1
|
Kleinendorst L, Siegelaar SE, Roessler HI, Meiwand L, van den Boogaard M, de Bruin-Bon RHACM, van Duinen KF, Planken RN, Jaspars EH, Kemperman PMJH, Bouma BJ, Nichols CG, Bekkenk MW, van Haaften GW, van Haelst MM. Treatment of overactive K ATP channels with glibenclamide in a zebrafish model and a clinical trial in humans with Cantú syndrome. Sci Rep 2025; 15:17704. [PMID: 40399303 PMCID: PMC12095479 DOI: 10.1038/s41598-025-00547-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 04/29/2025] [Indexed: 05/23/2025] Open
Abstract
This study explores the efficacy of glibenclamide, a KATP channel inhibitor, for treating Cantú syndrome (CS), a genetic disorder characterized by hypertrichosis and cardiovascular abnormalities. Treatment with glibenclamide for Cantú syndrome has only been reported in a single case report. In this study, we tested this repurposed drug in both a zebrafish model and an open-label trial with CS patients. CS zebrafish embryos, created using CRISPR/Cas9, were treated with glibenclamide. Their cardiac function was assessed using high-speed imaging. In the trial part of the study, four adults with CS used 2.5 mg glibenclamide daily for 8 months. Hypertrichosis, cardiac function, and edema were evaluated and glucose levels were monitored continuously. In the zebrafish model of CS glibenclamide reversed cardiac abnormalities. However, in the clinical trial, the effects on hypertrichosis were mixed, and there were no significant changes in cardiac phenotype or leg edema. One participant reported reduced facial erythema and puffiness, which relapsed post-trial. The treatment was generally safe, with multiple instances of level 1 hypoglycemia but no severe adverse events. In conclusion, glibenclamide can reverse cardiac abnormalities in a CS zebrafish model. Its effect on hypertrichosis and cardiovascular features in humans with CS are unclear and dosage increases are challenging due to hypoglycemia, which is important knowledge for treatment considerations in this rare genetic syndrome.Trial registration: EudraCT Number 2019-004651-36. Date of first registration 21/05/2021.
Collapse
Affiliation(s)
- Lotte Kleinendorst
- Department of Human Genetics, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Sarah E Siegelaar
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism Research Institute, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Helen I Roessler
- Emma Center for Personalized Medicine, Amsterdam UMC, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Lema Meiwand
- Department of Human Genetics, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Malou van den Boogaard
- Department of Cardiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Rianne H A C M de Bruin-Bon
- Department of Cardiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Kirsten F van Duinen
- Department of Dermatology, Ziekenhuis Nij Smellinghe, Compagnonsplein 1, Drachten, 9202 NN, The Netherlands
| | - R Nils Planken
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
- Department of Radiology, Mayo Clinic Rochester, 200 1st Street, Rochester, MN, 55905, USA
| | - Elisabeth H Jaspars
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Patrick M J H Kemperman
- Department of Dermatology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Berto J Bouma
- Department of Cardiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Colin G Nichols
- Department of Cell Biology and Physiology, Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St Louis, MO, 63110, USA
| | - Marcel W Bekkenk
- Department of Dermatology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Gijs W van Haaften
- Department of Genetics, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, The Netherlands
| | - Mieke M van Haelst
- Department of Human Genetics, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands.
- Emma Center for Personalized Medicine, Amsterdam UMC, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands.
| |
Collapse
|
2
|
Yan G, Wang Y, Yu L, Bo J, Fang H, Chen W, Zhang Y, Chen H, Hong Z. Depolymerization of oyster glycosaminoglycans for the enhancement of α-glucosidase inhibition and its application in hypoglycemic studies. Food Res Int 2025; 205:116008. [PMID: 40032484 DOI: 10.1016/j.foodres.2025.116008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 03/05/2025]
Abstract
Glycosaminoglycans are a class of naturally occurring macromolecular heteropolysaccharides with α-glucosidase inhibitory effects, which play an essential role in regulating postprandial hyperglycemia. However, the significant molecular weight hinders their inhibitory activity. In this study, oyster glycosaminoglycans were used for purification and depolymerization studies to obtain oyster glycosaminoglycan oligosaccharides (OGAG-Oli) and to study their inhibitory activity against α-glucosidase in vitro and hypoglycemic effect on hyperglycemic zebrafish in vivo. The results showed that the OGAG-Oli with optimal α-glucosidase inhibition obtained by the β-elimination mechanism of chondroitinase ABC had a molecular weight of about 2400 Da with five disaccharide repeating units, and their inhibitory effect on α-glucosidase was significantly enhanced with an IC50 value of 0.12 mg/mL, which was related to the strong binding capacity with α-glucosidase as evidenced by molecular docking. FT-IR and NMR results showed that the conformation of OGAG-Oli was predominantly in the α-configuration, which indicated that its inhibitory effect on α-glucosidase mainly depended on the α-glucosidic bond therein. The enzymatic depolymerization process of glycosaminoglycans was optimized, and the process conditions were determined as phosphate buffer solution pH 8.0, reaction time 3 h, reaction temperature 30 °C, chondroitin enzyme ABC activity 0.5 mU. The hypoglycemic ability of OGAG-Oli was evaluated in diabetic zebrafish, and the results showed that OGAG-Oli at a high-dose (0.12 mg/g, bw/d) could extremely significantly reduce fasting blood glucose in zebrafish, with an effect close to that of acarbose (p > 0.05), and exceptionally significantly (p < 0.01) reduced the elevation of TC, TG, and LDL-C caused by prolonged hyperglycemia, and extremely significantly (p < 0.01) increased the HDL-C level. This study showed that the structurally modified oyster glycosaminoglycans had intense α-glucosidase inhibitory activity, and exhibited biological activity of reducing glucose uptake and consequently improving body weight and lipid metabolism in diabetic zebrafish. As a bioactive ingredient of natural origin, this study provides a theoretical basis for the development and utilization of glycosaminoglycans with hypoglycemic effects in foods and drugs.
Collapse
Affiliation(s)
- Guangyu Yan
- Xiamen Ocean Vocational College, Xiamen 361022, China
| | - Yaru Wang
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, China; College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350001, China
| | - Lei Yu
- Xiamen Ocean Vocational College, Xiamen 361022, China
| | - Jun Bo
- Key Laboratory of Marine Ecological Conservation and Restoration, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, China
| | - Hua Fang
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, China; Xiamen Ocean Vocational College, Xiamen 361022, China
| | - Weizhu Chen
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, China; Xiamen Ocean Vocational College, Xiamen 361022, China
| | - Yiping Zhang
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, China; Xiamen Ocean Vocational College, Xiamen 361022, China
| | - Hui Chen
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, China; Xiamen Ocean Vocational College, Xiamen 361022, China.
| | - Zhuan Hong
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, China; Xiamen Ocean Vocational College, Xiamen 361022, China.
| |
Collapse
|
3
|
Schmitner N, Thumer S, Regele D, Mayer E, Bergerweiss I, Helker C, Stainier DYR, Meyer D, Kimmel RA. Conserved glucokinase regulation in zebrafish confirms therapeutic utility for pharmacologic modulation in diabetes. Commun Biol 2024; 7:1557. [PMID: 39580550 PMCID: PMC11585571 DOI: 10.1038/s42003-024-07264-5] [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: 04/17/2024] [Accepted: 11/13/2024] [Indexed: 11/25/2024] Open
Abstract
Glucokinase (GCK) is an essential enzyme for blood glucose homeostasis. Because of its importance in glucose metabolism, GCK is considered an attractive target for the development of antidiabetic drugs. However, a viable therapeutic agent has still to emerge, prompting efforts to improve understanding of the complex regulation and biological effects of GCK. Using the vertebrate organism zebrafish, an attractive model to study metabolic diseases and pharmacological responses, we dissected the complexities of gck regulation and unraveled effects of Gck modulation. We found that while gck expression in zebrafish islet cells is constitutive, gck expression in the liver is regulated by nutritional status, confirming similarity to the mammalian system. A combination of transgenic gck reporter lines and our diabetes model, the pdx1 mutant, allowed monitoring of gck expression under pathological conditions, revealing reduced gck expression and activity in the liver, which was unresponsive to nutrient stimulation, and decreased expression in the islet due to the reduced number of β-cells. Gck activation substantially ameliorated hyperglycemia in pdx1 mutants, without inducing oxidative stress responses in liver or islet. In-depth characterization of Gck activity and regulation at the cellular level in a whole-organism diabetes model clarifies its applicability as a drug target for therapies.
Collapse
Affiliation(s)
- Nicole Schmitner
- Institute of Molecular Biology, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria.
| | - Sophie Thumer
- Institute of Molecular Biology, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Dominik Regele
- Institute of Molecular Biology, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Elena Mayer
- Institute of Molecular Biology, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Ines Bergerweiss
- Institute of Molecular Biology, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Christian Helker
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Philipps-University Marburg, Marburg, Germany
| | | | - Dirk Meyer
- Institute of Molecular Biology, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Robin A Kimmel
- Institute of Molecular Biology, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
4
|
Kodama T, Watanabe S, Kayanuma I, Sasaki A, Kurokawa D, Baba O, Jimbo M, Furukawa F. Gluconeogenesis during development of the grass puffer (Takifugu niphobles). Comp Biochem Physiol A Mol Integr Physiol 2024; 295:111663. [PMID: 38735624 DOI: 10.1016/j.cbpa.2024.111663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/14/2024]
Abstract
During the development of teleost fish, the sole nutrient source is the egg yolk. The yolk consists mostly of proteins and lipids, with only trace amounts of carbohydrates such as glycogen and glucose. However, past evidence in some fishes showed transient increase in glucose during development, which may have supported the development of the embryos. Recently, we found in zebrafish that the yolk syncytial layer (YSL), an extraembryonic tissue surrounding the yolk, undergoes gluconeogenesis. However, in other teleost species, the knowledge on such gluconeogenic functions during early development is lacking. In this study, we used a marine fish, the grass puffer (Takifugu niphobles) and assessed possible gluconeogenic functions of their YSL, to understand the difference or shared features of gluconeogenesis between these species. A liquid chromatography (LC) / mass spectrometry (MS) analysis revealed that glucose and glycogen content significantly increased in the grass puffer during development. Subsequent real-time PCR results showed that most of the genes involved in gluconeogenesis increased in segmentation stages and/or during hatching. Among these genes, many were expressed in the YSL and liver, as shown by in situ hybridization analysis. In addition, glycogen immunostaining revealed that this carbohydrate source was accumulated in many tissues at segmentation stage but exclusively in the liver in hatched individuals. Taken together, these results suggest that developing grass puffer undergoes gluconeogenesis and glycogen synthesis during development, and that gluconeogenic activity is shared in YSL of zebrafish and grass puffer.
Collapse
Affiliation(s)
- Takafumi Kodama
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Seiya Watanabe
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Isana Kayanuma
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Akira Sasaki
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Daisuke Kurokawa
- Misaki Marine Biological Station, Graduate School of Science, The University of Tokyo, 1024 Koajiro, Misaki, Miura, Kanagawa 238-0225, Japan
| | - Otto Baba
- Oral and Maxillofacial Anatomy, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan
| | - Mitsuru Jimbo
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Fumiya Furukawa
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan.
| |
Collapse
|
5
|
Gao Y, Wu Y, Tie F, Wang H. Stilbenoids from fenugreek seeds alleviate insulin resistance by regulating the PI3K/AKT/mTOR signaling pathway in a type 2 diabetes zebrafish model. Heliyon 2024; 10:e32007. [PMID: 39040253 PMCID: PMC11260975 DOI: 10.1016/j.heliyon.2024.e32007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 07/24/2024] Open
Abstract
Insulin resistance (IR) is the main cause of type 2 diabetes mellitus (T2DM). The specific targets and underlying mechanisms responsible for the ameliorative effects of the stilbenoid compounds found in fenugreek seeds for ameliorating IR require further study. Here, we were predicted by using the network pharmacology prediction, molecular docking and molecular dynamics simulation approach the targets in common and the potential mechanismsof three stilbenoid compounds (rhaponticin, desoxyrhaponticin, and rhapontigenin) in relation to T2DM and IR. The results showed that the compounds may improve IR through the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) signaling pathway. Molecular docking studies revealed that they exhibit high binding affinity with the structural domains of peroxisome proliferator-activated receptor gamma (PPARG), glyceraldehyde-3-phosphate dehydrogenase (GAPDH), PI3K, and AKT. These results suggest that PPARG and GAPDH may be the potential targets for these three compounds in the treatment of T2DM.Subsequently, experiments using the zebrafish T2DM model showed that the stilbenoid compounds had varying degrees of efficacy in improving IR through the PI3K/AKT/mTOR signaling pathway, and rhaponticin had the most promising effects. The findings implicate a potential mechanism of action for the three stilbenoid compounds in enhancing insulin resistance (IR) through modulation of the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Yidan Gao
- Key Laboratory of Tibetan Medicine Research, Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Science, Xining, 810008, PR China
| | - Yun Wu
- Key Laboratory of Tibetan Medicine Research, Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Science, Xining, 810008, PR China
| | - Fangfang Tie
- Key Laboratory of Tibetan Medicine Research, Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Science, Xining, 810008, PR China
| | - Honglun Wang
- Key Laboratory of Tibetan Medicine Research, Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Science, Xining, 810008, PR China
| |
Collapse
|
6
|
Xue J, Wu S, Zhu Q, Liu X, He Z, Ye W, Wang P, Wu F. Enrichment and purification of Torreya grandis peptides by macroporous resin and its hypoglycemic mechanism revealed by transcriptome analysis. INDUSTRIAL CROPS AND PRODUCTS 2024; 213:118445. [DOI: 10.1016/j.indcrop.2024.118445] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
7
|
Juliana CA, Benjet J, De Leon DD. Characterization of the zebrafish as a model of ATP-sensitive potassium channel hyperinsulinism. BMJ Open Diabetes Res Care 2024; 12:e003735. [PMID: 38575153 PMCID: PMC11005463 DOI: 10.1136/bmjdrc-2023-003735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 01/25/2024] [Indexed: 04/06/2024] Open
Abstract
INTRODUCTION Congenital hyperinsulinism (HI) is the leading cause of persistent hypoglycemia in infants. Current models to study the most common and severe form of HI resulting from inactivating mutations in the ATP-sensitive potassium channel (KATP) are limited to primary islets from patients and the Sur1 -/- mouse model. Zebrafish exhibit potential as a novel KATPHI model since they express canonical insulin secretion pathway genes and those with identified causative HI mutations. Moreover, zebrafish larvae transparency provides a unique opportunity for in vivo visualization of pancreatic islets. RESEARCH DESIGN AND METHODS We evaluated zebrafish as a model for KATPHI using a genetically encoded Ca2+ sensor (ins:gCaMP6s) expressed under control of the insulin promoter in beta cells of an abcc8 -/- zebrafish line. RESULTS We observed significantly higher islet cytosolic Ca2+ in vivo in abcc8 -/- compared with abcc8 +/+ zebrafish larvae. Additionally, abcc8 -/- larval zebrafish had significantly lower whole body glucose and higher whole body insulin levels compared with abcc8 +/+ controls. However, adult abcc8 -/- zebrafish do not show differences in plasma glucose, plasma insulin, or glucose tolerance when compared with abcc8 +/+ zebrafish. CONCLUSIONS Our results identify that zebrafish larvae, but not adult fish, are a demonstrable novel model for advancement of HI research.
