1
|
Guan X, Fan Y, Six R, Van Soom A, Pavani KC, Peelman L. MicroRNAs bta-novel-miR-117, bta-novel-miR-234 and bta-novel-miR-417 have adverse effects on blastocyst formation. Theriogenology 2025; 233:88-99. [PMID: 39613498 DOI: 10.1016/j.theriogenology.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024]
Abstract
In a previous study we found that the levels of the novel microRNAs (miRNAs) bta-novel-miR-117 bta-novel-miR-234 and bta-novel-miR-417 (P < 0.001) are significantly up-regulated in extracellular vesicles (EVs) in the culture medium of degenerating embryos compared to blastocysts. Because the functions of these novel miRNAs are still unknown, we investigated their regulatory roles during bovine blastocyst development by adding their mimics and inhibitors to the culture medium. The addition of mimics for bta-novel-miR-117, bta-novel-miR-234 and bta-novel-miR-417 resulted in a decreased blastocyst rate, and supplementation of bta-novel-miR-234 inhibitors increased the cleavage rate significantly (P < 0.001). Low-input transcriptome analysis and RT-qPCR results revealed that bta-novel-miR-117, bta-novel-miR-234 and bta-novel-miR-417 co-target genes such as ANKEF1, HAND2 and SLC2A2, downregulated their expression significantly (P < 0.001). These genes associated with glucose transmembrane transport and plasma membrane raft metabolism play crucial roles in embryonic development. The results suggest that overexpressing of these three novel miRNAs impairs embryonic development, and they might serve as biomarkers to detect failing bovine embryos.
Collapse
Affiliation(s)
- Xuefeng Guan
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820, Merelbeke, Belgium
| | - Yuan Fan
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820, Merelbeke, Belgium
| | - Rani Six
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820, Merelbeke, Belgium
| | - Ann Van Soom
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, University of Ghent, Salisburylaan 133, B-9820, Merelbeke, Belgium
| | - Krishna Chaitanya Pavani
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, University of Ghent, Salisburylaan 133, B-9820, Merelbeke, Belgium; Department for Reproductive Medicine, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Gent, Belgium.
| | - Luc Peelman
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820, Merelbeke, Belgium.
| |
Collapse
|
2
|
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
|
3
|
Shen Z, Hou Y, Zhao G, Tan L, Chen J, Dong Z, Ni C, Pei L. Physiological functions of glucose transporter-2: From cell physiology to links with diabetes mellitus. Heliyon 2024; 10:e25459. [PMID: 38333863 PMCID: PMC10850595 DOI: 10.1016/j.heliyon.2024.e25459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 01/26/2024] [Accepted: 01/26/2024] [Indexed: 02/10/2024] Open
Abstract
Glucose is a sugar crucial for human health since it participates in many biochemical reactions. It produces adenosine 5'-triphosphate (ATP) and nucleosides through glucose metabolic and pentose phosphate pathways. These processes require many transporter proteins to assist in transferring glucose across cells, and the most notable ones are glucose transporter-2 (GLUT-2) and sodium/glucose cotransporter 1 (SGLT1). Glucose enters small intestinal epithelial cells from the intestinal lumen by crossing the brush boundary membrane via the SGLT1 cotransporter. It exits the cells by traversing the basolateral membrane through the activity of the GLUT-2 transporter, supplying energy throughout the body. Dysregulation of these glucose transporters is involved in the pathogenesis of several metabolic diseases, such as diabetes. Natural loss of GLUT-2 or its downregulation causes abnormal blood glucose concentrations in the body, such as fasting hypoglycemia and glucose tolerance. Therefore, understanding GLUT-2 physiology is necessary for exploring the mechanisms of diabetes and targeted treatment development. This article reviews how the apical GLUT-2 transporter maintains normal physiological functions of the human body and the adaptive changes this transporter produces under pathological conditions such as diabetes.
