Day 19). Confirming DM-like state in zebrafish, we found higher whole-body glucose, triglyceride, total cholesterol, low-density lipoprotein levels and glucagon mRNA expression, and lower high-density lipoprotein levels. DM zebrafish also showed anxiety-like behavior, elevated whole-body cortisol and cytokines IFN-γ and IL-4, as well as higher brain mRNA expression of the glucocorticoid receptor, CD11b (a microglial biomarker), pro-inflammatory cytokines IL-6 and TNF-α (but not IL-1β or anti-inflammatory cytokines IL-4 and IL-10), GFAP (an astrocytal biomarker), neurotrophin BDNF, its receptors p75 and TrkB, as well as apoptotic Bax and Caspase-3 (but not BCl-2) genes. Collectively, this supports the overlapping nature of DM-related affective pathogenesis and emphasizes the role of peripheral and central inflammation and apoptosis in DM-related affective and neuroendocrine deficits in zebrafish.
Collapse
Affiliation(s)
- JiaJia Wang
- Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China; Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Zhanjiang, China; Marine Medicine Development Center, Shenzhen Institute, Guangdong Ocean University, Shenzhen, China
| | - YanJun Li
- Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - Ke Lai
- Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - QiMei Zhong
- Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China; Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Zhanjiang, China; Marine Medicine Development Center, Shenzhen Institute, Guangdong Ocean University, Shenzhen, China
| | - Konstantin A Demin
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China; Ural Federal University, Ekaterinburg, Russia.
| | - Cai Song
- Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China; Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Zhanjiang, China; Marine Medicine Development Center, Shenzhen Institute, Guangdong Ocean University, Shenzhen, China.
| |
Collapse
|
26
|
Flores EM, Nguyen AT, Odem MA, Eisenhoffer GT, Krachler AM. The zebrafish as a model for gastrointestinal tract-microbe interactions. Cell Microbiol 2020; 22:e13152. [PMID: 31872937 DOI: 10.1111/cmi.13152] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/07/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023]
Abstract
The zebrafish (Danio rerio) has become a widely used vertebrate model for bacterial, fungal, viral, and protozoan infections. Due to its genetic tractability, large clutch sizes, ease of manipulation, and optical transparency during early life stages, it is a particularly useful model to address questions about the cellular microbiology of host-microbe interactions. Although its use as a model for systemic infections, as well as infections localised to the hindbrain and swimbladder having been thoroughly reviewed, studies focusing on host-microbe interactions in the zebrafish gastrointestinal tract have been neglected. Here, we summarise recent findings regarding the developmental and immune biology of the gastrointestinal tract, drawing parallels to mammalian systems. We discuss the use of adult and larval zebrafish as models for gastrointestinal infections, and more generally, for studies of host-microbe interactions in the gut.
Collapse
Affiliation(s)
- Erika M Flores
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.,M.D. Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Anh T Nguyen
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.,M.D. Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Max A Odem
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas
| | - George T Eisenhoffer
- M.D. Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas.,Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anne Marie Krachler
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.,M.D. Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| |
Collapse
|
27
|
Nath AK, Ma J, Chen ZZ, Li Z, Vitery MDC, Kelley ML, Peterson RT, Gerszten RE, Yeh JRJ. Genetic deletion of gpr27 alters acylcarnitine metabolism, insulin sensitivity, and glucose homeostasis in zebrafish. FASEB J 2019; 34:1546-1557. [PMID: 31914600 DOI: 10.1096/fj.201901466r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 10/31/2019] [Accepted: 11/11/2019] [Indexed: 02/06/2023]
Abstract
G protein-coupled receptors (GPCRs) comprise the largest group of membrane receptors in eukaryotic genomes and collectively they regulate nearly all cellular processes. Despite the widely recognized importance of this class of proteins, many GPCRs remain understudied. G protein-coupled receptor 27 (Gpr27) is an orphan GPCR that displays high conservation during vertebrate evolution. Although, GPR27 is known to be expressed in tissues that regulate metabolism including the pancreas, skeletal muscle, and adipose tissue, its functions are poorly characterized. Therefore, to investigate the potential roles of Gpr27 in energy metabolism, we generated a whole body gpr27 knockout zebrafish line. Loss of gpr27 potentiated the elevation in glucose levels induced by pharmacological or nutritional perturbations. We next leveraged a mass spectrometry metabolite profiling platform to identify other potential metabolic functions of Gpr27. Notably, genetic deletion of gpr27 elevated medium-chain acylcarnitines, in particular C6-hexanoylcarnitine, C8-octanoylcarnitine, C9-nonanoylcarnitine, and C10-decanoylcarnitine, lipid species known to be associated with insulin resistance in humans. Concordantly, gpr27 deletion in zebrafish abrogated insulin-dependent Akt phosphorylation and glucose utilization. Finally, loss of gpr27 increased the expression of key enzymes in carnitine shuttle complex, in particular the homolog to the brain-specific isoform of CPT1C which functions as a hypothalamic energy senor. In summary, our findings shed light on the biochemical functions of Gpr27 by illuminating its role in lipid metabolism, insulin signaling, and glucose homeostasis.
Collapse
Affiliation(s)
- Anjali K Nath
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Broad Institute, Cambridge, MA, USA
| | - Junyan Ma
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Zsu-Zsu Chen
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Zhuyun Li
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, USA
| | | | - Michelle L Kelley
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, USA
| | | | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Broad Institute, Cambridge, MA, USA
| | - Jing-Ruey J Yeh
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
28
|
Qiu L, Jia K, Huang L, Liao X, Guo X, Lu H. Hepatotoxicity of tricyclazole in zebrafish (Danio rerio). CHEMOSPHERE 2019; 232:171-179. [PMID: 31154177 DOI: 10.1016/j.chemosphere.2019.05.159] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/17/2019] [Accepted: 05/18/2019] [Indexed: 06/09/2023]
Abstract
Tricyclazole is widely used in agriculture as a pesticide, but its toxicity in vertebrates is currently poorly evaluated. In this study, we used zebrafish to assess the toxicity of tricyclazole. We found that tricyclazole induces liver damage, or hepatotoxicity, in zebrafish, during both development and adulthood. In embryos, we found that tricyclazole affected the liver development rather than other endodermal tissues such as gut and pancreas. In both embryos and adult zebrafish livers, tricyclazole disrupted the relationship between oxidant and antioxidant system and resulted in reactive oxygen species (ROS) overload. Meanwhile, it triggered hepatocyte apoptosis and disturbed carbohydrate/lipid metabolism and energy demand systems. These results suggested that tricyclazole could cause severe consequences for vertebrate hepatic development and function.