Collapse
Affiliation(s)
- Christine A Juliana
- Congenital Hyperinsulinism Center, Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Joshua Benjet
- Congenital Hyperinsulinism Center, Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Diva D De Leon
- Congenital Hyperinsulinism Center, Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Furukawa F, Aoyagi A, Sano K, Sameshima K, Goto M, Tseng YC, Ikeda D, Lin CC, Uchida K, Okumura SI, Yasumoto K, Jimbo M, Hwang PP. Gluconeogenesis in the extraembryonic yolk syncytial layer of the zebrafish embryo. PNAS NEXUS 2024; 3:pgae125. [PMID: 38585339 PMCID: PMC10997050 DOI: 10.1093/pnasnexus/pgae125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 03/11/2024] [Indexed: 04/09/2024]
Abstract
Yolk-consuming (lecithotrophic) embryos of oviparous animals, such as those of fish, need to make do with the maternally derived yolk. However, in many cases, yolk possesses little carbohydrates and sugars, including glucose, the essential monosaccharide. Interestingly, increases in the glucose content were found in embryos of some teleost fishes; however, the origin of this glucose has been unknown. Unveiling new metabolic strategies in fish embryos has a potential for better aquaculture technologies. In the present study, using zebrafish, we assessed how these embryos obtain the glucose. We employed stable isotope (13C)-labeled substrates and injected them to the zebrafish embryos. Our liquid chromatography-mass spectrometry-based isotope tracking revealed that among all tested substrate, glutamate was most actively metabolized to produce glucose in the zebrafish embryos. Expression analysis for gluconeogenic genes found that many of these were expressed in the yolk syncytial layer (YSL), an extraembryonic tissue found in teleost fishes. Generation 0 (G0) knockout of pck2, a gene encoding the key enzyme for gluconeogenesis from Krebs cycle intermediates, reduced gluconeogenesis from glutamate, suggesting that this gene is responsible for gluconeogenesis from glutamate in the zebrafish embryos. These results showed that teleost YSL undergoes gluconeogenesis, likely contributing to the glucose supplementation to the embryos with limited glucose source. Since many other animal lineages lack YSL, further comparative analysis will be interesting.
Collapse
Affiliation(s)
- Fumiya Furukawa
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Sec. 2, Nankang, Taipei 11529, Taiwan ROC
| | - Akihiro Aoyagi
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Kaori Sano
- Department of Chemistry, Faculty of Science, Josai University, 1-1 Keyakidai, Sakado, Saitama 350-0295, Japan
| | - Keita Sameshima
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Miku Goto
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Yung-Che Tseng
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Sec. 2, Nankang, Taipei 11529, Taiwan ROC
| | - Daisuke Ikeda
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Ching-Chun Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Sec. 2, Nankang, Taipei 11529, Taiwan ROC
| | - Katsuhisa Uchida
- Department of Marine Biology and Environmental Sciences, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuen Kibanadai-Nishi, Miyazaki 889-2192, Japan
| | - Sei-ichi Okumura
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Ko Yasumoto
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Mitsuru Jimbo
- School of Marine Biosciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Pung-Pung Hwang
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Sec. 2, Nankang, Taipei 11529, Taiwan ROC
| |
Collapse
|
9
|
Spratt J, Dias JM, Kolonelou C, Kiriako G, Engström E, Petrova E, Karampelias C, Cervenka I, Papanicolaou N, Lentini A, Reinius B, Andersson O, Ambrosetti E, Ruas JL, Teixeira AI. Multivalent insulin receptor activation using insulin-DNA origami nanostructures. NATURE NANOTECHNOLOGY 2024; 19:237-245. [PMID: 37813939 PMCID: PMC10873203 DOI: 10.1038/s41565-023-01507-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 08/15/2023] [Indexed: 10/11/2023]
Abstract
Insulin binds the insulin receptor (IR) and regulates anabolic processes in target tissues. Impaired IR signalling is associated with multiple diseases, including diabetes, cancer and neurodegenerative disorders. IRs have been reported to form nanoclusters at the cell membrane in several cell types, even in the absence of insulin binding. Here we exploit the nanoscale spatial organization of the IR to achieve controlled multivalent receptor activation. To control insulin nanoscale spatial organization and valency, we developed rod-like insulin-DNA origami nanostructures carrying different numbers of insulin molecules with defined spacings. Increasing the insulin valency per nanostructure markedly extended the residence time of insulin-DNA origami nanostructures at the receptors. Both insulin valency and spacing affected the levels of IR activation in adipocytes. Moreover, the multivalent insulin design associated with the highest levels of IR activation also induced insulin-mediated transcriptional responses more effectively than the corresponding monovalent insulin nanostructures. In an in vivo zebrafish model of diabetes, treatment with multivalent-but not monovalent-insulin nanostructures elicited a reduction in glucose levels. Our results show that the control of insulin multivalency and spatial organization with nanoscale precision modulates the IR responses, independent of the insulin concentration. Therefore, we propose insulin nanoscale organization as a design parameter in developing new insulin therapies.
Collapse
Affiliation(s)
- Joel Spratt
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - José M Dias
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Christina Kolonelou
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Georges Kiriako
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Enya Engström
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ekaterina Petrova
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Christos Karampelias
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Igor Cervenka
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Natali Papanicolaou
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Antonio Lentini
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Björn Reinius
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Olov Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Elena Ambrosetti
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Jorge L Ruas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ana I Teixeira
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
10
|
Li A, Yan C, Qiu J, Ji Y, Fu Y, Yan W. Adverse effects of plastic leachate and its component 2,4-DTBP on the early development of zebrafish embryos. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 904:167246. [PMID: 37741407 DOI: 10.1016/j.scitotenv.2023.167246] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/03/2023] [Accepted: 09/19/2023] [Indexed: 09/25/2023]
Abstract
Plastic waste has become a global environmental problem threatening the health of aquatic organisms especially via leachate. In this study, the test of zebrafish embryo showed adverse effects of leachate from some agricultural mulching films after UV light aging for 60 h. A typical phenolic antioxidant 2,4-di-tert-butylphenol (2,4-DTBP) was detected in the leachate and tested further for the zebrafish embryo biotoxicity. The microplastic leachate (6, 8 g/L, mass concentration measured by weight of plastic) increased the death and malformation rates, and reduced the hatching rate, heart rate, and body length of zebrafish larvae in the 96-hour early development period. Similar adverse effects were also caused by the 2,4-DTBP (0.01, 0.1, 1.0 mg/L, corresponding to 0.049, 0.49, and 4.85 μM) to some degree but could not completely explain the significant influences caused by the plastic leachate. Transcriptome analysis of zebrafish embryos exposed to the 2,4-DTBP for 96 h showed that the protein, fat, and carbohydrate digestion and absorption pathways, pancreatic secretion, PPAR signaling pathway, tryptophan metabolism, and adipocytokine signaling pathway were considerably down-regulated, but the cholesterol metabolism pathway was up-regulated in larval zebrafish. The altered transcriptional expression of mRNA at early development stage (96 h post fertilization) of zebrafish suggested that the 2,4-DTBP caused reduction of digestive capacity and pancreatic secretory function, and adversely affected processes associated with energy metabolism and glycolipid metabolism of larval zebrafish. This study helps us further understanding the effects of plastic leachate on the early development of fishes.
Collapse
Affiliation(s)
- Aifeng Li
- Key Laboratory of Marine Environment and Ecology, Ocean University of China, Ministry of Education, Qingdao 266100, China; College of Environmental Science and Engineering, Ocean University of China, Qingdao 266100, China.
| | - Chen Yan
- Key Laboratory of Marine Environment and Ecology, Ocean University of China, Ministry of Education, Qingdao 266100, China.
| | - Jiangbing Qiu
- Key Laboratory of Marine Environment and Ecology, Ocean University of China, Ministry of Education, Qingdao 266100, China; College of Environmental Science and Engineering, Ocean University of China, Qingdao 266100, China.
| | - Ying Ji
- Key Laboratory of Marine Environment and Ecology, Ocean University of China, Ministry of Education, Qingdao 266100, China.
| | - Yilei Fu
- Key Laboratory of Marine Environment and Ecology, Ocean University of China, Ministry of Education, Qingdao 266100, China.
| | - Wenhui Yan
- Key Laboratory of Marine Environment and Ecology, Ocean University of China, Ministry of Education, Qingdao 266100, China.
| |
Collapse
|
11
|
Wang K, Zhang R, Zhao X, Ma Y, Ren L, Ren Y, Chen G, Ye D, Wu J, Hu X, Guo Y, Xi R, Meng M, Yao Q, Li P, Chen Q, James TD. Reversible Recognition-Based Boronic Acid Probes for Glucose Detection in Live Cells and Zebrafish. J Am Chem Soc 2023. [PMID: 37023253 PMCID: PMC10119935 DOI: 10.1021/jacs.2c13694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Glucose, a critical source of energy, directly determines the homeostasis of the human body. However, due to the lack of robust imaging probes, the mechanism underlying the changes of glucose homeostasis in the human body remains unclear. Herein, diboronic acid probes with good biocompatibility and high sensitivity were synthesized based on an ortho-aminomethylphenylboronic acid probe, phenyl(di)boronic acid (PDBA). Significantly, by introducing the water-solubilizing group -CN directly opposite the boronic acid group and -COOCH3 or -COOH groups to the β site of the anthracene in PDBA, we obtained the water-soluble probe Mc-CDBA with sensitive response (F/F0 = 47.8, detection limit (LOD) = 1.37 μM) and Ca-CDBA with the highest affinity for glucose (Ka = 4.5 × 103 M-1). On this basis, Mc-CDBA was used to identify glucose heterogeneity between normal and tumor cells. Finally, Mc-CDBA and Ca-CDBA were used for imaging glucose in zebrafish. Our research provides a new strategy for designing efficient boronic acid glucose probes and powerful new tools for the evaluation of glucose-related diseases.
Collapse
Affiliation(s)
- Kai Wang
- Institute of Materia Medica, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, People's Republic of China
- Department of Chemistry, University of Bath, Bath BA2 7AY, U.K
| | - Ruixiao Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, People's Republic of China
| | - Xiujie Zhao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, People's Republic of China
| | - Yan Ma
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, People's Republic of China
| | - Lijuan Ren
- Institute of Materia Medica, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, People's Republic of China
| | - Youxiao Ren
- Institute of Materia Medica, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, People's Republic of China
| | - Gaofei Chen
- Institute of Materia Medica, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, People's Republic of China
| | - Dingming Ye
- Institute of Materia Medica, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, People's Republic of China
| | - Jinfang Wu
- Institute of Materia Medica, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, People's Republic of China
| | - Xinyuan Hu
- Institute of Materia Medica, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, People's Republic of China
| | - Yuanqiang Guo
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, People's Republic of China
| | - Rimo Xi
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, People's Republic of China
| | - Meng Meng
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, People's Republic of China
| | - Qingqiang Yao
- Institute of Materia Medica, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, People's Republic of China
| | - Ping Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University, Jinan 250014, People's Republic of China
| | - Qixin Chen
- Institute of Materia Medica, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, People's Republic of China
| | - Tony D James
- Department of Chemistry, University of Bath, Bath BA2 7AY, U.K
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, People's Republic of China
| |
Collapse
|
12
|
Research Progress on the Construction and Application of a Diabetic Zebrafish Model. Int J Mol Sci 2023; 24:ijms24065195. [PMID: 36982274 PMCID: PMC10048833 DOI: 10.3390/ijms24065195] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023] Open
Abstract
Diabetes is a metabolic disease characterized by high blood glucose levels. With economic development and lifestyle changes, the prevalence of diabetes is increasing yearly. Thus, it has become an increasingly serious public health problem in countries around the world. The etiology of diabetes is complex, and its pathogenic mechanisms are not completely clear. The use of diabetic animal models is helpful in the study of the pathogenesis of diabetes and the development of drugs. The emerging vertebrate model of zebrafish has many advantages, such as its small size, large number of eggs, short growth cycle, simple cultivation of adult fish, and effective improvement of experimental efficiency. Thus, this model is highly suitable for research as an animal model of diabetes. This review not only summarizes the advantages of zebrafish as a diabetes model, but also summarizes the construction methods and challenges of zebrafish models of type 1 diabetes, type 2 diabetes, and diabetes complications. This study provides valuable reference information for further study of the pathological mechanisms of diabetes and the research and development of new related therapeutic drugs.
Collapse
|
13
|
Sargent S, Brennan A, Clark JK. Regenerative potential and limitations in a zebrafish model of hyperglycemia-induced nerve degeneration. Dev Dyn 2023. [PMID: 36879394 DOI: 10.1002/dvdy.583] [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: 12/02/2021] [Revised: 02/04/2023] [Accepted: 02/25/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Previous work from our lab has described a model of motor nerve degeneration in hyperglycemic zebrafish larvae which resembles mammalian models of diabetic peripheral neuropathy (DPN). Here, we optimized the hyperglycemic-induction protocol, characterized deficits in nerve structure and behavioral function, and then examined the regenerative potential following recovery from the hyperglycemic state. RESULTS In agreement with our previous work, hyperglycemia induced motor nerve degeneration and behavioral deficits. However, the optimized protocol initiated disruption of tight junctions within the blood-nerve barrier, a phenotype apparent in mammalian models of DPN. Following a 10-day recovery period, regeneration of motor nerve components was apparent, but behavioral deficits persisted. We next examined the effect of hyperglycemia on the musculoskeletal system and found subtle deficits in muscle that resolved following recovery, and robust deficits in the skeletal system which persisted following recovery. CONCLUSION Here we optimized our previous model of hyperglycemia-induced motor nerve degeneration to more closely align with that observed in mammalian models and then characterized the regenerative potential following recovery from hyperglycemia. Notably, we observed striking impairments to skeletal development, which underscores the global impact hyperglycemia has across systems, and provides a framework for elucidating molecular mechanisms responsible for regenerative events moving forward.