Collapse
Affiliation(s)
- Zhean Shen
- Xinjiang Institute of Technology, Aksu, China
| | - Yingze Hou
- Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Guo Zhao
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Libi Tan
- School of Laboratory Medicine and Biotechnology, Southern Medical University, China
| | - Jili Chen
- Department of Nutrition and Food Hygiene School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Ziqi Dong
- School of Public Health, Peking University Health Science Center, Beijing 100021, China
| | - Chunxiao Ni
- Hangzhou Lin ‘an District Center for Disease Control and Prevention, Hangzhou, China
| | | |
Collapse
|
4
|
Iida A, Tsuda N, Yoshida J, Nomura J, Ratanayotha A, Kawai T, Hondo E. Glucose absorption activity and gene expression of sugar transporters in the trophotaenia of the viviparous teleost Xenotoca eiseni. Biochim Biophys Acta Gen Subj 2023; 1867:130464. [PMID: 37717926 DOI: 10.1016/j.bbagen.2023.130464] [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: 07/10/2023] [Revised: 09/07/2023] [Accepted: 09/12/2023] [Indexed: 09/19/2023]
Abstract
In viviparous reproductive systems, nutrient transfer from mother to embryo plays a critical role in the generation of offspring. Herein, we investigated the mother-to-embryo nutrient transfer machinery in the viviparous teleost Xenotoca eiseni, which belongs to the family Goodeidae. The intraovarian embryo absorbs maternal supplements via the hindgut-derived placental structure termed the trophotaenia. Tracer analysis indicated that the trophotaenia can take up glucose analogs in ex vivo cultured embryos. The candidate genes for absorption, sglt1, glut2, atp1a, and atp1b, were determined from published transcriptomes. These genes were expressed in the trophotaenia of X. eiseni embryos. Fluorescent immunohistochemistry of Na+/K+ ATPase indicated the polarity of epithelial cells in the trophotaenia. The presented evidence suggests that the epithelial cell layer transports monosaccharides from the apical membrane of epithelial cells in a basolateral direction. Taken together, this study provides insight into how maternal fish maintain their offspring during gestation and will aid in the development of strategies to improve offspring generation in these fish.
Collapse
Affiliation(s)
- Atsuo Iida
- Department of Animal Sciences Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi 464-8602, Japan; Department of Bioresource Sciences, School of Agricultural Sciences, Nagoya University, Nagoya, Aichi 464-8602, Japan.
| | - Natsuho Tsuda
- Department of Bioresource Sciences, School of Agricultural Sciences, Nagoya University, Nagoya, Aichi 464-8602, Japan
| | - Junki Yoshida
- Department of Animal Sciences Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi 464-8602, Japan
| | - Jumpei Nomura
- Department of Animal Sciences Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi 464-8602, Japan
| | - Adisorn Ratanayotha
- Laboratory of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; Department of Anatomy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkoknoi, Bangkok 10700, Thailand
| | - Takafumi Kawai
- Laboratory of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Eiichi Hondo
- Department of Animal Sciences Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi 464-8602, Japan; Department of Bioresource Sciences, School of Agricultural Sciences, Nagoya University, Nagoya, Aichi 464-8602, Japan
| |
Collapse
|
5
|
Sportiello M, Poindexter A, Reilly EC, Geber A, Lambert Emo K, Jones TN, Topham DJ. Mouse Memory CD8 T Cell Subsets Defined by Tissue-Resident Memory Integrin Expression Exhibit Distinct Metabolic Profiles. Immunohorizons 2023; 7:652-669. [PMID: 37855738 PMCID: PMC10615656 DOI: 10.4049/immunohorizons.2300040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/24/2023] [Indexed: 10/20/2023] Open
Abstract
Tissue-resident memory CD8 T cells (TRM) principally reside in peripheral nonlymphoid tissues, such as lung and skin, and confer protection against a variety of illnesses ranging from infections to cancers. The functions of different memory CD8 T cell subsets have been linked with distinct metabolic pathways and differ from other CD8 T cell subsets. For example, skin-derived memory T cells undergo fatty acid oxidation and oxidative phosphorylation to a greater degree than circulating memory and naive cells. Lung TRMs defined by the cell-surface expression of integrins exist as distinct subsets that differ in gene expression and function. We hypothesize that TRM subsets with different integrin profiles will use unique metabolic programs. To test this, differential expression and pathway analysis were conducted on RNA sequencing datasets from mouse lung TRMs yielding significant differences related to metabolism. Next, metabolic models were constructed, and the predictions were interrogated using functional metabolite uptake assays. The levels of oxidative phosphorylation, mitochondrial mass, and neutral lipids were measured. Furthermore, to investigate the potential relationships to TRM development, T cell differentiation studies were conducted in vitro with varying concentrations of metabolites. These demonstrated that lipid conditions impact T cell survival, and that glucose concentration impacts the expression of canonical TRM marker CD49a, with no effect on central memory-like T cell marker CCR7. In summary, it is demonstrated that mouse resident memory T cell subsets defined by integrin expression in the lung have unique metabolic profiles, and that nutrient abundance can alter differentiation.