Collapse
Affiliation(s)
- Lingyu Qiu
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, Jiangxi, China
| | - Kun Jia
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, Jiangxi, China; Center for Developmental Biology of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi, China
| | - Lirong Huang
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, Jiangxi, China; Center for Developmental Biology of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi, China
| | - Xinjun Liao
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, Jiangxi, China; Center for Developmental Biology of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi, China
| | - Xinchun Guo
- School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, Jiangxi, China.
| | - Huiqiang Lu
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, Jiangxi, China; Center for Developmental Biology of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi, China.
| |
Collapse
|
29
|
Dhillon SS, Torell F, Donten M, Lundstedt-Enkel K, Bennett K, Rännar S, Trygg J, Lundstedt T. Metabolic profiling of zebrafish embryo development from blastula period to early larval stages. PLoS One 2019; 14:e0213661. [PMID: 31086370 PMCID: PMC6516655 DOI: 10.1371/journal.pone.0213661] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/15/2019] [Indexed: 11/18/2022] Open
Abstract
The zebrafish embryo is a popular model for drug screening, disease modelling and molecular genetics. In this study, samples were obtained from zebrafish at different developmental stages. The stages that were chosen were 3/4, 4/5, 24, 48, 72 and 96 hours post fertilization (hpf). Each sample included fifty embryos. The samples were analysed using gas chromatography time-of-flight mass spectrometry (GC-TOF-MS). Principle component analysis (PCA) was applied to get an overview of the data and orthogonal projection to latent structure discriminant analysis (OPLS-DA) was utilised to discriminate between the developmental stages. In this way, changes in metabolite profiles during vertebrate development could be identified. Using a GC-TOF-MS metabolomics approach it was found that nucleotides and metabolic fuel (glucose) were elevated at early stages of embryogenesis, whereas at later stages amino acids and intermediates in the Krebs cycle were abundant. This agrees with zebrafish developmental biology, as organs such as the liver and pancreas develop at later stages. Thus, metabolomics of zebrafish embryos offers a unique opportunity to investigate large scale changes in metabolic processes during important developmental stages in vertebrate development. In terms of stability of the metabolic profile and viability of the embryos, it was concluded at 72 hpf was a suitable time point for the use of zebrafish as a model system in numerous scientific applications.
Collapse
Affiliation(s)
- Sundeep S. Dhillon
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Frida Torell
- Computational Life Science Cluster (CLiC), Department of Chemistry, Umeå University, Umeå, Sweden
- Accelerator Lab (ACL), Karlsruhe Institute of Technology, Karlsruhe, Germany
- AcureOmics, Umeå, Sweden
| | | | - Katrin Lundstedt-Enkel
- AcureOmics, Umeå, Sweden
- Environmental toxicology, Organismal Biology, Uppsala University, Uppsala, Sweden
| | | | - Stefan Rännar
- AcureOmics, Umeå, Sweden
- Corporate Research, Sartorius AG, Göttingen, Germany
| | - Johan Trygg
- Computational Life Science Cluster (CLiC), Department of Chemistry, Umeå University, Umeå, Sweden
- Corporate Research, Sartorius AG, Göttingen, Germany
- * E-mail:
| | - Torbjörn Lundstedt
- AcureOmics, Umeå, Sweden
- Deparment of Pharmaceutical Bioscience, BMC, Uppsala University, Uppsala, Sweden
| |
Collapse
|
30
|
Dickmeis T, Feng Y, Mione MC, Ninov N, Santoro M, Spaink HP, Gut P. Nano-Sampling and Reporter Tools to Study Metabolic Regulation in Zebrafish. Front Cell Dev Biol 2019; 7:15. [PMID: 30873407 PMCID: PMC6401643 DOI: 10.3389/fcell.2019.00015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 01/31/2019] [Indexed: 01/09/2023] Open
Abstract
In the past years, evidence has emerged that hallmarks of human metabolic disorders can be recapitulated in zebrafish using genetic, pharmacological or dietary interventions. An advantage of modeling metabolic diseases in zebrafish compared to other "lower organisms" is the presence of a vertebrate body plan providing the possibility to study the tissue-intrinsic processes preceding the loss of metabolic homeostasis. While the small size of zebrafish is advantageous in many aspects, it also has shortcomings such as the difficulty to obtain sufficient amounts for biochemical analyses in response to metabolic challenges. A workshop at the European Zebrafish Principal Investigator meeting in Trento, Italy, was dedicated to discuss the advantages and disadvantages of zebrafish to study metabolic disorders. This perspective article by the participants highlights strategies to achieve improved tissue-resolution for read-outs using "nano-sampling" approaches for metabolomics as well as live imaging of zebrafish expressing fluorescent reporter tools that inform on cellular or subcellular metabolic processes. We provide several examples, including the use of reporter tools to study the heterogeneity of pancreatic beta-cells within their tissue environment. While limitations exist, we believe that with the advent of new technologies and more labs developing methods that can be applied to minimal amounts of tissue or single cells, zebrafish will further increase their utility to study energy metabolism.
Collapse
Affiliation(s)
- Thomas Dickmeis
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Yi Feng
- Centre for Inflammation Research, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland
| | | | - Nikolay Ninov
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | | | - Herman P. Spaink
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Philipp Gut
- Nestlé Research, EPFL Innovation Park, Lausanne, Switzerland
| |
Collapse
|
31
|
Weger M, Weger BD, Görling B, Poschet G, Yildiz M, Hell R, Luy B, Akcay T, Güran T, Dickmeis T, Müller F, Krone N. Glucocorticoid deficiency causes transcriptional and post-transcriptional reprogramming of glutamine metabolism. EBioMedicine 2018; 36:376-389. [PMID: 30266295 PMCID: PMC6197330 DOI: 10.1016/j.ebiom.2018.09.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/07/2018] [Accepted: 09/13/2018] [Indexed: 01/12/2023] Open
Abstract
Background Deficient glucocorticoid biosynthesis leading to adrenal insufficiency is life-threatening and is associated with significant co-morbidities. The affected pathways underlying the pathophysiology of co-morbidities due to glucocorticoid deficiency remain poorly understood and require further investigation. Methods To explore the pathophysiological processes related to glucocorticoid deficiency, we have performed global transcriptional, post-transcriptional and metabolic profiling of a cortisol-deficient zebrafish mutant with a disrupted ferredoxin (fdx1b) system. Findings fdx1b−/− mutants show pervasive reprogramming of metabolism, in particular of glutamine-dependent pathways such as glutathione metabolism, and exhibit changes of oxidative stress markers. The glucocorticoid-dependent post-transcriptional regulation of key enzymes involved in de novo purine synthesis was also affected in this mutant. Moreover, fdx1b−/− mutants exhibit crucial features of primary adrenal insufficiency, and mirror metabolic changes detected in primary adrenal insufficiency patients. Interpretation Our study provides a detailed map of metabolic changes induced by glucocorticoid deficiency as a consequence of a disrupted ferredoxin system in an animal model of adrenal insufficiency. This improved pathophysiological understanding of global glucocorticoid deficiency informs on more targeted translational studies in humans suffering from conditions associated with glucocorticoid deficiency. Fund Marie Curie Intra-European Fellowships for Career Development, HGF-programme BIFTM, Deutsche Forschungsgemeinschaft, BBSRC.