Collapse
Affiliation(s)
- Sheridan Sargent
- Department of Biological Sciences, Salisbury University, Salisbury, Maryland, USA
| | - Anna Brennan
- Department of Biological Sciences, Salisbury University, Salisbury, Maryland, USA
| | | |
Collapse
|
14
|
Lin B, Ma J, Fang Y, Lei P, Wang L, Qu L, Wu W, Jin L, Sun D. Advances in Zebrafish for Diabetes Mellitus with Wound Model. Bioengineering (Basel) 2023; 10:bioengineering10030330. [PMID: 36978721 PMCID: PMC10044998 DOI: 10.3390/bioengineering10030330] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/01/2023] [Accepted: 03/04/2023] [Indexed: 03/08/2023] Open
Abstract
Diabetic foot ulcers cause great suffering and are costly for the healthcare system. Normal wound healing involves hemostasis, inflammation, proliferation, and remodeling. However, the negative factors associated with diabetes, such as bacterial biofilms, persistent inflammation, impaired angiogenesis, inhibited cell proliferation, and pathological scarring, greatly interfere with the smooth progress of the entire healing process. It is this impaired wound healing that leads to diabetic foot ulcers and even amputations. Therefore, drug screening is challenging due to the complexity of damaged healing mechanisms. The establishment of a scientific and reasonable animal experimental model contributes significantly to the in-depth research of diabetic wound pathology, prevention, diagnosis, and treatment. In addition to the low cost and transparency of the embryo (for imaging transgene applications), zebrafish have a discrete wound healing process for the separate study of each stage, resulting in their potential as the ideal model animal for diabetic wound healing in the future. In this review, we examine the reasons behind the delayed healing of diabetic wounds, systematically review various studies using zebrafish as a diabetic wound model by different induction methods, as well as summarize the challenges and improvement strategies which provide references for establishing a more reasonable diabetic wound zebrafish model.
Collapse
Affiliation(s)
- Bangchang Lin
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310000, China
| | - Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Yimeng Fang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Pengyu Lei
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Lei Wang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Linkai Qu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China
- Correspondence: (W.W.); (L.J.); (D.S.)
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
- Wenzhou City and WenZhouOuTai Medical Laboratory Co., Ltd. Joint Doctoral Innovation Station, Wenzhou Association for Science and Technology, Wenzhou 325000, China
- Correspondence: (W.W.); (L.J.); (D.S.)
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
- Correspondence: (W.W.); (L.J.); (D.S.)
| |
Collapse
|
15
|
Kim I, Seok SH, Lee HY. Development of a Zebrafish Larvae Model for Diabetic Heart Failure With Reduced Ejection Fraction. Korean Circ J 2022; 53:34-46. [PMID: 36627738 PMCID: PMC9834558 DOI: 10.4070/kcj.2022.0210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/19/2022] [Accepted: 11/09/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Diabetes mellitus (DM)-associated heart failure (HF) causes high morbidity and mortality. In this study, we established a zebrafish larvae model for in vivo research on diabetic HF. METHODS DM-like phenotypes were induced by treating zebrafish larvae with a combination of D-glucose (GLU) and streptozotocin (STZ). HF was induced by treatment with terfenadine (TER), a potassium channel blocker. Additionally, myocardial contractility, motility, and viability were evaluated. RESULTS The zebrafish larvae treated with a combination of GLU and STZ showed significantly higher whole-body glucose concentrations, lower insulin levels, and higher phosphoenolpyruvate carboxykinase levels, which are markers of abnormal glucose homeostasis, than the group treated with only GLU, with no effect on viability. When treated with TER, DM zebrafish showed significantly less myocardial fractional shortening and more irregular contractions than the non-DM zebrafish. Furthermore, in DM-HF with reduced ejection fraction (rEF) zebrafish, a significant increase in the levels of natriuretic peptide B, a HF biomarker, markedly reduced motility, and reduced survival rates were observed. CONCLUSIONS We established a DM-HFrEF zebrafish model by sequentially treating zebrafish larvae with GLU, STZ, and TER. Our findings indicate the potential utility of the developed zebrafish larvae model not only in screening studies of new drug candidates for DM-HFrEF but also in mechanistic studies to understand the pathophysiology of DM-HFrEF.
Collapse
Affiliation(s)
- Inho Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Seung Hyeok Seok
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Hae-Young Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
16
|
Lu K, Liang XF, Liu T, Cai W, Zhuang W, Zhang Y, Bibi A. DNA methylation of pck1 might contribute to the programming effects of early high-carbohydrate diets feeding to the glucose metabolism across two generations in zebrafish (Danio rerio). FISH PHYSIOLOGY AND BIOCHEMISTRY 2022; 48:1619-1633. [PMID: 36481836 DOI: 10.1007/s10695-022-01149-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 11/19/2022] [Indexed: 06/17/2023]
Abstract
The purpose of this study is to assess the effects of early high-carbohydrate stimulus on glucose metabolism in zebrafish (Danio rerio) over two generations and explore the mechanisms that explain those nutritional programming effects via epigenetic modifications. The larvae were delivered a high-carbohydrate diet (53.66%) that was used as an early nutritional stimulus from the first feeding to the end of the yolk sac (FF) and 5 days after yolk-sac exhaustion (YE). The larvae (F0) and their offspring (F1) were then both fed the control diet (22.69%) until adulthood (15 weeks), and they were challenged with a high-carbohydrate diet (35.36%) at the 16th week. The results indicated that early stimulus immediately raised the mRNA levels of genes involved in glycolysis and gluconeogenesis. At the end of F0 challenge, both treatment groups decreased the plasma glucose levels, increased the expression levels of glucokinase (gck), and inhibited the mRNA during gluconeogenesis. When challenged in F1, the glucose levels were lower in FF (F1), and the mRNA levels of phosphoenolpyruvate carboxykinase 1 (pck1) were decreased in FF (F1) and YE (F1). Besides, in both experimental groups (F0 and F1), the CpG island of pck1 maintained lower levels of hypermethylated expression from F0 adult, 24 h post-fertilization embryo, to F1 adult. In conclusion, these results indicated that an early high-carbohydrate stimulus could significantly reprogram glucose metabolism in adult zebrafish, that those modifications could be partially transmitted to the next generation, and that the DNA methylation of pck1 might work as a stable epigenetic marker to contribute to those processes.
Collapse
Affiliation(s)
- Ke Lu
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China
| | - Xu-Fang Liang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China.
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China.
| | - Tong Liu
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China
| | - Wenjing Cai
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China
| | - Wuyuan Zhuang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China
| | - Yanpeng Zhang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China
| | - Asima Bibi
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China
| |
Collapse
|
17
|
Keller RM, Beaver LM, Prater MC, Truong L, Tanguay RL, Stevens JF, Hord NG. Nitrate exposure reprograms hepatic amino acid and nutrient sensing pathways prior to exercise: A metabolomic and transcriptomic investigation in zebrafish (Danio rerio). Front Mol Biosci 2022; 9:903130. [PMID: 35928228 PMCID: PMC9343839 DOI: 10.3389/fmolb.2022.903130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Scope: Nitrate supplementation is a popular ergogenic aid that improves exercise performance by reducing oxygen consumption during exercise. We investigated the effect of nitrate exposure and exercise on metabolic pathways in zebrafish liver.Materials and methods: Fish were exposed to sodium nitrate (606.9 mg/L), or control water, for 21 days and analyzed at intervals during an exercise test. We utilized untargeted liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis and measured gene expression of 24 genes central to energy metabolism and redox signaling.Results: We observed a greater abundance of metabolites involved in endogenous nitric oxide (NO) metabolism and amino acid metabolism in nitrate-treated liver at rest, compared to rested controls. In the absence of exercise, nitrate treatment upregulated expression of genes central to nutrient sensing (pgc1a), protein synthesis (mtor) and purine metabolism (pnp5a and ampd1) and downregulated expression of genes involved in mitochondrial fat oxidation (acaca and cpt2).Conclusion: Our data support a role for sub-chronic nitrate treatment in the improvement of exercise performance, in part, by improving NO bioavailability, sparing arginine, and modulating hepatic gluconeogenesis and glycolytic capacity in the liver.
Collapse
Affiliation(s)
- Rosa M. Keller
- University of California, San Francisco, San Francisco, CA, United States
| | - Laura M. Beaver
- University of California, San Francisco, San Francisco, CA, United States
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Mary C. Prater
- Department of Foods and Nutrition, College of Family and Consumer Sciences, University of Georgia, Athens, GA, United States
| | - Lisa Truong
- Sinnhuber Aquatic Research Laboratory and the Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, United States
| | - Robyn L. Tanguay
- Sinnhuber Aquatic Research Laboratory and the Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, United States
| | - Jan F. Stevens
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
- College of Pharmacy, Oregon State University, Corvallis, OR, United States
| | - Norman G. Hord
- OU Health, Harold Hamm Diabetes Center, Department of Nutritional Sciences, College of Allied Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- *Correspondence: Norman G. Hord,
| |
Collapse
|
18
|
Fang Y, Wan JP, Zhang RJ, Sun F, Yang L, Zhao SX, Dong M, Song HD. Tpo knockout in zebrafish partially recapitulates clinical manifestations of congenital hypothyroidism and reveals the involvement of TH in proper development of glucose homeostasis. Gen Comp Endocrinol 2022; 323-324:114033. [PMID: 35367205 DOI: 10.1016/j.ygcen.2022.114033] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/26/2022]
Abstract
Congenital hypothyroidism (CH) is a highly prevalent but treatable neonatal endocrine disorder. Thyroid peroxidase (TPO) catalyzes key reactions in thyroid hormone (TH) synthesis. TPO mutations have been found to underlie approximately 5% of congenital hypothyroidism in Chinese patients with more severe phenotypes, the treatment of whom usually requires a higher dose of L-thyroxine. The Tpo gene of zebrafish has 66% homology with the human TPO gene, and synteny analysis has indicated that it is likely a human TPO ortholog. In this study, we generated a tpo-/- mutant zebrafish line through knockout of tpo with CRISPR/Cas9 and investigated the associated phenotypes. Tpo-/- mutant zebrafish displayed growth retardation; an increased number of thyroid follicular cells; and abnormal extrathyroidal phenotypes including pigmentation defects, erythema in the thoracic region, delayed scale development and failure of swim bladder secondary lobe formation. All these abnormal phenotypes were reversed by 30 nM thyroxine (T4) treatment starting at 1 month of age. Tpo-/- mutants also showed increased glucose levels during larval stages, and the increases were induced at least in part by increasing glucagon and decreasing insulin expression. Our work indicates that tpo-mutant zebrafish may serve as a human congenital hypothyroidism model for studying TPO- and TH-related disease mechanisms.
Collapse
Affiliation(s)
- Ya Fang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jia-Ping Wan
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Rui-Jia Zhang
- Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, China
| | - Feng Sun
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Liu Yang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Shuang-Xia Zhao
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Mei Dong
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Huai-Dong Song
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
19
|
High glucose-induced ROS-accumulation in embryo-larval stages of zebrafish leads to mitochondria-mediated apoptosis. Apoptosis 2022; 27:509-520. [PMID: 35596834 DOI: 10.1007/s10495-022-01731-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2022] [Indexed: 12/30/2022]
Abstract
In recent decades, diabetes mellitus has become a major chronic disease threatening human health worldwide, and the age of patients tends to be younger; however, the pathogenesis remains unclear, resulting in many difficulties in its treatment. As an ideal model animal, zebrafish can simulate the processes of human diabetes well. In this study, we successfully established a model of diabetic zebrafish larvae in a previous work. Furthermore, transcriptome analysis was completed, and the results suggested that 10.59% of differentially expressed genes (DEGs) related to the apoptosis pathway need to be considered. Then, glucose-induced developmental toxicity, reactive oxygen species (ROS) accumulation, antioxidant system function, apoptosis and mitochondrial dysfunction were measured in zebrafish larvae. We hope that this study will provide valuable reference information for type 2 juvenile diabetes treatment.
Collapse
|
20
|
Schmitner N, Recheis C, Thönig J, Kimmel RA. Differential Responses of Neural Retina Progenitor Populations to Chronic Hyperglycemia. Cells 2021; 10:cells10113265. [PMID: 34831487 PMCID: PMC8622914 DOI: 10.3390/cells10113265] [Citation(s) in RCA: 5] [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: 09/30/2021] [Revised: 11/08/2021] [Accepted: 11/18/2021] [Indexed: 12/30/2022] Open
Abstract
Diabetic retinopathy is a frequent complication of longstanding diabetes, which comprises a complex interplay of microvascular abnormalities and neurodegeneration. Zebrafish harboring a homozygous mutation in the pancreatic transcription factor pdx1 display a diabetic phenotype with survival into adulthood, and are therefore uniquely suitable among zebrafish models for studying pathologies associated with persistent diabetic conditions. We have previously shown that, starting at three months of age, pdx1 mutants exhibit not only vascular but also neuro-retinal pathologies manifesting as photoreceptor dysfunction and loss, similar to human diabetic retinopathy. Here, we further characterize injury and regenerative responses and examine the effects on progenitor cell populations. Consistent with a negative impact of hyperglycemia on neurogenesis, stem cells of the ciliary marginal zone show an exacerbation of aging-related proliferative decline. In contrast to the robust Müller glial cell proliferation seen following acute retinal injury, the pdx1 mutant shows replenishment of both rod and cone photoreceptors from slow-cycling, neurod-expressing progenitors which first accumulate in the inner nuclear layer. Overall, we demonstrate a diabetic retinopathy model which shows pathological features of the human disease evolving alongside an ongoing restorative process that replaces lost photoreceptors, at the same time suggesting an unappreciated phenotypic continuum between multipotent and photoreceptor-committed progenitors.