Collapse
Affiliation(s)
- Meltem Weger
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | - Benjamin D Weger
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Benjamin Görling
- Institute for Organic Chemistry and Institute for Biological Interfaces 4 - Magnetic Resonance, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Gernot Poschet
- Centre for Organismal Studies (COS), Heidelberg University, 69120 Heidelberg, Germany
| | - Melek Yildiz
- Kanuni Sultan Süleyman Education and Research Hospital, Küçükçekmece, Istanbul, Turkey
| | - Rüdiger Hell
- Centre for Organismal Studies (COS), Heidelberg University, 69120 Heidelberg, Germany
| | - Burkhard Luy
- Institute for Organic Chemistry and Institute for Biological Interfaces 4 - Magnetic Resonance, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Teoman Akcay
- Istinye University Gaziosmanpasa Medical Park Hospital Gaziosmanpasa, Istanbul, Turkey
| | - Tülay Güran
- Marmara University, Department of Pediatric Endocrinology and Diabetes, Pendik, Istanbul, Turkey
| | - Thomas Dickmeis
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Ferenc Müller
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Nils Krone
- Department of Oncology & Metabolism, University of Sheffield, Sheffield S10 2TH, UK; Department of Biomedical Science, The Bateson Centre, Firth Court, Western Bank, Sheffield S10 2TN, UK..
| |
Collapse
|
32
|
Silver Nanoparticles Synthesized Using Eysenhardtia polystachya and Assessment of the Inhibition of Glycation in Multiple Stages In Vitro and in the Zebrafish Model. J CLUST SCI 2018. [DOI: 10.1007/s10876-018-1448-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
33
|
Elmonem MA, Berlingerio SP, van den Heuvel LP, de Witte PA, Lowe M, Levtchenko EN. Genetic Renal Diseases: The Emerging Role of Zebrafish Models. Cells 2018; 7:cells7090130. [PMID: 30200518 PMCID: PMC6162634 DOI: 10.3390/cells7090130] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 08/27/2018] [Accepted: 08/29/2018] [Indexed: 12/14/2022] Open
Abstract
The structural and functional similarity of the larval zebrafish pronephros to the human nephron, together with the recent development of easier and more precise techniques to manipulate the zebrafish genome have motivated many researchers to model human renal diseases in the zebrafish. Over the last few years, great advances have been made, not only in the modeling techniques of genetic diseases in the zebrafish, but also in how to validate and exploit these models, crossing the bridge towards more informative explanations of disease pathophysiology and better designed therapeutic interventions in a cost-effective in vivo system. Here, we review the significant progress in these areas giving special attention to the renal phenotype evaluation techniques. We further discuss the future applications of such models, particularly their role in revealing new genetic diseases of the kidney and their potential use in personalized medicine.
Collapse
Affiliation(s)
- Mohamed A Elmonem
- Department of Pediatric Nephrology & Development and Regeneration, University Hospitals Leuven, KU Leuven-University of Leuven, Herestraat 49, Box 817, 3000 Leuven, Belgium.
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, 11628 Cairo, Egypt.
| | - Sante Princiero Berlingerio
- Department of Pediatric Nephrology & Development and Regeneration, University Hospitals Leuven, KU Leuven-University of Leuven, Herestraat 49, Box 817, 3000 Leuven, Belgium.
| | - Lambertus P van den Heuvel
- Department of Pediatric Nephrology & Development and Regeneration, University Hospitals Leuven, KU Leuven-University of Leuven, Herestraat 49, Box 817, 3000 Leuven, Belgium.
- Department of Pediatric Nephrology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.
| | - Peter A de Witte
- Laboratory for Molecular Bio-Discovery, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven-University of Leuven, 3000 Leuven, Belgium.
| | - Martin Lowe
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK.
| | - Elena N Levtchenko
- Department of Pediatric Nephrology & Development and Regeneration, University Hospitals Leuven, KU Leuven-University of Leuven, Herestraat 49, Box 817, 3000 Leuven, Belgium.
| |
Collapse
|
34
|
van Leeuwen EM, Emri E, Merle BMJ, Colijn JM, Kersten E, Cougnard-Gregoire A, Dammeier S, Meester-Smoor M, Pool FM, de Jong EK, Delcourt C, Rodrigez-Bocanegra E, Biarnés M, Luthert PJ, Ueffing M, Klaver CCW, Nogoceke E, den Hollander AI, Lengyel I. A new perspective on lipid research in age-related macular degeneration. Prog Retin Eye Res 2018; 67:56-86. [PMID: 29729972 DOI: 10.1016/j.preteyeres.2018.04.006] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/25/2018] [Accepted: 04/30/2018] [Indexed: 12/15/2022]
Abstract
There is an urgency to find new treatment strategies that could prevent or delay the onset or progression of AMD. Different classes of lipids and lipoproteins metabolism genes have been associated with AMD in a multiple ways, but despite the ever-increasing knowledge base, we still do not understand fully how circulating lipids or local lipid metabolism contribute to AMD. It is essential to clarify whether dietary lipids, systemic or local lipoprotein metabolismtrafficking of lipids in the retina should be targeted in the disease. In this article, we critically evaluate what has been reported in the literature and identify new directions needed to bring about a significant advance in our understanding of the role for lipids in AMD. This may help to develop potential new treatment strategies through targeting the lipid homeostasis.
Collapse
Affiliation(s)
- Elisabeth M van Leeuwen
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Eszter Emri
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Benedicte M J Merle
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, Team LEHA, UMR 1219, F-33000, Bordeaux, France
| | - Johanna M Colijn
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Eveline Kersten
- Department of Ophthalmology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
| | - Audrey Cougnard-Gregoire
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, Team LEHA, UMR 1219, F-33000, Bordeaux, France
| | - Sascha Dammeier
- Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Germany
| | - Magda Meester-Smoor
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Eiko K de Jong
- Department of Ophthalmology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
| | - Cécile Delcourt
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, Team LEHA, UMR 1219, F-33000, Bordeaux, France
| | | | | | | | - Marius Ueffing
- Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Germany
| | - Caroline C W Klaver
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Ophthalmology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
| | - Everson Nogoceke
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Anneke I den Hollander
- Department of Ophthalmology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Imre Lengyel
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom.
| |
Collapse
|
35
|
Epigenetics in teleost fish: From molecular mechanisms to physiological phenotypes. Comp Biochem Physiol B Biochem Mol Biol 2018; 224:210-244. [PMID: 29369794 DOI: 10.1016/j.cbpb.2018.01.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 01/08/2018] [Accepted: 01/16/2018] [Indexed: 02/07/2023]
Abstract
While the field of epigenetics is increasingly recognized to contribute to the emergence of phenotypes in mammalian research models across different developmental and generational timescales, the comparative biology of epigenetics in the large and physiologically diverse vertebrate infraclass of teleost fish remains comparatively understudied. The cypriniform zebrafish and the salmoniform rainbow trout and Atlantic salmon represent two especially important teleost orders, because they offer the unique possibility to comparatively investigate the role of epigenetic regulation in 3R and 4R duplicated genomes. In addition to their sequenced genomes, these teleost species are well-characterized model species for development and physiology, and therefore allow for an investigation of the role of epigenetic modifications in the emergence of physiological phenotypes during an organism's lifespan and in subsequent generations. This review aims firstly to describe the evolution of the repertoire of genes involved in key molecular epigenetic pathways including histone modifications, DNA methylation and microRNAs in zebrafish, rainbow trout, and Atlantic salmon, and secondly, to discuss recent advances in research highlighting a role for molecular epigenetics in shaping physiological phenotypes in these and other teleost models. Finally, by discussing themes and current limitations of the emerging field of teleost epigenetics from both theoretical and technical points of view, we will highlight future research needs and discuss how epigenetics will not only help address basic research questions in comparative teleost physiology, but also inform translational research including aquaculture, aquatic toxicology, and human disease.
Collapse
|
36
|
Kopp R, Martínez IO, Legradi J, Legler J. Exposure to endocrine disrupting chemicals perturbs lipid metabolism and circadian rhythms. J Environ Sci (China) 2017; 62:133-137. [PMID: 29289284 DOI: 10.1016/j.jes.2017.10.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 09/15/2017] [Accepted: 10/25/2017] [Indexed: 06/07/2023]
Abstract
A growing body of evidence indicates that exposure to environmental chemicals can contribute to the etiology of obesity by inappropriately stimulating adipogenesis as well as perturbing lipid metabolism and energy balance. One potential mechanism by which chemical exposure can influence lipid metabolism is through disturbance of circadian rhythms, endogenously-driven cycles of roughly 24hr in length that coordinate biochemical, physiological, and behavioral processes in all organisms. Here we show for the first time that exposure to endocrine disrupting compounds (EDCs), including the pesticide tributyltin, two commercial flame retardants, and a UV-filter chemical found in sunscreens, can perturb both circadian clocks and lipid metabolism in vertebrates. Exposure of developing zebrafish to EDCs affects core clock activity and leads to a remarkable increase in lipid accumulation that is reminiscent of the effects observed for longdaysin, a known disruptor of circadian rhythms. Our data reveal a novel obesogenic mechanism of action for environmental chemicals, an observation which warrants further research. Because circadian clocks regulate a wide variety of physiological processes, identification of environmental chemicals capable of perturbing these systems may provide important insights into the development of environmentally-induced metabolic disease.
Collapse
Affiliation(s)
- Renate Kopp
- Institute for Environmental Studies (IVM), VU University Amsterdam, 1081, HV, The Netherlands.
| | - Irene Ozáez Martínez
- Biology and Environmental Toxicology Group, Faculty of Sciences, National Distance Education University, 28015 Madrid, Spain
| | - Jessica Legradi
- Institute for Environmental Studies (IVM), VU University Amsterdam, 1081, HV, The Netherlands
| | - Juliette Legler
- Institute for Environmental Studies (IVM), VU University Amsterdam, 1081, HV, The Netherlands; Institute for Environment, Health and Societies, College of Health and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, United Kingdom.
| |
Collapse
|
37
|
The tumor suppressor LKB1 regulates starvation-induced autophagy under systemic metabolic stress. Sci Rep 2017; 7:7327. [PMID: 28779098 PMCID: PMC5544676 DOI: 10.1038/s41598-017-07116-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 06/21/2017] [Indexed: 12/19/2022] Open
Abstract
Autophagy is an evolutionarily conserved process that degrades cellular components to restore energy homeostasis under limited nutrient conditions. How this starvation-induced autophagy is regulated at the whole-body level is not fully understood. Here, we show that the tumor suppressor Lkb1, which activates the key energy sensor AMPK, also regulates starvation-induced autophagy at the organismal level. Lkb1-deficient zebrafish larvae fail to activate autophagy in response to nutrient restriction upon yolk termination, shown by reduced levels of the autophagy-activating proteins Atg5, Lc3-II and Becn1, and aberrant accumulation of the cargo receptor and autophagy substrate p62. We demonstrate that the autophagy defect in lkb1 mutants can be partially rescued by inhibiting mTOR signaling but not by inhibiting the PI3K pathway. Interestingly, mTOR-independent activation of autophagy restores degradation of the aberrantly accumulated p62 in lkb1 mutants and prolongs their survival. Our data uncover a novel critical role for Lkb1 in regulating starvation-induced autophagy at the organismal level, providing mechanistic insight into metabolic adaptation during development.