Collapse
|
21
|
Fakhlaei R, Selamat J, Razis AFA, Sukor R, Ahmad S, Amani Babadi A, Khatib A. In Vivo Toxicity Evaluation of Sugar Adulterated Heterotrigona itama Honey Using Zebrafish Model. Molecules 2021; 26:molecules26206222. [PMID: 34684803 PMCID: PMC8538600 DOI: 10.3390/molecules26206222] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 12/21/2022] Open
Abstract
Honey is prone to be adulterated through mixing with sugars, cheap and low-quality honey, and other adulterants. Consumption of adulterated honey may cause several health issues such as weight gain, diabetes, and liver and kidney dysfunction. Therefore, studying the impact of consumption of adulterated honey on consumers is critical since there is a lack of study in this field. Hence, the aims of this paper were: (1) to determine the lethal concentration (LC50) of adulterated honey using zebrafish embryo, (2) to elucidate toxicology of selected adulterated honey based on lethal dose (LD50) using adult zebrafish, (3) to determine the effects of adulterated honey on histological changes of zebrafish, and (4) to screen the metabolites profile of adulterated honey by using zebrafish blood serum. The LC50 of Heterotrigona itama honey (acacia honey) and its sugar adulterants (light corn sugar, cane sugar, inverted sugar, and palm sugar in the proportion of 1-3% (w/w) from the total volume) was determined by the toxicological assessment of honey samples on zebrafish embryos (different exposure concentrations in 24, 48, 72, and 96 h postfertilization (hpf)). Pure H. itama honey represents the LC50 of 34.40 ± 1.84 (mg/mL) at 96 hpf, while the inverted sugar represents the lowest LC50 (5.03 ± 0.92 mg/mL) among sugar adulterants. The highest concentration (3%) of sugar adulterants were used to study the toxicology of adulterated honey using adult zebrafish in terms of acute, prolong-acute, and sub-acute tests. The results of the LD50 from the sub-acute toxicity test of pure H. itama honey was 2.33 ± 0.24 (mg/mL). The histological studies of internal organs showed a lesion in the liver, kidney, and spleen of adulterated treated-honey groups compared to the control group. Furthermore, the LC-MS/MS results revealed three endogenous metabolites in both the pure and adulterated honey treated groups, as follows: (1) S-Cysteinosuccinic acid, (2) 2,3-Diphosphoglyceric acid, and (3) Cysteinyl-Tyrosine. The results of this study demonstrated that adulterated honey caused mortality, which contributes to higher toxicity, and also suggested that the zebrafish toxicity test could be a standard method for assessing the potential toxicity of other hazardous food additives. The information gained from this research will permit an evaluation of the potential risk associated with the consumption of adulterated compared to pure honey.
Collapse
Affiliation(s)
- Rafieh Fakhlaei
- Food Safety and Food Integrity (FOSFI), Institute of Tropical Agriculture and Food Security, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (R.F.); (R.S.)
| | - Jinap Selamat
- Food Safety and Food Integrity (FOSFI), Institute of Tropical Agriculture and Food Security, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (R.F.); (R.S.)
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
- Correspondence: ; Tel.: +60-38-9769-1099
| | - Ahmad Faizal Abdull Razis
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Rashidah Sukor
- Food Safety and Food Integrity (FOSFI), Institute of Tropical Agriculture and Food Security, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (R.F.); (R.S.)
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | - Syahida Ahmad
- Department of Biochemistry, Faculty of Biotechnology & Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | - Arman Amani Babadi
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 55469-14177, Iran;
| | - Alfi Khatib
- Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, Kuantan 25200, Pahang, Malaysia;
| |
Collapse
|
22
|
Tomazi R, Figueira ÂC, Ferreira AM, Ferreira DQ, de Souza GC, de Souza Pinheiro WB, Pinheiro Neto JR, da Silva GA, de Lima HB, da Silva Hage-Melim LI, Pereira ACM, Carvalho JCT, da Silva de Almeida SSM. Hypoglycemic Activity of Aqueous Extract of Latex from Hancornia speciosa Gomes: A Study in Zebrafish and In Silico. Pharmaceuticals (Basel) 2021; 14:ph14090856. [PMID: 34577555 PMCID: PMC8472165 DOI: 10.3390/ph14090856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/20/2021] [Accepted: 08/22/2021] [Indexed: 11/29/2022] Open
Abstract
Hancornia speciosa Gomes is a tree native to Brazil and has therapeutic potential for several diseases. Ethnopharmacological surveys have reported that the plant is used as a hypoglycemic agent and to lose weight. This study aimed to evaluate the effects of the aqueous extract from H. speciosa latex (LxHs) in a zebrafish model of diabetes. The extract was evaluated through high-performance thin-layer chromatography (HTPLC), nuclear magnetic resonance (NMR), and Fourier-transform infrared spectroscopy (FT-IR). We then tested treatments with LxHs (500, 1000, and 1500 mg/kg) by assessing blood glucose levels in alloxan-induced diabetic animals, and metformin was used as a control. The toxicity was evaluated through histopathology of the pancreas and biochemical assessment of serum levels of AST, ALT, creatinine, and urea. The extract was also assessed for acute toxicity through several parameters in embryos and adult animals. Finally, we performed in silico analysis through the SEA server and docking using the software GOLD. The phytochemical study showed the compounds cornoside, dihydrocornoide, and 1-O-methyl-myoinositol (bornesitol). The treatment with all doses of LxHs significantly decreased alloxan-induced hyperglycemia without any significant histological or biochemical abnormalities. No significant frequency of teratogenesis was observed in the embryos exposed to the extract, and no significant behavioral changes or deaths were observed in adult animals. In silico, the results showed a potential interaction between inositol and enzymes involved in carbohydrates’ metabolism. Overall, the results show a hypoglycemic activity of the extract in vivo, with no apparent toxicity. The computational studies suggest this could be at least partially due to the presence of bornesitol, since inositols can interact with carbohydrates’ enzymes.
Collapse
Affiliation(s)
- Rosana Tomazi
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia da Rede Bionorte (Ppg-Bionorte), Instituto Federal de Educação, Ciência e Tecnologia do Amapá (IFAP), Rodovia BR-210, km 03, S/n—Brasil Novo, Macapá 68909-398, AP, Brazil; (R.T.); (Â.C.F.)
- Laboratório de Pesquisa em Fármacos, Departamento de Ciências Biológicas e da Saúde, Universidade Federal do Amapá (UNIFAP), Rod. Juscelino Kubitschek, km 02—Jardim Marco Zero, Macapá 68903-419, AP, Brazil; (A.M.F.); (D.Q.F.); (G.C.d.S.); (A.C.M.P.)
- Laboratório de Farmacognosia e Fitoquímica, Departamento de Ciências Biológicas e da Saúde, Universidade Federal do Amapá (UNIFAP), Rod. Juscelino Kubitschek, km 02—Jardim Marco Zero, Macapá 68903-419, AP, Brazil;
| | - Ângela Costa Figueira
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia da Rede Bionorte (Ppg-Bionorte), Instituto Federal de Educação, Ciência e Tecnologia do Amapá (IFAP), Rodovia BR-210, km 03, S/n—Brasil Novo, Macapá 68909-398, AP, Brazil; (R.T.); (Â.C.F.)
| | - Adriana Maciel Ferreira
- Laboratório de Pesquisa em Fármacos, Departamento de Ciências Biológicas e da Saúde, Universidade Federal do Amapá (UNIFAP), Rod. Juscelino Kubitschek, km 02—Jardim Marco Zero, Macapá 68903-419, AP, Brazil; (A.M.F.); (D.Q.F.); (G.C.d.S.); (A.C.M.P.)
| | - Diego Quaresma Ferreira
- Laboratório de Pesquisa em Fármacos, Departamento de Ciências Biológicas e da Saúde, Universidade Federal do Amapá (UNIFAP), Rod. Juscelino Kubitschek, km 02—Jardim Marco Zero, Macapá 68903-419, AP, Brazil; (A.M.F.); (D.Q.F.); (G.C.d.S.); (A.C.M.P.)
| | - Gisele Custódio de Souza
- Laboratório de Pesquisa em Fármacos, Departamento de Ciências Biológicas e da Saúde, Universidade Federal do Amapá (UNIFAP), Rod. Juscelino Kubitschek, km 02—Jardim Marco Zero, Macapá 68903-419, AP, Brazil; (A.M.F.); (D.Q.F.); (G.C.d.S.); (A.C.M.P.)
| | - Wandson Braamcamp de Souza Pinheiro
- Laboratório de Química Industrial, Instituto de Química, Universidade Federal do Pará (UFPA), Rua. Augusto Corrêa, Guamá, 01, Belém 66075-110, AP, Brazil; (W.B.d.S.P.); (J.R.P.N.); (G.A.d.S.)
| | - José Rodrigues Pinheiro Neto
- Laboratório de Química Industrial, Instituto de Química, Universidade Federal do Pará (UFPA), Rua. Augusto Corrêa, Guamá, 01, Belém 66075-110, AP, Brazil; (W.B.d.S.P.); (J.R.P.N.); (G.A.d.S.)
| | - Geilson Alcantara da Silva
- Laboratório de Química Industrial, Instituto de Química, Universidade Federal do Pará (UFPA), Rua. Augusto Corrêa, Guamá, 01, Belém 66075-110, AP, Brazil; (W.B.d.S.P.); (J.R.P.N.); (G.A.d.S.)
| | - Henrique Barros de Lima
- Laboratório de Química Medicinal, Departamento de Ciências Biológicas e da Saúde, Universidade Federal do Amapá (UNIFAP), Rod. Juscelino Kubitschek, km 02—Jardim Marco Zero, Macapá 68903-419, AP, Brazil; (H.B.d.L.); (L.I.d.S.H.-M.)
| | - Lorane Izabel da Silva Hage-Melim
- Laboratório de Química Medicinal, Departamento de Ciências Biológicas e da Saúde, Universidade Federal do Amapá (UNIFAP), Rod. Juscelino Kubitschek, km 02—Jardim Marco Zero, Macapá 68903-419, AP, Brazil; (H.B.d.L.); (L.I.d.S.H.-M.)
| | - Arlindo César Matias Pereira
- Laboratório de Pesquisa em Fármacos, Departamento de Ciências Biológicas e da Saúde, Universidade Federal do Amapá (UNIFAP), Rod. Juscelino Kubitschek, km 02—Jardim Marco Zero, Macapá 68903-419, AP, Brazil; (A.M.F.); (D.Q.F.); (G.C.d.S.); (A.C.M.P.)
| | - José Carlos Tavares Carvalho
- Laboratório de Pesquisa em Fármacos, Departamento de Ciências Biológicas e da Saúde, Universidade Federal do Amapá (UNIFAP), Rod. Juscelino Kubitschek, km 02—Jardim Marco Zero, Macapá 68903-419, AP, Brazil; (A.M.F.); (D.Q.F.); (G.C.d.S.); (A.C.M.P.)
- Correspondence:
| | - Sheylla Susan Moreira da Silva de Almeida
- Laboratório de Farmacognosia e Fitoquímica, Departamento de Ciências Biológicas e da Saúde, Universidade Federal do Amapá (UNIFAP), Rod. Juscelino Kubitschek, km 02—Jardim Marco Zero, Macapá 68903-419, AP, Brazil;
| |
Collapse
|
23
|
Wang L, Kong H, Jin M, Li X, Stoika R, Lin H, Liu K. Synthesis of disaccharide modified berberine derivatives and their anti-diabetic investigation in zebrafish using a fluorescence-based technology. Org Biomol Chem 2021; 18:3563-3574. [PMID: 32347284 DOI: 10.1039/d0ob00327a] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Berberine is a naturally occurring isoquinoline alkaloid and has been used as an important functional food additive in China due to its various pharmacological activities. Berberine exhibits great potential for developing anti-diabetic agents against type 2 diabetes mellitus (T2DM), as it can reduce the blood glucose level in many animal models. However, the low anti-diabetic activity and poor bioavailability of berberine (below 5%) by oral administration significantly limit its practical applications. To solve these problems, this article focuses on the structural modification of berberine using some disaccharide groups, because the carbohydrate moiety has been proved to improve the bioavailability and enhance the receptor-binding affinity of drugs. Anti-diabetic investigation of the synthesized compounds was performed in a zebrafish model using a fluorescently labelled glucose analog 2-deoxy-2-[(7-nitro-2,1,3-benzoxadiazol-4-yl)amino]-d-glucose (2-NBDG) as a glucose tracker. The results indicated that the modification of berberine with carbohydrate groups could give derivatives with improved anti-diabetic activity, in particular the diglucose modified berberine derivative 1 which could dramatically promote the uptake of 2-NBDG in both zebrafish larvae and their eyes even at very low concentrations. Furthermore, the fluorescence-based anti-diabetic investigation method in zebrafish shows great potential for anti-diabetic drug screening.
Collapse
Affiliation(s)
- Lizhen Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, Shandong Province, China.
| | | | | | | | | | | | | |
Collapse
|
24
|
Characteristics of the New Insulin-Resistant Zebrafish Model. Pharmaceuticals (Basel) 2021; 14:ph14070642. [PMID: 34358068 PMCID: PMC8308799 DOI: 10.3390/ph14070642] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 01/09/2023] Open
Abstract
Insulin resistance, which occurs when insulin levels are sufficiently high over a prolonged period, causing the cells to fail to respond normally to the hormone. As a system for insulin resistance and diabetes drug development, insulin-resistant rodent models have been clearly established, but there is a limitation to high-throughput drug screening. Recently, zebrafish have been identified as an excellent system for drug discovery and identification of therapeutic targets, but studies on insulin resistance models have not been extensively performed. Therefore, we aimed to make a rapid insulin-resistant zebrafish model that complements the existing rodent models. To establish this model, zebrafish were treated with 10 μM insulin for 48 h. This model showed characteristics of insulin-resistant disease such as damaged pancreatic islets. Then we confirmed the recovery of the pancreatic islets after pioglitazone treatment. In addition, it was found that insulin-resistant drugs have as significant an effect in zebrafish as in humans, and these results proved the value of the zebrafish insulin resistance model for drug selection. In addition, RNA sequencing was performed to elucidate the mechanism involved. KEGG pathway enrichment analysis of differentially expressed genes showed that insulin resistance altered gene expression due to the MAPK signaling and calcium signaling pathways. This model demonstrates the utility of the zebrafish model for drug testing and drug discovery in insulin resistance and diabetes.
Collapse
|
25
|
Moortgat S, Manfroid I, Pendeville H, Freeman S, Bourdouxhe J, Benoit V, Merhi A, Philippe C, Faivre L, Maystadt I. Broadening the phenotypic spectrum and physiological insights related to EIF2S3 variants. Hum Mutat 2021; 42:827-834. [PMID: 33942450 DOI: 10.1002/humu.24215] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 04/09/2021] [Accepted: 04/27/2021] [Indexed: 01/20/2023]
Abstract
Mental deficiency, epilepsy, hypogonadism, microcephaly, and obesity syndrome is a severe X-linked syndrome caused by pathogenic variants in EIF2S3. The gene encodes the γ subunit of the eukaryotic translation initiation factor-2, eIF2, essential for protein translation. A recurrent frameshift variant is described in severely affected patients while missense variants usually cause a moderate phenotype. We identified a novel missense variant (c.433A>G, p.(Met145Val)) in EIF2S3 in a mildly affected patient. Studies on zebrafish confirm the pathogenicity of this novel variant and three previously published missense variants. CRISPR/Cas9 knockout of eif2s3 in zebrafish embryos recapitulate the human microcephaly and show increased neuronal cell death. Abnormal high glucose levels were identified in mutant embryos, caused by beta cell and pancreatic progenitor deficiency, not related to apoptosis. Additional studies in patient-derived fibroblasts did not reveal apoptosis. Our results provide new insights into disease physiopathology, suggesting tissue-dependent mechanisms.