Collapse
|
38
|
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: 207] [Impact Index Per Article: 29.6] [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
|
39
|
O’Hare EA, Yang R, Yerges-Armstrong L, Sreenivasan U, McFarland R, Leitch CC, Wilson MH, Narina S, Gorden A, Ryan K, Shuldiner AR, Farber SA, Wood GC, Still CD, Gerhard GS, Robishaw JD, Sztalryd C, Zaghloul NA. TM6SF2 rs58542926 impacts lipid processing in liver and small intestine. Hepatology 2017; 65:1526-1542. [PMID: 28027591 PMCID: PMC5397347 DOI: 10.1002/hep.29021] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 12/22/2016] [Accepted: 12/22/2016] [Indexed: 12/21/2022]
Abstract
The transmembrane 6 superfamily member 2 (TM6SF2) loss-of-function variant rs58542926 is a genetic risk factor for nonalcoholic fatty liver disease and progression to fibrosis but is paradoxically associated with lower levels of hepatically derived triglyceride-rich lipoproteins. TM6SF2 is expressed predominantly in liver and small intestine, sites for triglyceride-rich lipoprotein biogenesis and export. In light of this, we hypothesized that TM6SF2 may exhibit analogous effects on both liver and intestine lipid homeostasis. To test this, we genotyped rs58542926 in 983 bariatric surgery patients from the Geisinger Medical Center for Nutrition and Weight Management, Geisinger Health System, in Pennsylvania and from 3,556 study participants enrolled in the Amish Complex Disease Research Program. Although these two cohorts have different metabolic profiles, carriers in both cohorts had improved fasting lipid profiles. Importantly, following a high-fat challenge, carriers in the Amish Complex Disease Research Program cohort exhibited significantly lower postprandial serum triglycerides, suggestive of a role for TM6SF2 in the small intestine. To gain further insight into this putative role, effects of TM6SF2 deficiency were studied in a zebrafish model and in cultured human Caco-2 enterocytes. In both systems TM6SF2 deficiency resulted in defects in small intestine metabolism in response to dietary lipids, including significantly increased lipid accumulation, decreased lipid clearance, and increased endoplasmic reticulum stress. CONCLUSIONS These data strongly support a role of TM6SF2 in the regulation of postprandial lipemia, potentially through a similar function for TM6SF2 in the lipidation and/or export of both hepatically and intestinally derived triglyceride-rich lipoproteins. (Hepatology 2017;65:1526-1542).
Collapse
Affiliation(s)
- Elizabeth A. O’Hare
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Rongze Yang
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Laura Yerges-Armstrong
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Urmilla Sreenivasan
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Rebecca McFarland
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Carmen C. Leitch
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Meredith H. Wilson
- Carnegie Institution for Science, Department of Embryology, Baltimore, MD 21218, USA
| | - Shilpa Narina
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Alexis Gorden
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Kathy Ryan
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Alan R. Shuldiner
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Steve A. Farber
- Carnegie Institution for Science, Department of Embryology, Baltimore, MD 21218, USA
| | - G. Craig Wood
- Geisinger Clinic, Geisinger Obesity Research Institute, Danville PA 17822, USA
| | | | - Glenn S. Gerhard
- Geisinger Clinic, Geisinger Obesity Research Institute, Danville PA 17822, USA
| | - Janet D. Robishaw
- Geisinger Clinic, Geisinger Obesity Research Institute, Danville PA 17822, USA
| | - Carole Sztalryd
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Baltimore VA Medical Center, VA Research Service, Geriatric Research, Education and Clinical Center (GRECC) and VA Maryland Health Care System, 10N Green Street Baltimore 21201, USA
| | - Norann A. Zaghloul
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
40
|
Seebacher F, Tallis J, McShea K, James RS. Obesity-induced decreases in muscle performance are not reversed by weight loss. Int J Obes (Lond) 2017; 41:1271-1278. [PMID: 28337027 DOI: 10.1038/ijo.2017.81] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 02/22/2017] [Accepted: 03/14/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND/OBJECTIVES Obesity can affect muscle phenotypes, and may thereby constrain movement and energy expenditure. Weight loss is a common and intuitive intervention for obesity, but it is not known whether the effects of obesity on muscle function are reversible by weight loss. Here we tested whether obesity-induced changes in muscle metabolic and contractile phenotypes are reversible by weight loss. SUBJECTS/METHODS We used zebrafish (Danio rerio) in a factorial design to compare energy metabolism, locomotor capacity, muscle isometric force and work-loop power output, and myosin heavy chain (MHC) composition between lean fish, diet-induced obese fish, and fish that were obese and then returned to lean body mass following diet restriction. RESULTS Obesity increased resting metabolic rates (P<0.001) and decreased maximal metabolic rates (P=0.030), but these changes were reversible by weight loss, and were not associated with changes in muscle citrate synthase activity. In contrast, obesity-induced decreases in locomotor performance (P=0.0034), and isolated muscle isometric stress (P=0.01), work-loop power output (P<0.001) and relaxation rates (P=0.012) were not reversed by weight loss. Similarly, obesity-induced decreases in concentrations of fast and slow MHCs, and a shift toward fast MHCs were not reversed by weight loss. CONCLUSION Obesity-induced changes in locomotor performance and muscle contractile function were not reversible by weight loss. These results show that weight loss alone may not be a sufficient intervention.
Collapse
Affiliation(s)
- F Seebacher
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - J Tallis
- Centre for Applied Biological and Exercise Sciences, Coventry University, Coventry, UK
| | - K McShea
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - R S James
- Centre for Applied Biological and Exercise Sciences, Coventry University, Coventry, UK
| |
Collapse
|
41
|
Leulier F, MacNeil LT, Lee WJ, Rawls JF, Cani PD, Schwarzer M, Zhao L, Simpson SJ. Integrative Physiology: At the Crossroads of Nutrition, Microbiota, Animal Physiology, and Human Health. Cell Metab 2017; 25:522-534. [PMID: 28273475 PMCID: PMC6200423 DOI: 10.1016/j.cmet.2017.02.001] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/30/2017] [Accepted: 01/31/2017] [Indexed: 02/07/2023]
Abstract
Nutrition is paramount in shaping all aspects of animal biology. In addition, the influence of the intestinal microbiota on physiology is now widely recognized. Given that diet also shapes the intestinal microbiota, this raises the question of how the nutritional environment and microbial assemblages together influence animal physiology. This research field constitutes a new frontier in the field of organismal biology that needs to be addressed. Here we review recent studies using animal models and humans and propose an integrative framework within which to define the study of the diet-physiology-microbiota systems and ultimately link it to human health. Nutritional Geometry sits centrally in the proposed framework and offers means to define diet compositions that are optimal for individuals and populations.
Collapse
Affiliation(s)
- François Leulier
- Institut de Génomique Fonctionnelle de Lyon (IGFL), Université de Lyon, Ecole Normale Supérieure de Lyon, CNRS UMR 5242, Université Claude Bernard Lyon 1, F-69364 Lyon Cedex 07, France.
| | - Lesley T MacNeil
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S4K1, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S4K1, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S4K1, Canada
| | - Won-Jae Lee
- School of Biological Science, Institute of Molecular Biology and Genetics, National Creative Research Initiative Center for Hologenomics, Seoul National University, Seoul 151-742, South Korea
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Center for the Genomics of Microbial Systems, Duke University School of Medicine, Durham, NC 27710, USA
| | - Patrice D Cani
- Université Catholique de Louvain, Louvain Drug Research Institute, WELBIO (Walloon Excellence in Life Sciences and Biotechnology), Metabolism and Nutrition Research Group, B-1200 Brussels, Belgium
| | - Martin Schwarzer
- Institut de Génomique Fonctionnelle de Lyon (IGFL), Université de Lyon, Ecole Normale Supérieure de Lyon, CNRS UMR 5242, Université Claude Bernard Lyon 1, F-69364 Lyon Cedex 07, France
| | - Liping Zhao
- State Key Laboratory of Microbial Metabolism and Ministry of Education Key Laboratory of Systems Biomedicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Stephen J Simpson
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia; School of Life and Environmental Sciences, University of Sydney, Sydney 2006, Australia
| |
Collapse
|
42
|
Elmonem MA, Khalil R, Khodaparast L, Khodaparast L, Arcolino FO, Morgan J, Pastore A, Tylzanowski P, Ny A, Lowe M, de Witte PA, Baelde HJ, van den Heuvel LP, Levtchenko E. Cystinosis (ctns) zebrafish mutant shows pronephric glomerular and tubular dysfunction. Sci Rep 2017; 7:42583. [PMID: 28198397 PMCID: PMC5309805 DOI: 10.1038/srep42583] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 01/12/2017] [Indexed: 01/05/2023] Open
Abstract
The human ubiquitous protein cystinosin is responsible for transporting the disulphide amino acid cystine from the lysosomal compartment into the cytosol. In humans, Pathogenic mutations of CTNS lead to defective cystinosin function, intralysosomal cystine accumulation and the development of cystinosis. Kidneys are initially affected with generalized proximal tubular dysfunction (renal Fanconi syndrome), then the disease rapidly affects glomeruli and progresses towards end stage renal failure and multiple organ dysfunction. Animal models of cystinosis are limited, with only a Ctns knockout mouse reported, showing cystine accumulation and late signs of tubular dysfunction but lacking the glomerular phenotype. We established and characterized a mutant zebrafish model with a homozygous nonsense mutation (c.706 C > T; p.Q236X) in exon 8 of ctns. Cystinotic mutant larvae showed cystine accumulation, delayed development, and signs of pronephric glomerular and tubular dysfunction mimicking the early phenotype of human cystinotic patients. Furthermore, cystinotic larvae showed a significantly increased rate of apoptosis that could be ameliorated with cysteamine, the human cystine depleting therapy. Our data demonstrate that, ctns gene is essential for zebrafish pronephric podocyte and proximal tubular function and that the ctns-mutant can be used for studying the disease pathogenic mechanisms and for testing novel therapies for cystinosis.
Collapse
Affiliation(s)
- Mohamed A Elmonem
- Department of Paediatric Nephrology &Growth and Regeneration, University Hospitals Leuven KU Leuven - University of Leuven, Leuven, Belgium.,Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ramzi Khalil
- Department of Pathology, Leiden University Medical Centre, The Netherlands
| | - Ladan Khodaparast
- Department of Cellular and Molecular Medicine, Switch Laboratory, VIB, University Hospitals Leuven KU Leuven - University of Leuven, Leuven, Belgium
| | - Laleh Khodaparast
- Department of Cellular and Molecular Medicine, Switch Laboratory, VIB, University Hospitals Leuven KU Leuven - University of Leuven, Leuven, Belgium
| | - Fanny O Arcolino
- Department of Paediatric Nephrology &Growth and Regeneration, University Hospitals Leuven KU Leuven - University of Leuven, Leuven, Belgium
| | - Joseph Morgan
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Anna Pastore
- Laboratory of Proteomics and Metabolomics, Children's Hospital and Research Institute "Bambino Gesù" IRCCS, Rome, Italy
| | - Przemko Tylzanowski
- Department of Development and Regeneration, Laboratory for Developmental and Stem Cell Biology, Skeletal Biology and Engineering Research Centre, KU Leuven - University of Leuven, Leuven, Belgium.,Department of Biochemistry and Molecular Biology, Medical University, Lublin, Poland
| | - Annelii Ny
- Laboratory for Molecular Bio-discovery, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven - University of Leuven, Leuven, Belgium
| | - Martin Lowe
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Peter A de Witte
- Laboratory for Molecular Bio-discovery, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven - University of Leuven, Leuven, Belgium
| | - Hans J Baelde
- Department of Pathology, Leiden University Medical Centre, The Netherlands
| | - Lambertus P van den Heuvel
- Department of Paediatric Nephrology &Growth and Regeneration, University Hospitals Leuven KU Leuven - University of Leuven, Leuven, Belgium.,Department of Paediatric Nephrology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Elena Levtchenko
- Department of Paediatric Nephrology &Growth and Regeneration, University Hospitals Leuven KU Leuven - University of Leuven, Leuven, Belgium
| |
Collapse
|
43
|
Burggren WW, Dubansky B, Bautista NM. Cardiovascular Development in Embryonic and Larval Fishes. ACTA ACUST UNITED AC 2017. [DOI: 10.1016/bs.fp.2017.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
44
|
Caro M, Sansone A, Amezaga J, Navarro V, Ferreri C, Tueros I. Wine lees modulate lipid metabolism and induce fatty acid remodelling in zebrafish. Food Funct 2017; 8:1652-1659. [DOI: 10.1039/c6fo01754a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
This study investigates the ability of a polyphenolic extract obtained from a wine lees by-product to modulate zebrafish lipid metabolism.