Collapse
Affiliation(s)
- Stephanie Moortgat
- Center de Génétique Humaine, Institut de Pathologie et de Génétique, Gosselies, Belgium
| | - Isabelle Manfroid
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA-Research, Tour B34, Université de Liège, Liège (Sart-Tilman), Belgium
| | - Hélène Pendeville
- GIGA-Research, Zebrafish Platform, Tour B34, Université de Liège, Liège (Sart-Tilman), Belgium
| | - Stephen Freeman
- GIGA-Research, Imaging and Flow Cytometry Platform, Tour B34, Université de Liège, Liège (Sart-Tilman), Belgium
| | - Jordane Bourdouxhe
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA-Research, Tour B34, Université de Liège, Liège (Sart-Tilman), Belgium
| | - Valérie Benoit
- Center de Génétique Humaine, Institut de Pathologie et de Génétique, Gosselies, Belgium
| | - Ahmad Merhi
- Laboratory of Translational Oncology, Institut de Pathologie et de Génétique, Gosselies, Belgium.,IPG BioBank, Institut de Pathologie et de Génétique, 6041 Charleroi, Gosselies, Belgium
| | - Christophe Philippe
- Inserm UMR 1231 GAD, Genetics of Developmental disorders, Université de Bourgogne-Franche Comté, Dijon, France.,Unité Fonctionnelle « Innovation diagnostique dans les maladies rares », laboratoire de génétique moléculaire, plate-forme de biologie hospitalo-universitaire, CHU Dijon, Dijon, France
| | - Laurence Faivre
- Inserm UMR 1231 GAD, Genetics of Developmental disorders, Université de Bourgogne-Franche Comté, Dijon, France.,Center de Génétique et Center de Référence Maladies Rares « Anomalies du Développement et Syndromes Malformatifs de l'Interrégion Est », Hôpital d'Enfants, CHU, Dijon, France.,Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Centre Hospitalier Universitaire Dijon, Dijon, France
| | - Isabelle Maystadt
- Center de Génétique Humaine, Institut de Pathologie et de Génétique, Gosselies, Belgium.,Faculté de Médecine, URPhyM, UNamur, Namur, Belgium
| |
Collapse
|
26
|
MacDonald AJ, Yang YHC, Cruz AM, Beall C, Ellacott KLJ. Brain-Body Control of Glucose Homeostasis-Insights From Model Organisms. Front Endocrinol (Lausanne) 2021; 12:662769. [PMID: 33868184 PMCID: PMC8044781 DOI: 10.3389/fendo.2021.662769] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/12/2021] [Indexed: 12/15/2022] Open
Abstract
Tight regulation of blood glucose is essential for long term health. Blood glucose levels are defended by the correct function of, and communication between, internal organs including the gastrointestinal tract, pancreas, liver, and brain. Critically, the brain is sensitive to acute changes in blood glucose level and can modulate peripheral processes to defend against these deviations. In this mini-review we highlight select key findings showcasing the utility, strengths, and limitations of model organisms to study brain-body interactions that sense and control blood glucose levels. First, we discuss the large platform of genetic tools available to investigators studying mice and how this field may yet reveal new modes of communication between peripheral organs and the brain. Second, we discuss how rats, by virtue of their size, have unique advantages for the study of CNS control of glucose homeostasis and note that they may more closely model some aspects of human (patho)physiology. Third, we discuss the nascent field of studying the CNS control of blood glucose in the zebrafish which permits ease of genetic modification, large-scale measurements of neural activity and live imaging in addition to high-throughput screening. Finally, we briefly discuss glucose homeostasis in drosophila, which have a distinct physiology and glucoregulatory systems to vertebrates.
Collapse
Affiliation(s)
| | | | | | | | - Kate L. J. Ellacott
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom
| |
Collapse
|
27
|
Salehpour A, Rezaei M, Khoradmehr A, Tahamtani Y, Tamadon A. Which Hyperglycemic Model of Zebrafish ( Danio rerio) Suites My Type 2 Diabetes Mellitus Research? A Scoring System for Available Methods. Front Cell Dev Biol 2021; 9:652061. [PMID: 33791308 PMCID: PMC8005598 DOI: 10.3389/fcell.2021.652061] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/12/2021] [Indexed: 12/17/2022] Open
Abstract
Despite extensive studies on type 2 diabetes mellitus (T2DM), there is no definitive cure, drug, or prevention. Therefore, for developing new therapeutics, proper study models of T2DM is necessary to conduct further preclinical researches. Diabetes has been induced in animals using chemical, genetic, hormonal, antibody, viral, and surgical methods or a combination of them. Beside different approaches of diabetes induction, different animal species have been suggested. Although more than 85% of articles have proposed rat (genus Rattus) as the proper model for diabetes induction, zebrafish (Danio rerio) models of diabetes are being used more frequently in diabetes related studies. In this systematic review, we compare different aspects of available methods of inducing hyperglycemia referred as T2DM in zebrafish by utilizing a scoring system. Evaluating 26 approved models of T2DM in zebrafish, this scoring system may help researchers to compare different T2DM zebrafish models and select the best one regarding their own research theme. Eventually, glyoxalase1 (glo1-/-) knockout model of hyperglycemia achieved the highest score. In addition to assessment of hyperglycemic induction methods in zebrafish, eight most commonly proposed diabetic induction approval methods are suggested to help researchers confirm their subsequent proposed models.
Collapse
Affiliation(s)
- Aria Salehpour
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bandar Bushehr, Iran
| | - Mohammad Rezaei
- Department of Diabetes, Obesity and Metabolism, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, Iran
| | - Arezoo Khoradmehr
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bandar Bushehr, Iran
| | - Yaser Tahamtani
- Department of Diabetes, Obesity and Metabolism, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, Iran
- Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, Academic Center for Education, Culture and Research, Tehran, Iran
| | - Amin Tamadon
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bandar Bushehr, Iran
- Center of Marine Experimental and Comparative Medicine, The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bandar Bushehr, Iran
| |
Collapse
|
28
|
Gyimah E, Dong X, Xu H, Zhang Z, Mensah JK. Embryonic Exposure to Low Concentrations of Bisphenol A and S Altered Genes Related to Pancreatic β-Cell Development and DNA Methyltransferase in Zebrafish. ARCHIVES OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2021; 80:450-460. [PMID: 33471154 DOI: 10.1007/s00244-021-00812-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 01/02/2021] [Indexed: 06/12/2023]
Abstract
Bisphenol A (BPA) and bisphenol S (BPS) are implicated in the development of metabolic disorders, such diabetes mellitus. However, the epigenetic mechanism underlying the pancreatic β-cell dysregulation for both BPA/BPS needs clarification. This exploratory study was designed to investigate whether embryonic exposure to low BPA/BPS concentrations impair early pancreatic β-cell differentiation as well as DNA methylation in its gene expression profile using an in vivo model, zebrafish. Zebrafish embryos were exposed to 0, 0.01, 0.03, 0.1, 0.3, and 1.0 µM BPA/BPS at 4-h post fertilization (hpf) until 120 hpf. BPA/BPS-induced effects on pancreatic-related genes, insulin gene, and DNA methylation-associated genes were assessed at developmental stages (24-120 hpf), while glucose level was measure at the 120 hpf. The insulin expression levels decreased at 72-120 hpf for 1.0 µM BPA, while 0.32 and 0.24-fold of insulin expression were elicited by 0.3 and 1 µM BPS respectively at 72 hpf. Significant elevation of glucose levels; 16.3% (for 1.0 µM BPA), 7.20% (for 0.3 µM BPS), and 74.09% (for 1.0 µM BPS) higher than the control groups were observed. In addition, pancreatic-related genes pdx-1, foxa2, ptfla, and isl1 were significantly interfered compared with the untreated group. Moreover, the maintenance methylation gene, dnmt1, was monotonically and significantly decreased at early stage of development following BPA exposure but remained constant for BPS treatment relative to the control group. DNMT3a and DNMT3b orthologs were distinctively altered following BPA/BPS embryonic exposure. Our data indicated that embryonic exposure to low concentration of BPA/BPS can impair the normal expressions of pancreatic-associated genes and DNA methylation pattern of selected genes in zebrafish early development.
Collapse
Affiliation(s)
- Eric Gyimah
- Institute of Environmental Health and Ecological Security, School of Environment and Safety Engineering, Jiangsu University, Zhenjiang, 212013, China
| | - Xing Dong
- Institute of Environmental Health and Ecological Security, School of Environment and Safety Engineering, Jiangsu University, Zhenjiang, 212013, China
| | - Hai Xu
- Institute of Environmental Health and Ecological Security, School of Environment and Safety Engineering, Jiangsu University, Zhenjiang, 212013, China.
| | - Zhen Zhang
- Institute of Environmental Health and Ecological Security, School of Environment and Safety Engineering, Jiangsu University, Zhenjiang, 212013, China
| | - John Kenneth Mensah
- Department of Chemistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| |
Collapse
|
29
|
Banerji R, Huynh C, Figueroa F, Dinday MT, Baraban SC, Patel M. Enhancing glucose metabolism via gluconeogenesis is therapeutic in a zebrafish model of Dravet syndrome. Brain Commun 2021; 3:fcab004. [PMID: 33842883 PMCID: PMC8023476 DOI: 10.1093/braincomms/fcab004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 01/18/2023] Open
Abstract
Energy-producing pathways are novel therapeutic targets for the treatment of neurodevelopmental disorders. Here, we focussed on correcting metabolic defects in a catastrophic paediatric epilepsy, Dravet syndrome which is caused by mutations in sodium channel NaV1.1 gene, SCN1A. We utilized a translatable zebrafish model of Dravet syndrome (scn1lab) which exhibits key characteristics of patients with Dravet syndrome and shows metabolic deficits accompanied by down-regulation of gluconeogenesis genes, pck1 and pck2. Using a metabolism-based small library screen, we identified compounds that increased gluconeogenesis via up-regulation of pck1 gene expression in scn1lab larvae. Treatment with PK11195, a pck1 activator and a translocator protein ligand, normalized dys-regulated glucose levels, metabolic deficits, translocator protein expression and significantly decreased electrographic seizures in mutant larvae. Inhibition of pck1 in wild-type larvae mimicked metabolic and behaviour defects observed in scn1lab mutants. Together, this suggests that correcting dys-regulated metabolic pathways can be therapeutic in neurodevelopmental disorders such as Dravet syndrome arising from ion channel dysfunction.
Collapse
Affiliation(s)
- Rajeswari Banerji
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, CA 80045, USA
| | - Christopher Huynh
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, CA 80045, USA
| | - Francisco Figueroa
- Department of Neurological Surgery, Epilepsy Research Laboratory, University of California, San Francisco, CA 94143, USA
| | - Matthew T Dinday
- Department of Neurological Surgery, Epilepsy Research Laboratory, University of California, San Francisco, CA 94143, USA
| | - Scott C Baraban
- Department of Neurological Surgery, Epilepsy Research Laboratory, University of California, San Francisco, CA 94143, USA
| | - Manisha Patel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, CA 80045, USA
| |
Collapse
|
30
|
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: 0.4] [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
|
31
|
Crocins from Crocus sativus L. in the Management of Hyperglycemia. In Vivo Evidence from Zebrafish. Molecules 2020; 25:molecules25225223. [PMID: 33182581 PMCID: PMC7696463 DOI: 10.3390/molecules25225223] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/03/2020] [Accepted: 11/07/2020] [Indexed: 12/11/2022] Open
Abstract
Diabetes mellitus is a disease characterized by persistent high blood glucose levels and accompanied by impaired metabolic pathways. In this study, we used zebrafish to investigate the effect of crocins isolated from Crocus sativus L., on the control of glucose levels and pancreatic β-cells. Embryos were exposed to an aqueous solution of crocins and whole embryo glucose levels were measured at 48 h post-treatment. We showed that the application of crocins reduces zebrafish embryo glucose levels and enhances insulin expression. We also examined whether crocins are implicated in the metabolic pathway of gluconeogenesis. We showed that following a single application of crocins and glucose level reduction, the expression of phosphoenolpyruvate carboxykinase1 (pck1), a key gene involved in glucose metabolism, is increased. We propose a putative role for the crocins in glucose metabolism and insulin management.
Collapse
|
32
|
Lou B, Boger M, Bennewitz K, Sticht C, Kopf S, Morgenstern J, Fleming T, Hell R, Yuan Z, Nawroth PP, Kroll J. Elevated 4-hydroxynonenal induces hyperglycaemia via Aldh3a1 loss in zebrafish and associates with diabetes progression in humans. Redox Biol 2020; 37:101723. [PMID: 32980661 PMCID: PMC7519378 DOI: 10.1016/j.redox.2020.101723] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/31/2020] [Accepted: 09/11/2020] [Indexed: 12/12/2022] Open
Abstract
Increased methylglyoxal (MG) formation is associated with diabetes and its complications. In zebrafish, knockout of the main MG detoxifying system Glyoxalase 1, led to limited MG elevation but significantly elevated aldehyde dehydrogenases (ALDH) activity and aldh3a1 expression, suggesting the compensatory role of Aldh3a1 in diabetes. To evaluate the function of Aldh3a1 in glucose homeostasis and diabetes, aldh3a1−/− zebrafish mutants were generated using CRISPR-Cas9. Vasculature and pancreas morphology were analysed by zebrafish transgenic reporter lines. Corresponding reactive carbonyl species (RCS), glucose, transcriptome and metabolomics screenings were performed and ALDH activity was measured for further verification. Aldh3a1−/− zebrafish larvae displayed retinal vasodilatory alterations, impaired glucose homeostasis, which can be aggravated via pdx1 silencing induced hyperglycaemia. Unexpectedly, MG was not altered, but 4-hydroxynonenal (4-HNE), another prominent lipid peroxidation RCS exhibited high affinity with Aldh3a1, was increased in aldh3a1 mutants. 4-HNE was responsible for the retinal phenotype via pancreas disruption induced hyperglycaemia and can be rescued via l-Carnosine treatment. Furthermore, in type 2 diabetic patients, serum 4-HNE was increased and correlated with disease progression. Thus, our data suggest impaired 4-HNE detoxification and elevated 4-HNE concentration as biomarkers but also the possible inducers for diabetes, from genetic susceptibility to the pathological progression. Aldh3a1 mutant was generated using CRISPR/Cas9 and displayed impaired glucose homeostasis. Elevated 4-Hydroxynonenal (4-HNE) was responsible for hyperglycaemia in aldh3a1 mutants and was rescued by Carnosine. Patient serum 4-HNE level was correlated with HbA1c and fasting glucose. Impaired 4-HNE detoxification acts as possible inducers for diabetes, from genetic susceptibility to pathological progress.
Collapse
Affiliation(s)
- Bowen Lou
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Cardiovascular Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710048, China
| | - Mike Boger
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Katrin Bennewitz
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Carsten Sticht
- Center for Medical Research (ZMF), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefan Kopf
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Jakob Morgenstern
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Thomas Fleming
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Rüdiger Hell
- Metabolomics Core Technology Platform, Centre for Organismal Studies, Heidelberg University, Heidelberg, Germany
| | - Zuyi Yuan
- Cardiovascular Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710048, China
| | - Peter Paul Nawroth
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz-Zentrum, München, Heidelberg, Germany
| | - Jens Kroll
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
33
|
Frame JM, Kubaczka C, Long TL, Esain V, Soto RA, Hachimi M, Jing R, Shwartz A, Goessling W, Daley GQ, North TE. Metabolic Regulation of Inflammasome Activity Controls Embryonic Hematopoietic Stem and Progenitor Cell Production. Dev Cell 2020; 55:133-149.e6. [PMID: 32810442 DOI: 10.1016/j.devcel.2020.07.015] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 05/26/2020] [Accepted: 07/22/2020] [Indexed: 12/21/2022]
Abstract
Embryonic hematopoietic stem and progenitor cells (HSPCs) robustly proliferate while maintaining multilineage potential in vivo; however, an incomplete understanding of spatiotemporal cues governing their generation has impeded robust production from human induced pluripotent stem cells (iPSCs) in vitro. Using the zebrafish model, we demonstrate that NLRP3 inflammasome-mediated interleukin-1-beta (IL1β) signaling drives HSPC production in response to metabolic activity. Genetic induction of active IL1β or pharmacologic inflammasome stimulation increased HSPC number as assessed by in situ hybridization for runx1/cmyb and flow cytometry. Loss of inflammasome components, including il1b, reduced CD41+ HSPCs and prevented their expansion in response to metabolic cues. Cell ablation studies indicated that macrophages were essential for initial inflammasome stimulation of Il1rl1+ HSPCs. Significantly, in human iPSC-derived hemogenic precursors, transient inflammasome stimulation increased multilineage hematopoietic colony-forming units and T cell progenitors. This work establishes the inflammasome as a conserved metabolic sensor that expands HSPC production in vivo and in vitro.
Collapse
Affiliation(s)
- Jenna M Frame
- Stem Cell Program, Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Caroline Kubaczka
- Stem Cell Program, Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Timothy L Long
- Stem Cell Program, Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Virginie Esain
- Stem Cell Program, Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Rebecca A Soto
- Stem Cell Program, Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Developmental and Regenerative Biology Program, Harvard Medical School, Boston, MA 02115, USA
| | - Mariam Hachimi
- Stem Cell Program, Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Ran Jing
- Stem Cell Program, Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Arkadi Shwartz
- Genetics Division, Brigham & Women's Hospital, Boston, MA 02115, USA
| | - Wolfram Goessling
- Developmental and Regenerative Biology Program, Harvard Medical School, Boston, MA 02115, USA; Genetics Division, Brigham & Women's Hospital, Boston, MA 02115, USA; Gastroenterology Division, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - George Q Daley
- Stem Cell Program, Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Developmental and Regenerative Biology Program, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Trista E North
- Stem Cell Program, Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Developmental and Regenerative Biology Program, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
34
|
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: 21] [Impact Index Per Article: 4.2] [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
|
35
|
Wiggenhauser LM, Qi H, Stoll SJ, Metzger L, Bennewitz K, Poschet G, Krenning G, Hillebrands JL, Hammes HP, Kroll J. Activation of Retinal Angiogenesis in Hyperglycemic pdx1 -/- Zebrafish Mutants. Diabetes 2020; 69:1020-1031. [PMID: 32139597 DOI: 10.2337/db19-0873] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 02/26/2020] [Indexed: 11/13/2022]
Abstract
Progression from the initial vascular response upon hyperglycemia to a proliferative stage with neovacularizations is the hallmark of proliferative diabetic retinopathy. Here, we report on the novel diabetic pdx1 -/- zebrafish mutant as a model for diabetic retinopathy that lacks the transcription factor pdx1 through CRISPR-Cas9-mediated gene knockout leading to disturbed pancreatic development and hyperglycemia. Larval pdx1 -/- mutants prominently show vasodilation of blood vessels through increased vascular thickness in the hyaloid network as direct developmental precursor of the adult retinal vasculature in zebrafish. In adult pdx1 -/- mutants, impaired glucose homeostasis induces increased hyperbranching and hypersprouting with new vessel formation in the retina and aggravation of the vascular alterations from the larval to the adult stage. Both vascular aspects respond to antiangiogenic and antihyperglycemic pharmacological interventions in the larval stage and are accompanied by alterations in the nitric oxide metabolism. Thus, the pdx1 -/- mutant represents a novel model to study mechanisms of hyperglycemia-induced retinopathy wherein extensive proangiogenic alterations in blood vessel morphology and metabolic alterations underlie the vascular phenotype.
Collapse
Affiliation(s)
- Lucas M Wiggenhauser
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Haozhe Qi
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sandra J Stoll
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lena Metzger
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Katrin Bennewitz
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gernot Poschet
- Metabolomics Core Technology Platform, Centre for Organismal Studies, Heidelberg University, Heidelberg, Germany
| | - Guido Krenning
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jan-Luuk Hillebrands
- Pathology Section, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Hans-Peter Hammes
- Fifth Medical Department and European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jens Kroll
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
36
|
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: 7.0] [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
|
37
|
Arjmand B, Tayanloo-Beik A, Foroughi Heravani N, Alaei S, Payab M, Alavi-Moghadam S, Goodarzi P, Gholami M, Larijani B. Zebrafish for Personalized Regenerative Medicine; A More Predictive Humanized Model of Endocrine Disease. Front Endocrinol (Lausanne) 2020; 11:396. [PMID: 32765420 PMCID: PMC7379230 DOI: 10.3389/fendo.2020.00396] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 05/18/2020] [Indexed: 12/18/2022] Open
Abstract
Regenerative medicine is a multidisciplinary field that aims to determine different factors and develop various methods to regenerate impaired tissues, organs, and cells in the disease and impairment conditions. When treatment procedures are specified according to the individual's information, the leading role of personalized regenerative medicine will be revealed in developing more effective therapies. In this concept, endocrine disorders can be considered as potential candidates for regenerative medicine application. Diabetes mellitus as a worldwide prevalent endocrine disease causes different damages such as blood vessel damages, pancreatic damages, and impaired wound healing. Therefore, a global effort has been devoted to diabetes mellitus investigations. Hereupon, the preclinical study is a fundamental step. Up to now, several species of animals have been modeled to identify the mechanism of multiple diseases. However, more recent researches have been demonstrated that animal models with the ability of tissue regeneration are more suitable choices for regenerative medicine studies in endocrine disorders, typically diabetes mellitus. Accordingly, zebrafish has been introduced as a model that possesses the capacity to regenerate different organs and tissues. Especially, fine regeneration in zebrafish has been broadly investigated in the regenerative medicine field. In addition, zebrafish is a suitable model for studying a variety of different situations. For instance, it has been used for developmental studies because of the special characteristics of its larva. In this review, we discuss the features of zebrafish that make it a desirable animal model, the advantages of zebrafish and recent research that shows zebrafish is a promising animal model for personalized regenerative diseases. Ultimately, we conclude that as a newly introduced model, zebrafish can have a leading role in regeneration studies of endocrine diseases and provide a good perception of underlying mechanisms.
Collapse
Affiliation(s)
- Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Najmeh Foroughi Heravani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Setareh Alaei
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Moloud Payab
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Goodarzi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Gholami
- Department of Toxicology and Pharmacology, Toxicology and Poisoning Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- *Correspondence: Bagher Larijani
| |
Collapse
|
38
|
Tu W, Martínez R, Navarro-Martin L, Kostyniuk DJ, Hum C, Huang J, Deng M, Jin Y, Chan HM, Mennigen JA. Bioconcentration and Metabolic Effects of Emerging PFOS Alternatives in Developing Zebrafish. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2019; 53:13427-13439. [PMID: 31609598 DOI: 10.1021/acs.est.9b03820] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The novel PFOS alternatives, 6:2 chlorinated polyfluorinated ether sulfonate (F-53B) and sodium p-perfluorous nonenoxybenzenesulfonate (OBS), are emerging in the Chinese market, but little is known about their ecological risks. In this study, zebrafish embryos were exposed to PFOS, F-53B, and OBS to evaluate their bioconcentration and acute metabolic consequences. Per- and polyfluoroalkyl substances (PFASs) accumulated in larvae in the order of F-53B > PFOS > OBS, with the bioconcentration factors ranging from 20 to 357. Exposure to F-53B and PFOS, but not OBS, increased energy expenditure, and reduced feed intake in a concentration-dependent manner and the expression of genes involved in metabolic pathways at the transcriptional and translational levels. Molecular docking revealed that the binding affinities of PFASs to glucokinase were decreased in the following order: F-53B > PFOS > OBS. Finally, the results of Point of Departure (PoD) indicate that metabolic end points at the molecular and organismal level are most sensitive to F-53B followed by PFOS and OBS. Collectively, F-53B has the highest bioconcentration potential and the strongest metabolism-disrupting effects, followed by PFOS and OBS. Our findings have important implications for the assessment of early developmental metabolic effects of PFOS alternatives F-53B and OBS in wildlife and humans.
Collapse
Affiliation(s)
- Wenqing Tu
- Research Institute of Poyang Lake , Jiangxi Academy of Sciences , Nanchang 330012 , China
| | - Rubén Martínez
- Department of Environmental Chemistry , Institute of Environmental Assessment and Water Research, IDAEA-CSIC , Jordi Girona, Barcelona 18-26 08034 , Spain
- Department of Cellular Biology, Physiology and Immunology , Universitat de Barcelona (UB) , Barcelona 585 08007 , Spain
| | - Laia Navarro-Martin
- Department of Environmental Chemistry , Institute of Environmental Assessment and Water Research, IDAEA-CSIC , Jordi Girona, Barcelona 18-26 08034 , Spain
| | - Daniel J Kostyniuk
- Department of Biology , University of Ottawa , Ottawa , Ontario K1N 6N5 , Canada
| | - Christine Hum
- Department of Biology , University of Ottawa , Ottawa , Ontario K1N 6N5 , Canada
| | - Jing Huang
- Research Institute of Poyang Lake , Jiangxi Academy of Sciences , Nanchang 330012 , China
| | - Mi Deng
- Research Institute of Poyang Lake , Jiangxi Academy of Sciences , Nanchang 330012 , China
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering , Zhejiang University of Technology , Hangzhou , 310032 , China
| | - Hing Man Chan
- Department of Biology , University of Ottawa , Ottawa , Ontario K1N 6N5 , Canada
| | | |
Collapse
|
39
|
Mullapudi ST, Boezio GLM, Rossi A, Marass M, Matsuoka RL, Matsuda H, Helker CSM, Yang YHC, Stainier DYR. Disruption of the pancreatic vasculature in zebrafish affects islet architecture and function. Development 2019; 146:dev.173674. [PMID: 31597659 DOI: 10.1242/dev.173674] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 10/03/2019] [Indexed: 12/14/2022]
Abstract
A dense local vascular network is crucial for pancreatic endocrine cells to sense metabolites and secrete hormones, and understanding the interactions between the vasculature and the islets may allow for therapeutic modulation in disease conditions. Using live imaging in two models of vascular disruption in zebrafish, we identified two distinct roles for the pancreatic vasculature. At larval stages, expression of a dominant negative version of Vegfaa (dnVegfaa) in β-cells led to vascular and endocrine cell disruption with a minor impairment in β-cell function. In contrast, expression of a soluble isoform of Vegf receptor 1 (sFlt1) in β-cells blocked the formation of the pancreatic vasculature and drastically stunted glucose response, although islet architecture was not affected. Notably, these effects of dnVegfaa or sFlt1 were not observed in animals lacking vegfaa, vegfab, kdrl, kdr or flt1 function, indicating that they interfere with multiple ligands and/or receptors. In adults, disrupted islet architecture persisted in dnVegfaa-expressing animals, whereas sFlt1-expressing animals displayed large sheets of β-cells along their pancreatic ducts, accompanied by impaired glucose tolerance in both models. Thus, our study reveals novel roles for the vasculature in patterning and function of the islet.
Collapse
Affiliation(s)
- Sri Teja Mullapudi
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Giulia L M Boezio
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Andrea Rossi
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Michele Marass
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Ryota L Matsuoka
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Hiroki Matsuda
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Christian S M Helker
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Yu Hsuan Carol Yang
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| |
Collapse
|
40
|
Schmöhl F, Peters V, Schmitt CP, Poschet G, Büttner M, Li X, Weigand T, Poth T, Volk N, Morgenstern J, Fleming T, Nawroth PP, Kroll J. CNDP1 knockout in zebrafish alters the amino acid metabolism, restrains weight gain, but does not protect from diabetic complications. Cell Mol Life Sci 2019; 76:4551-4568. [PMID: 31073745 PMCID: PMC11105213 DOI: 10.1007/s00018-019-03127-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/22/2019] [Accepted: 04/30/2019] [Indexed: 12/12/2022]
Abstract
The gene CNDP1 was associated with the development of diabetic nephropathy. Its enzyme carnosinase 1 (CN1) primarily hydrolyzes the histidine-containing dipeptide carnosine but other organ and metabolic functions are mainly unknown. In our study we generated CNDP1 knockout zebrafish, which showed strongly decreased CN1 activity and increased intracellular carnosine levels. Vasculature and kidneys of CNDP1-/- zebrafish were not affected, except for a transient glomerular alteration. Amino acid profiling showed a decrease of certain amino acids in CNDP1-/- zebrafish, suggesting a specific function for CN1 in the amino acid metabolisms. Indeed, we identified a CN1 activity for Ala-His and Ser-His. Under diabetic conditions increased carnosine levels in CNDP1-/- embryos could not protect from respective organ alterations. Although, weight gain through overfeeding was restrained by CNDP1 loss. Together, zebrafish exhibits CN1 functions, while CNDP1 knockout alters the amino acid metabolism, attenuates weight gain but cannot protect organs from diabetic complications.