Collapse
Affiliation(s)
- M. Caro
- New Foods
- AZTI
- 48160 Derio
- Spain
| | | | | | - V. Navarro
- Department of Pharmacy and Food Sciences
- University of the Basque Country
- 01006 Vitoria-Gasteiz
- Spain
| | | | | |
Collapse
|
45
|
Noyes PD, Garcia GR, Tanguay RL. ZEBRAFISH AS AN IN VIVO MODEL FOR SUSTAINABLE CHEMICAL DESIGN. GREEN CHEMISTRY : AN INTERNATIONAL JOURNAL AND GREEN CHEMISTRY RESOURCE : GC 2016; 18:6410-6430. [PMID: 28461781 PMCID: PMC5408959 DOI: 10.1039/c6gc02061e] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Heightened public awareness about the many thousands of chemicals in use and present as persistent contaminants in the environment has increased the demand for safer chemicals and more rigorous toxicity testing. There is a growing recognition that the use of traditional test models and empirical approaches is impractical for screening for toxicity the many thousands of chemicals in the environment and the hundreds of new chemistries introduced each year. These realities coupled with the green chemistry movement have prompted efforts to implement more predictive-based approaches to evaluate chemical toxicity early in product development. While used for many years in environmental toxicology and biomedicine, zebrafish use has accelerated more recently in genetic toxicology, high throughput screening (HTS), and behavioral testing. This review describes major advances in these testing methods that have positioned the zebrafish as a highly applicable model in chemical safety evaluations and sustainable chemistry efforts. Many toxic responses have been shown to be shared among fish and mammals owing to their generally well-conserved development, cellular networks, and organ systems. These shared responses have been observed for chemicals that impair endocrine functioning, development, and reproduction, as well as those that elicit cardiotoxicity and carcinogenicity, among other diseases. HTS technologies with zebrafish enable screening large chemical libraries for bioactivity that provide opportunities for testing early in product development. A compelling attribute of the zebrafish centers on being able to characterize toxicity mechanisms across multiple levels of biological organization from the genome to receptor interactions and cellular processes leading to phenotypic changes such as developmental malformations. Finally, there is a growing recognition of the links between human and wildlife health and the need for approaches that allow for assessment of real world multi-chemical exposures. The zebrafish is poised to be an important model in bridging these two conventionally separate areas of toxicology and characterizing the biological effects of chemical mixtures that could augment its role in sustainable chemistry.
Collapse
Affiliation(s)
- Pamela D. Noyes
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331
| | - Gloria R. Garcia
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331
| | - Robert L. Tanguay
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331
| |
Collapse
|
46
|
Masud S, Torraca V, Meijer AH. Modeling Infectious Diseases in the Context of a Developing Immune System. Curr Top Dev Biol 2016; 124:277-329. [PMID: 28335862 DOI: 10.1016/bs.ctdb.2016.10.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Zebrafish has been used for over a decade to study the mechanisms of a wide variety of inflammatory disorders and infections, with models ranging from bacterial, viral, to fungal pathogens. Zebrafish has been especially relevant to study the differentiation, specialization, and polarization of the two main innate immune cell types, the macrophages and the neutrophils. The optical accessibility and the early appearance of myeloid cells that can be tracked with fluorescent labels in zebrafish embryos and the ability to use genetics to selectively ablate or expand immune cell populations have permitted studying the interaction between infection, development, and metabolism. Additionally, zebrafish embryos are readily colonized by a commensal flora, which facilitated studies that emphasize the requirement for immune training by the natural microbiota to properly respond to pathogens. The remarkable conservation of core mechanisms required for the recognition of microbial and danger signals and for the activation of the immune defenses illustrates the high potential of the zebrafish model for biomedical research. This review will highlight recent insight that the developing zebrafish has contributed to our understanding of host responses to invading microbes and the involvement of the microbiome in several physiological processes. These studies are providing a mechanistic basis for developing novel therapeutic approaches to control infectious diseases.
Collapse
Affiliation(s)
- Samrah Masud
- Institute of Biology, Leiden University, Leiden, The Netherlands
| | - Vincenzo Torraca
- Institute of Biology, Leiden University, Leiden, The Netherlands
| | | |
Collapse
|
47
|
Hugo SE, Schlegel A. A genetic screen for zebrafish mutants with hepatic steatosis identifies a locus required for larval growth. J Anat 2016; 230:407-413. [PMID: 27976367 DOI: 10.1111/joa.12570] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2016] [Indexed: 01/14/2023] Open
Abstract
In a screen for zebrafish larval mutants with excessive liver lipid accumulation (hepatic steatosis), we identified harvest moon (hmn). Cytoplasmic lipid droplets, surrounded by multivesicular structures and mitochondria whose cristae appeared swollen, are seen in hmn mutant hepatocytes. Whole body triacylglycerol is increased in hmn mutant larvae. When we attempted to raise mutants, which were morphologically normal at the developmental stage that the screen was conducted, to adulthood, we observed that most hmn mutants do not survive to the juvenile period when raised. An arrest in growth occurs in the late larval period without obvious organ defects. Maternal zygotic mutants have no additional defects, suggesting that the mutation affects a late developmental process. The developmental window between embryogenesis and the metamorphosis remains under-studied, and hmn mutants might be useful for exploring the molecular and anatomic processes occurring during this transition period.
Collapse
Affiliation(s)
- Sarah E Hugo
- University of Utah Molecular Medicine (U2M2) Program, University of Utah School of Medicine, Salt Lake City, UT, USA.,Division of Endocrinology, Metabolism and Diabetes, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Amnon Schlegel
- University of Utah Molecular Medicine (U2M2) Program, University of Utah School of Medicine, Salt Lake City, UT, USA.,Division of Endocrinology, Metabolism and Diabetes, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA.,Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA.,Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
48
|
Weger BD, Weger M, Görling B, Schink A, Gobet C, Keime C, Poschet G, Jost B, Krone N, Hell R, Gachon F, Luy B, Dickmeis T. Extensive Regulation of Diurnal Transcription and Metabolism by Glucocorticoids. PLoS Genet 2016; 12:e1006512. [PMID: 27941970 PMCID: PMC5191836 DOI: 10.1371/journal.pgen.1006512] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 12/27/2016] [Accepted: 11/29/2016] [Indexed: 12/31/2022] Open
Abstract
Altered daily patterns of hormone action are suspected to contribute to metabolic disease. It is poorly understood how the adrenal glucocorticoid hormones contribute to the coordination of daily global patterns of transcription and metabolism. Here, we examined diurnal metabolite and transcriptome patterns in a zebrafish glucocorticoid deficiency model by RNA-Seq, NMR spectroscopy and liquid chromatography-based methods. We observed dysregulation of metabolic pathways including glutaminolysis, the citrate and urea cycles and glyoxylate detoxification. Constant, non-rhythmic glucocorticoid treatment rescued many of these changes, with some notable exceptions among the amino acid related pathways. Surprisingly, the non-rhythmic glucocorticoid treatment rescued almost half of the entire dysregulated diurnal transcriptome patterns. A combination of E-box and glucocorticoid response elements is enriched in the rescued genes. This simple enhancer element combination is sufficient to drive rhythmic circadian reporter gene expression under non-rhythmic glucocorticoid exposure, revealing a permissive function for the hormones in glucocorticoid-dependent circadian transcription. Our work highlights metabolic pathways potentially contributing to morbidity in patients with glucocorticoid deficiency, even under glucocorticoid replacement therapy. Moreover, we provide mechanistic insight into the interaction between the circadian clock and glucocorticoids in the transcriptional regulation of metabolism.
Collapse
Affiliation(s)
- Benjamin D. Weger
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen, Germany
- Nestlé Institute of Health Sciences SA, EPFL Innovation Park, Bâtiment H, Lausanne, Switzerland
| | - Meltem Weger
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen, Germany
- Centre for Endocrinology, Diabetes and Metabolism, University of Birmingham, Birmingham, United Kingdom
| | - Benjamin Görling
- Institute for Organic Chemistry, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen, Germany
- Institute for Biological Interfaces 4 –Magnetic Resonance, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen, Germany
| | - Andrea Schink
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen, Germany
| | - Cédric Gobet
- Nestlé Institute of Health Sciences SA, EPFL Innovation Park, Bâtiment H, Lausanne, Switzerland
- Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Céline Keime
- Plateforme Biopuces et séquençage, IGBMC, 1 rue Laurent Fries, Parc d'Innovation, Illkirch, France
| | - Gernot Poschet
- Centre for Organismal Studies (COS), University of Heidelberg, Heidelberg, Germany
| | - Bernard Jost
- Plateforme Biopuces et séquençage, IGBMC, 1 rue Laurent Fries, Parc d'Innovation, Illkirch, France
| | - Nils Krone
- Academic Unit of Child Health, Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Rüdiger Hell
- Centre for Organismal Studies (COS), University of Heidelberg, Heidelberg, Germany
| | - Frédéric Gachon
- Nestlé Institute of Health Sciences SA, EPFL Innovation Park, Bâtiment H, Lausanne, Switzerland
- Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Burkhard Luy
- Institute for Organic Chemistry, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen, Germany
- Institute for Biological Interfaces 4 –Magnetic Resonance, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen, Germany
| | - Thomas Dickmeis
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen, Germany
- * E-mail:
| |
Collapse
|
49
|
Yan AF, Chen T, Chen S, Tang DS, Liu F, Jiang X, Huang W, Ren CH, Hu CQ. Signal transduction mechanism for glucagon-induced leptin gene expression in goldfish liver. Int J Biol Sci 2016; 12:1544-1554. [PMID: 27994518 PMCID: PMC5166495 DOI: 10.7150/ijbs.16612] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 10/12/2016] [Indexed: 12/24/2022] Open
Abstract
Leptin is a peripheral satiety hormone that also plays important roles in energy homeostasis in vertebrates ranging from fish to mammals. In teleost fish, however, the regulatory mechanism for leptin gene expression still remains unclear. In this study, we found that glucagon, a key hormone in glucose homeostasis, was effective at elevating the leptin-AI and leptin-AII transcript levels in goldfish liver via both in vivo intraperitoneal injection and in vitro cells incubation approaches. The responses of leptin-AI and leptin-AII mRNA to glucagon treatment were highly comparable. In contrast, blockade of local glucagon action could reduce the basal and induced leptin-AI and leptin-AII mRNA expression. The stimulation of leptin levels by glucagon was caused by the activation of adenylate cyclase (AC)/cyclic-AMP (cAMP)/ protein kinase A (PKA), and probably cAMP response element-binding protein (CREB) cascades. Our study described the effect and signal transduction mechanism of glucagon on leptin gene expression in goldfish liver, and may also provide new insight into leptin as a mediator in the regulatory network of energy metabolism in the fish model.
Collapse
Affiliation(s)
- Ai-Fen Yan
- School of stomatology and medicine, Foshan University, Foshan 528000, China
| | - Ting Chen
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB); South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510275, China
| | - Shuang Chen
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Dong-Sheng Tang
- School of stomatology and medicine, Foshan University, Foshan 528000, China
| | - Fang Liu
- School of stomatology and medicine, Foshan University, Foshan 528000, China
| | - Xiao Jiang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB)
| | - Wen Huang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB)
| | - Chun-Hua Ren
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB); South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510275, China
| | - Chao-Qun Hu
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB); South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510275, China
| |
Collapse
|
50
|
Yang S, Ott CJ, Rossmann MP, Superdock M, Zon LI, Zhou Y. Chromatin immunoprecipitation and an open chromatin assay in zebrafish erythrocytes. Methods Cell Biol 2016; 135:387-412. [PMID: 27443937 DOI: 10.1016/bs.mcb.2016.04.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Zebrafish is an excellent genetic and developmental model for the study of vertebrate development and disease. Its ability to produce an abundance of transparent, externally developed embryos has facilitated large-scale genetic and chemical screens for the identification of critical genes and chemical factors that modulate developmental pathways. These studies can have profound implications for the diagnosis and treatment of a variety of human diseases. Recent advancements in molecular and genomic studies have provided valuable tools and resources for comprehensive and high-resolution analysis of epigenomes during cell specification and lineage differentiation throughout development. In this chapter, we describe two simple methods to evaluate protein-DNA interaction and chromatin architecture in erythrocytes from adult zebrafish. These are chromatin immunoprecipitation coupled with next-generation sequencing (ChIP-seq) and an assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq). These techniques, together with gene expression profiling, are useful for analyzing epigenomic regulation of cell specification, differentiation, and function during zebrafish development in both normal and disease models.
Collapse
Affiliation(s)
- S Yang
- Boston Children's Hospital, Boston, MA, United States; Dana Farber Cancer Institute, Harvard Stem Cell Institute, Boston, MA, United States; Harvard Medical School and Howard Hughes Medical Institute, Boston, MA, United States
| | - C J Ott
- Dana Farber Cancer Institute, Harvard Stem Cell Institute, Boston, MA, United States
| | - M P Rossmann
- Harvard University, Harvard, Cambridge, MA, United States
| | - M Superdock
- Boston Children's Hospital, Boston, MA, United States; Dana Farber Cancer Institute, Harvard Stem Cell Institute, Boston, MA, United States; Harvard Medical School and Howard Hughes Medical Institute, Boston, MA, United States
| | - L I Zon
- Boston Children's Hospital, Boston, MA, United States; Dana Farber Cancer Institute, Harvard Stem Cell Institute, Boston, MA, United States; Harvard Medical School and Howard Hughes Medical Institute, Boston, MA, United States; Harvard University, Harvard, Cambridge, MA, United States
| | - Y Zhou
- Boston Children's Hospital, Boston, MA, United States; Dana Farber Cancer Institute, Harvard Stem Cell Institute, Boston, MA, United States; Harvard Medical School and Howard Hughes Medical Institute, Boston, MA, United States; Harvard University, Harvard, Cambridge, MA, United States
| |
Collapse
|