Collapse
Affiliation(s)
- Felix Schmöhl
- European Center for Angioscience (ECAS), Department of Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Verena Peters
- Center for Paediatric and Adolescent Medicine, University of Heidelberg, Im Neuenheimer Feld 669, 69120, Heidelberg, Germany
| | - Claus Peter Schmitt
- Center for Paediatric and Adolescent Medicine, University of Heidelberg, Im Neuenheimer Feld 669, 69120, Heidelberg, Germany
| | - Gernot Poschet
- Center for Organismal Studies (COS), University of Heidelberg, Im Neuenheimer Feld 360, 69120, Heidelberg, Germany
| | - Michael Büttner
- Center for Organismal Studies (COS), University of Heidelberg, Im Neuenheimer Feld 360, 69120, Heidelberg, Germany
| | - Xiaogang Li
- European Center for Angioscience (ECAS), Department of Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Tim Weigand
- Center for Paediatric and Adolescent Medicine, University of Heidelberg, Im Neuenheimer Feld 669, 69120, Heidelberg, Germany
| | - Tanja Poth
- CMCP-Center for Model System and Comparative Pathology, Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Nadine Volk
- Tissue Bank of the National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Jakob Morgenstern
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Thomas Fleming
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Peter P Nawroth
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Center for Diabetes Research (DZD), 85764, München-Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz-Zentrum, München, Im Neuenheimer Feld 410, F02 Room 02.414-02.434, 69120, Heidelberg, Germany
| | - Jens Kroll
- European Center for Angioscience (ECAS), Department of Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany.
| |
Collapse
|
41
|
Ennerfelt H, Voithofer G, Tibbo M, Miller D, Warfield R, Allen S, Kennett Clark J. Disruption of peripheral nerve development in a zebrafish model of hyperglycemia. J Neurophysiol 2019; 122:862-871. [DOI: 10.1152/jn.00318.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Diabetes mellitus-induced hyperglycemia is associated with a number of pathologies such as retinopathy, nephropathy, delayed wound healing, and diabetic peripheral neuropathy (DPN). Approximately 50% of patients with diabetes mellitus will develop DPN, which is characterized by disrupted sensory and/or motor functioning, with treatment limited to pain management. Zebrafish ( Danio rerio) are an emerging animal model used to study a number of metabolic disorders, including diabetes. Diabetic retinopathy, nephropathy, and delayed wound healing have all been demonstrated in zebrafish. Recently, our laboratory has demonstrated that following the ablation of the insulin-producing β-cells of the pancreas (and subsequent hyperglycemia), the peripheral nerves begin to show signs of dysregulation. In this study, we take a different approach, taking advantage of the transdermal absorption abilities of zebrafish larvae to extend the period of hyperglycemia. Following 5 days of 60 mM d-glucose treatment, we observed motor axon defasciculation, disturbances in perineurial glia sheath formation, decreased myelination of motor axons, and sensory neuron mislocalization. This study extends our understanding of the structural changes of the peripheral nerve following induction of hyperglycemia and does so in an animal model capable of potential DPN drug discovery in the future. NEW & NOTEWORTHY Zebrafish are emerging as a robust model system for the study of diabetic complications such as retinopathy, nephropathy, and impaired wound healing. We present a novel model of diabetic peripheral neuropathy in zebrafish in which the integrity of the peripheral nerve is dysregulated following the induction of hyperglycemia. By using this model, future studies can focus on elucidating the underlying molecular mechanisms currently unknown.
Collapse
Affiliation(s)
- Hannah Ennerfelt
- Department of Biological Sciences, Salisbury University, Salisbury, Maryland
- Department of Psychology, Salisbury University, Salisbury, Maryland
| | - Gabrielle Voithofer
- Department of Biological Sciences, Salisbury University, Salisbury, Maryland
- Department of Psychology, Salisbury University, Salisbury, Maryland
| | - Morgan Tibbo
- Department of Biological Sciences, Salisbury University, Salisbury, Maryland
- Department of Psychology, Salisbury University, Salisbury, Maryland
| | - Derrick Miller
- Department of Chemistry, Salisbury University, Salisbury, Maryland
| | - Rebecca Warfield
- Department of Biological Sciences, Salisbury University, Salisbury, Maryland
- Department of Psychology, Salisbury University, Salisbury, Maryland
| | - Samantha Allen
- Department of Biological Sciences, Salisbury University, Salisbury, Maryland
| | | |
Collapse
|
42
|
Yun Y, Zhang Y, Li G, Chen S, Sang N. Embryonic exposure to oxy-polycyclic aromatic hydrocarbon interfere with pancreatic β-cell development in zebrafish via altering DNA methylation and gene expression. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 660:1602-1609. [PMID: 30743951 DOI: 10.1016/j.scitotenv.2018.12.476] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 12/26/2018] [Accepted: 12/31/2018] [Indexed: 06/09/2023]
Abstract
Oxygenated polycyclic aromatic hydrocarbons (OPAHs) are a class of anthropogenic, persistent and very toxic PAH contaminant associated with developmental toxicity. Abnormal glucose metabolism disturbs energy balances that impair the early development of vertebrates, but the mechanisms by which maternal OPAH exposure alters glucose homeostasis in offspring are not well understood. Studies have suggested that epigenetic changes, particularly in DNA methylation, provide a memory of plastic developmental responses to the environment, leading to the generation of novel offspring phenotypes. The objective of this study is to test the hypothesis that embryonic exposure to low-dose OPAH can impair early β-cell differentiation in zebrafish (Danio rerio) by altering DNA methylation and gene expression. The zebrafish embryos were exposed to 0, 0.03, 0.1, 0.3, 1 and 3 μM 9‑fluorenone (9-FLO) at 3 h postfertilization (hpf) until 120 hpf to assess pancreatic organogenesis. 9-FLO exposure reduced total body length, eye length and heart rate, decreased insulin generation, interfered with glucose metabolism, and altered the expression of pancreatic organogenesis-related genes pdx-1, foxa2, isl1 and ptf1a. In particular, low-dose embryonic 9-FLO exposure significantly decreased β-cell differentiation marker gene pdx-1 mRNA levels, indicating that pancreatic endocrine is a more sensitive target response to embryonic low-dose OPAH exposure. Additionally, we found that DNA methyltransferases dnmt1 and dnmt3 were elevated and the DNA methylation at promoter regions of pdx-1 was increased at an early stage of development. These data demonstrated that the low-dose OPAH embryonic exposure can impair pancreatic endocrine development by increasing DNA methylation at the promoter regions of pdx-1 that are essential for β-cell differentiation.
Collapse
Affiliation(s)
- Yang Yun
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Yujie Zhang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Guangke Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Shaoyu Chen
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, USA; University of Louisville Alcohol Research Center, Louisville, KY, USA
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| |
Collapse
|
43
|
Rocker A, Howell J, Voithofer G, Clark JK. Acute effects of hyperglycemia on the peripheral nervous system in zebrafish (Danio rerio) following nitroreductase-mediated β-cell ablation. Am J Physiol Regul Integr Comp Physiol 2019; 316:R395-R405. [DOI: 10.1152/ajpregu.00258.2018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Diabetic peripheral neuropathy (DPN) is estimated to affect 50% of diabetic patients. Although DPN is highly prevalent, molecular mechanisms remain unknown and treatment is limited to pain relief and glycemic control. We provide a novel model of acute DPN in zebrafish ( Danio rerio) larvae. Beginning 5 days postfertilization (dpf), zebrafish expressing nitroreductase in their pancreatic β-cells were treated with metronidazole (MTZ) for 48 h and checked for β-cell ablation 7 dpf. In experimental design, this was meant to serve as proof of concept that β-cell ablation and hyperglycemia are possible at this time point, but we were surprised to find changes in both sensory and motor nerve components. Compared with controls, neurod+ sensory neurons were often observed outside the dorsal root ganglia in MTZ-treated fish. Fewer motor nerves were properly ensheathed by nkx2.2a+ perineurial cells, and tight junctions were disrupted along the motor nerve in MTZ-treated fish compared with controls. Not surprisingly, the motor axons of the MTZ-treated group were defasciculated compared with the control group, myelination was attenuated, and there was a subtle difference in Schwann cell number between the MTZ-treated and control group. All structural changes occurred in the absence of behavioral changes in the larvae at this time point, suggesting that peripheral nerves are influenced by acute hyperglycemia before becoming symptomatic. Moving forward, this novel animal model of DPN will allow us to access the molecular mechanisms associated with the acute changes in the hyperglycemic peripheral nervous system, which may help direct therapeutic approaches.
Collapse
Affiliation(s)
- Amanda Rocker
- Department of Biological Sciences, Salisbury University, Salisbury, Maryland
| | - Julia Howell
- Department of Biological Sciences, Salisbury University, Salisbury, Maryland
| | - Gabrielle Voithofer
- Department of Biological Sciences, Salisbury University, Salisbury, Maryland
| | | |
Collapse
|
44
|
Krishnan J, Rohner N. Sweet fish: Fish models for the study of hyperglycemia and diabetes. J Diabetes 2019; 11:193-203. [PMID: 30264455 DOI: 10.1111/1753-0407.12860] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 08/01/2018] [Accepted: 09/09/2018] [Indexed: 01/15/2023] Open
Abstract
Fish are good for your health in more ways than you may expect. For one, eating fish is a common dietary recommendation for a healthy diet. However, fish have much more to provide than omega-3 fatty acids to your circulatory system. Some fish species now serve as important and innovative model systems for diabetes research, providing novel and unique advantages compared with classical research models. Not surprisingly, the largest share of diabetes research in fish occurs in the laboratory workhorse among fish, the zebrafish (Danio rerio). Established as a genetic model system to study development, these small cyprinid fish have eventually conquered almost every scientific discipline and, over the past decade, have emerged as an important model system for metabolic diseases, including diabetes mellitus. In this review we highlight the practicability of using zebrafish to study diabetes and hyperglycemia, and summarize some of the recent research and breakthroughs made using this model. Equally exciting is the appearance of another emerging discipline, one that is taking advantage of evolution by studying cases of naturally occurring insulin resistance in fish species. We briefly discuss two such models in this review, namely the rainbow trout (Oncorhynchus mykiss) and the cavefish (Astyanax mexicanus).
Collapse
Affiliation(s)
- Jaya Krishnan
- Stowers Institute for Medical Research, Kansas City, Missouri, USA
| | - Nicolas Rohner
- Stowers Institute for Medical Research, Kansas City, Missouri, USA
- Department of Molecular and Integrative Physiology, KU Medical Center, Kansas City, Missouri, USA
| |
Collapse
|
45
|
Zhao J, Zong W, Zhao Y, Gou D, Liang S, Shen J, Wu Y, Zheng X, Wu R, Wang X, Niu F, Wang A, Zhang Y, Xiong JW, Chen L, Liu Y. In vivo imaging of β-cell function reveals glucose-mediated heterogeneity of β-cell functional development. eLife 2019; 8:41540. [PMID: 30694176 PMCID: PMC6395064 DOI: 10.7554/elife.41540] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 01/29/2019] [Indexed: 12/22/2022] Open
Abstract
How pancreatic β-cells acquire function in vivo is a long-standing mystery due to the lack of technology to visualize β-cell function in living animals. Here, we applied a high-resolution two-photon light-sheet microscope for the first in vivo imaging of Ca2+activity of every β-cell in Tg (ins:Rcamp1.07) zebrafish. We reveal that the heterogeneity of β-cell functional development in vivo occurred as two waves propagating from the islet mantle to the core, coordinated by islet vascularization. Increasing amounts of glucose induced functional acquisition and enhancement of β-cells via activating calcineurin/nuclear factor of activated T-cells (NFAT) signaling. Conserved in mammalians, calcineurin/NFAT prompted high-glucose-stimulated insulin secretion of neonatal mouse islets cultured in vitro. However, the reduction in low-glucose-stimulated insulin secretion was dependent on optimal glucose but independent of calcineurin/NFAT. Thus, combination of optimal glucose and calcineurin activation represents a previously unexplored strategy for promoting functional maturation of stem cell-derived β-like cells in vitro. When the amount of sugar in our body rises, specialised cells known as β-cells respond by releasing insulin, a hormone that acts on various organs to keep blood sugar levels within a healthy range. These cells cluster in small ‘islets’ inside our pancreas. If the number of working β-cells declines, diseases such as diabetes may appear and it becomes difficult to regulate the amount of sugar in our bodies. Understanding how β-cells normally develop and mature in the embryo could help us learn how to make new ones in the laboratory. In particular, researchers are interested in studying how different body signals, such as blood sugar levels, turn immature β-cells into fully productive cells. However, in mammals, the pancreas and its islets are buried deep inside the embryo and they cannot be observed easily. Here, Zhao et al. circumvented this problem by doing experiments on zebrafish embryos, which are transparent, grow outside their mother’s body, and have pancreatic islets that are similar to the ones found in mammals. A three-dimensional microscopy technique was used to watch individual β-cells activity over long periods, which revealed that the cells start being able to produce insulin at different times. The β-cells around the edge of each islet were the first to have access to blood sugar signals: they gained their hormone-producing role earlier than the cells in the core of an islet, which only sensed the information later on. Zhao et al. then exposed the zebrafish embryos to different amounts of sugar. This showed that there is an optimal concentration of sugar which helps β-cells develop by kick-starting a cascade of events inside the cell. Further experiments confirmed that the same pathway and optimal sugar concentration exist for mammalian islets grown in the laboratory. These findings may help researchers find better ways of making new β-cells to treat diabetic patients. In the future, using the three-dimensional imaging technique in zebrafish embryos may lead to more discoveries on how the pancreas matures.
Collapse
Affiliation(s)
- Jia Zhao
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Weijian Zong
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China.,China Department of Cognitive Sciences, Institute of Basic Medical Sciences, Beijing, China
| | - Yiwen Zhao
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Dongzhou Gou
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Shenghui Liang
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Jiayu Shen
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Yi Wu
- School of Software and Microelectronics, Peking University, Beijing, China
| | - Xuan Zheng
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Runlong Wu
- School of Electronics Engineering and Computer Science, Peking University, Beijing, China
| | - Xu Wang
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Fuzeng Niu
- State Key Laboratory of Advanced Optical Communication System and Networks, School of Electronics Engineering and Computer Science, Peking University, Beijing, China
| | - Aimin Wang
- State Key Laboratory of Advanced Optical Communication System and Networks, School of Electronics Engineering and Computer Science, Peking University, Beijing, China
| | - Yunfeng Zhang
- School of Electronics Engineering and Computer Science, Peking University, Beijing, China
| | - Jing-Wei Xiong
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Liangyi Chen
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Yanmei Liu
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China.,Institute for Brain Research and Rehabilitation (IBRR), Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, China
| |
Collapse
|
46
|
Zhao F, Wang H, Wei P, Jiang G, Wang W, Zhang X, Ru S. Impairment of bisphenol F on the glucose metabolism of zebrafish larvae. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 165:386-392. [PMID: 30218961 DOI: 10.1016/j.ecoenv.2018.09.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/25/2018] [Accepted: 09/02/2018] [Indexed: 06/08/2023]
Abstract
Bisphenol F (BPF) is a substitute of bisphenol A in the production of epoxy resin and polycarbonate. Its extensive use in consumer products leads to a wide human exposure at high levels. Although the adverse effects of BPF on animal health are of increasing public concern, its risks on systematic glucose metabolism and blood glucose concentrations still remain largely unknown. Using zebrafish larvae as the model animal, we investigated the disturbance of BPF exposure on glucose metabolism and the underlying mechanisms. Zebrafish larvae at 96 h post fertilization were exposed to 0.1, 1, 10, and 100 μg/L of BPF for 48 h. Compared with the control group, glucose levels of larvae increased significantly in the 10 and 100 μg/L exposure groups, which are associated with enhancement of gluconeogenesis and suppression of glycolysis induced by high doses of BPF. Additionally, both mRNA expressions and protein levels of insulin increased significantly in the 10 and 100 μg/L exposure groups, while transcription levels of genes encoding insulin receptor substrates decreased significantly in these groups, indicating a possibly decreased insulin sensitivity due to impairment of insulin signaling transduction downstream of insulin receptor. Further, compared with BPF alone, co-exposure of larvae to BPF and rosiglitazone, an insulin sensitizer, significantly attenuates increases in both glucose levels and mRNA expressions of a key gluconeogenesis enzyme. Our data therefore indicate impairing insulin signaling transduction may be the main mechanism through which BPF disrupts glucose metabolism and induces hyperglycemia. Results of the present study inform the health risk assessment of BPF and also suggest the use of zebrafish larvae in large-scale screening of chemicals with possible glucose metabolism disturbing effect.
Collapse
Affiliation(s)
- Fei Zhao
- Marine Life Science College, Ocean University of China, 5 Yushan Road, Qingdao 266003, Shandong Province, PR China
| | - Hongfang Wang
- Marine Life Science College, Ocean University of China, 5 Yushan Road, Qingdao 266003, Shandong Province, PR China
| | - Penghao Wei
- Marine Life Science College, Ocean University of China, 5 Yushan Road, Qingdao 266003, Shandong Province, PR China
| | - Guobin Jiang
- Marine Life Science College, Ocean University of China, 5 Yushan Road, Qingdao 266003, Shandong Province, PR China
| | - Wei Wang
- Marine Life Science College, Ocean University of China, 5 Yushan Road, Qingdao 266003, Shandong Province, PR China
| | - Xiaona Zhang
- Marine Life Science College, Ocean University of China, 5 Yushan Road, Qingdao 266003, Shandong Province, PR China
| | - Shaoguo Ru
- Marine Life Science College, Ocean University of China, 5 Yushan Road, Qingdao 266003, Shandong Province, PR China.
| |
Collapse
|
47
|
Mullapudi ST, Helker CS, Boezio GL, Maischein HM, Sokol AM, Guenther S, Matsuda H, Kubicek S, Graumann J, Yang YHC, Stainier DY. Screening for insulin-independent pathways that modulate glucose homeostasis identifies androgen receptor antagonists. eLife 2018; 7:42209. [PMID: 30520733 PMCID: PMC6300353 DOI: 10.7554/elife.42209] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 12/03/2018] [Indexed: 12/13/2022] Open
Abstract
Pathways modulating glucose homeostasis independently of insulin would open new avenues to combat insulin resistance and diabetes. Here, we report the establishment, characterization, and use of a vertebrate ‘insulin-free’ model to identify insulin-independent modulators of glucose metabolism. insulin knockout zebrafish recapitulate core characteristics of diabetes and survive only up to larval stages. Utilizing a highly efficient endoderm transplant technique, we generated viable chimeric adults that provide the large numbers of insulin mutant larvae required for our screening platform. Using glucose as a disease-relevant readout, we screened 2233 molecules and identified three that consistently reduced glucose levels in insulin mutants. Most significantly, we uncovered an insulin-independent beneficial role for androgen receptor antagonism in hyperglycemia, mostly by reducing fasting glucose levels. Our study proposes therapeutic roles for androgen signaling in diabetes and, more broadly, offers a novel in vivo model for rapid screening and decoupling of insulin-dependent and -independent mechanisms. Diabetes is a disease that affects the ability of the body to control the level of sugar in the blood. Individuals with diabetes are unable to make a hormone called insulin – which normally stimulates certain cells to absorb sugar from the blood – or their cells are less able to respond to this hormone. Most treatments for diabetes involve replacing the lost insulin or boosting the hormone’s activity in the body. However, these treatments can also cause individuals to gain weight or become more resistant to insulin, making it harder to control blood sugar levels. In addition to insulin, several other factors regulate the levels of sugar in the blood and some of them may operate independently of insulin. However, little is known about such factors because it is impractical to carry out large-scale screens to identify drugs that target them in humans or mice, which are often used as experimental models for human biology. To overcome this challenge, Mullapudi et al. turned to another animal known as the zebrafish and generated mutant fish that lack insulin. The mutant zebrafish had similar problems with regulating sugar levels as those observed in humans and mice with diabetes. This observation suggests that insulin is just as important in zebrafish as it is in humans and other mammals. The mutant zebrafish did not survive into adulthood, and so Mullapudi et al. transplanted healthy tissue into the zebrafish to allow them to produce enough insulin to survive. These adult zebrafish produced many offspring that still carried the insulin mutation. Mullapudi et al. used these mutant offspring to screen over 2,000 drugs for their ability to decrease blood sugar levels in the absence of insulin. The screen identified three promising candidate drugs, including a molecule that interferes with a receptor for a signal known as androgen. These findings will help researchers investigate new ways to treat diabetes. In the future, the screening approach developed by Mullapudi et al. could be adapted to search for new drugs to treat other human metabolic conditions.
Collapse
Affiliation(s)
- Sri Teja Mullapudi
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Christian Sm Helker
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Giulia Lm Boezio
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Hans-Martin Maischein
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Anna M Sokol
- Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Guenther
- ECCPS Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Hiroki Matsuda
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Kubicek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Johannes Graumann
- Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research, Berlin, Germany
| | - Yu Hsuan Carol Yang
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Didier Yr Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
48
|
Sokol AM, Uszczynska-Ratajczak B, Collins MM, Bazala M, Topf U, Lundegaard PR, Sugunan S, Guenther S, Kuenne C, Graumann J, Chan SSL, Stainier DYR, Chacinska A. Loss of the Mia40a oxidoreductase leads to hepato-pancreatic insufficiency in zebrafish. PLoS Genet 2018; 14:e1007743. [PMID: 30457989 PMCID: PMC6245507 DOI: 10.1371/journal.pgen.1007743] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 10/05/2018] [Indexed: 02/07/2023] Open
Abstract
Development and function of tissues and organs are powered by the activity of mitochondria. In humans, inherited genetic mutations that lead to progressive mitochondrial pathology often manifest during infancy and can lead to death, reflecting the indispensable nature of mitochondrial biogenesis and function. Here, we describe a zebrafish mutant for the gene mia40a (chchd4a), the life-essential homologue of the evolutionarily conserved Mia40 oxidoreductase which drives the biogenesis of cysteine-rich mitochondrial proteins. We report that mia40a mutant animals undergo progressive cellular respiration defects and develop enlarged mitochondria in skeletal muscles before their ultimate death at the larval stage. We generated a deep transcriptomic and proteomic resource that allowed us to identify abnormalities in the development and physiology of endodermal organs, in particular the liver and pancreas. We identify the acinar cells of the exocrine pancreas to be severely affected by mutations in the MIA pathway. Our data contribute to a better understanding of the molecular, cellular and organismal effects of mitochondrial deficiency, important for the accurate diagnosis and future treatment strategies of mitochondrial diseases. Mitochondrial pathologies which result from mutations in the nuclear DNA remain incurable and often lead to death. As mitochondria play various roles in cellular and tissue-specific contexts, the symptoms of mitochondrial pathologies can differ between patients. Thus, diagnosis and treatment of mitochondrial disorders remain challenging. To enhance this, the generation of new models that explore and define the consequences of mitochondria insufficiencies is of central importance. Here, we present a mia40a zebrafish mutant as a model for mitochondrial dysfunction, caused by an imbalance in mitochondrial protein biogenesis. This mutant shares characteristics with existing reports on mitochondria dysfunction, and has led us to identify novel phenotypes such as enlarged mitochondrial clusters in skeletal muscles. In addition, our transcriptomics and proteomics data contribute important findings to the existing knowledge on how faulty mitochondria impinge on vertebrate development in molecular, tissue and organ specific contexts.
Collapse
Affiliation(s)
- Anna M. Sokol
- International Institute of Molecular and Cell Biology, Warsaw, Poland
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- * E-mail: (AMS); (AC)
| | | | - Michelle M. Collins
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Michal Bazala
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Ulrike Topf
- International Institute of Molecular and Cell Biology, Warsaw, Poland
- Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Pia R. Lundegaard
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sreedevi Sugunan
- International Institute of Molecular and Cell Biology, Warsaw, Poland
- Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Stefan Guenther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Carsten Kuenne
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Johannes Graumann
- Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Sherine S. L. Chan
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Didier Y. R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Agnieszka Chacinska
- International Institute of Molecular and Cell Biology, Warsaw, Poland
- Centre of New Technologies, University of Warsaw, Warsaw, Poland
- * E-mail: (AMS); (AC)
| |
Collapse
|
49
|
Zang L, Maddison LA, Chen W. Zebrafish as a Model for Obesity and Diabetes. Front Cell Dev Biol 2018; 6:91. [PMID: 30177968 PMCID: PMC6110173 DOI: 10.3389/fcell.2018.00091] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 07/25/2018] [Indexed: 12/13/2022] Open
Abstract
Obesity and diabetes now considered global epidemics. The prevalence rates of diabetes are increasing in parallel with the rates of obesity and the strong connection between these two diseases has been coined as “diabesity.” The health risks of overweight or obesity include Type 2 diabetes mellitus (T2DM), coronary heart disease and cancer of numerous organs. Both obesity and diabetes are complex diseases that involve the interaction of genetics and environmental factors. The underlying pathogenesis of obesity and diabetes are not well understood and further research is needed for pharmacological and surgical management. Consequently, the use of animal models of obesity and/or diabetes is important for both improving the understanding of these diseases and to identify and develop effective treatments. Zebrafish is an attractive model system for studying metabolic diseases because of the functional conservation in lipid metabolism, adipose biology, pancreas structure, and glucose homeostasis. It is also suited for identification of novel targets associated with the risk and treatment of obesity and diabetes in humans. In this review, we highlight studies using zebrafish to model metabolic diseases, and discuss the advantages and disadvantages of studying pathologies associated with obesity and diabetes in zebrafish.
Collapse
Affiliation(s)
- Liqing Zang
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, United States.,Graduate School of Regional Innovation Studies, Mie University, Tsu, Japan
| | - Lisette A Maddison
- Center for Reproductive Biology, Washington State University, Pullman, WA, United States
| | - Wenbiao Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, United States
| |
Collapse
|
50
|
Lorincz R, Emfinger CH, Walcher A, Giolai M, Krautgasser C, Remedi MS, Nichols CG, Meyer D. In vivo monitoring of intracellular Ca 2+ dynamics in the pancreatic β-cells of zebrafish embryos. Islets 2018; 10:221-238. [PMID: 30521410 PMCID: PMC6300091 DOI: 10.1080/19382014.2018.1540234] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Assessing the response of pancreatic islet cells to glucose stimulation is important for understanding β-cell function. Zebrafish are a promising model for studies of metabolism in general, including stimulus-secretion coupling in the pancreas. We used transgenic zebrafish embryos expressing a genetically-encoded Ca2+ sensor in pancreatic β-cells to monitor a key step in glucose induced insulin secretion; the elevations of intracellular [Ca2+]i. In vivo and ex vivo analyses of [Ca2+]i demonstrate that β-cell responsiveness to glucose is well established in late embryogenesis and that embryonic β-cells also respond to free fatty acid and amino acid challenges. In vivo imaging of whole embryos further shows that indirect glucose administration, for example by yolk injection, results in a slow and asynchronous induction of β-cell [Ca2+]i responses, while intravenous glucose injections cause immediate and islet-wide synchronized [Ca2+]i fluctuations. Finally, we demonstrate that embryos with disrupted mutation of the CaV1.2 channel gene cacna1c are hyperglycemic and that this phenotype is associated with glucose-independent [Ca2+]i fluctuation in β-cells. The data reveal a novel central role of cacna1c in β-cell specific stimulus-secretion coupling in zebrafish and demonstrate that the novel approach we propose - to monitor the [Ca2+]i dynamics in embryonic β-cells in vivo - will help to expand the understanding of β-cell physiological functions in healthy and diseased states.
Collapse
Affiliation(s)
- Reka Lorincz
- Institute of Molecular Biology/CMBI, University of Innsbruck, Innsbruck, Austria
| | - Christopher H. Emfinger
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrea Walcher
- Institute of Molecular Biology/CMBI, University of Innsbruck, Innsbruck, Austria
| | - Michael Giolai
- Institute of Molecular Biology/CMBI, University of Innsbruck, Innsbruck, Austria
| | - Claudia Krautgasser
- Institute of Molecular Biology/CMBI, University of Innsbruck, Innsbruck, Austria
| | - Maria S. Remedi
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Colin G. Nichols
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University School of Medicine, St. Louis, MO, USA
| | - Dirk Meyer
- Institute of Molecular Biology/CMBI, University of Innsbruck, Innsbruck, Austria
- CONTACT Dirk Meyer Institute of Molecular Biology/CMBI, University of Innsbruck, Technikerstrasse 25, Innsbruck 6020, Austria
| |
Collapse
|