Collapse
Affiliation(s)
- Mike Sportiello
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY
- Medical Scientist Training Program, University of Rochester Medical Center, Rochester, NY
| | - Alexis Poindexter
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY
| | - Emma C. Reilly
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY
| | - Adam Geber
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY
- Medical Scientist Training Program, University of Rochester Medical Center, Rochester, NY
| | - Kris Lambert Emo
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY
| | - Taylor N. Jones
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY
| | - David J. Topham
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
6
|
Sun B, Chen H, Xue J, Li P, Fu X. The role of GLUT2 in glucose metabolism in multiple organs and tissues. Mol Biol Rep 2023; 50:6963-6974. [PMID: 37358764 PMCID: PMC10374759 DOI: 10.1007/s11033-023-08535-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 05/17/2023] [Indexed: 06/27/2023]
Abstract
The glucose transporter family has an important role in the initial stage of glucose metabolism; Glucose transporters 2 (GLUTs, encoded by the solute carrier family 2, SLC2A genes) is the major glucose transporter in β-cells of pancreatic islets and hepatocytes but is also expressed in the small intestine, kidneys, and central nervous system; GLUT2 has a relatively low affinity to glucose. Under physiological conditions, GLUT2 transports glucose into cells and allows the glucose concentration to reach balance on the bilateral sides of the cellular membrane; Variation of GLUT2 is associated with various endocrine and metabolic disorders; In this study, we discussed the role of GLUT2 in participating in glucose metabolism and regulation in multiple organs and tissues and its effects on maintaining glucose homeostasis.
Collapse
Affiliation(s)
- Bo Sun
- Endorcrine and Metabolism Department, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Department of Infantile Endocrine Genetic Metabolism, Gansu Maternal and child Health Care Hospital, Lanzhou, 730000, China
| | - Hui Chen
- Endorcrine and Metabolism Department, Lanzhou University Second Hospital, Lanzhou, 730000, China.
| | - Jisu Xue
- EndEnorcrine and Metabolism Department, Shenzhen Bao 'an People's Hospital (Group), Shenzhen, 518100, China
| | - Peiwu Li
- Key Laboratory of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Xu Fu
- Key Laboratory of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, 730000, China
| |
Collapse
|
7
|
Sudhakaran G, Rajesh R, Almutairi BO, Arokiyaraj S, Gopinath P, Arockiaraj J. Nimbin analogs stimulate glucose uptake and glycogen storage in the insulin signalling cascade by enhancing the IRTK, PI3K and Glut-4 mechanism in myotubes. Tissue Cell 2023; 82:102104. [PMID: 37207372 DOI: 10.1016/j.tice.2023.102104] [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: 02/13/2023] [Revised: 05/05/2023] [Accepted: 05/05/2023] [Indexed: 05/21/2023]
Abstract
BACKGROUND Diabetes Mellitus is a metabolic disorder characterized by insulin dysfunction or failure of the pancreatic β-cells to produce insulin resulting in hyperglycemia. Adverse effects of hyperglycemic conditions continue to be common, reducing treatment adherence. Intensified therapies are required for the constant loss of endogenous islet reserve. AIM This study aimed to evaluate the effect of Nimbin semi-natural analogs (N2, N5, N7, and N8) from A. indica on high glucose-induced ROS and apoptosis with insulin resistance in L6 myotubes evaluated along with Wortmannin and Genistein inhibitors and the expression of key genes in the insulin signalling pathway. MATERIALS AND METHODS The analogs were screened for anti-oxidant and anti-diabetic activity using cell-free assays; The ability of analogs to suppress ROS and prevent apoptosis induced by High glucose and uptake glucose and glycogen storage in L6 myotubes was evaluated using DCFH-DA, AO-PI and 2NBDG staining. Further, the glucose uptake was performed in the presence of Insulin Receptor Tyrosine Kinase (IRTK) inhibitors, and the expression of key genes PI3K, Glut-4, GS and IRTK in the insulin signalling pathway were evaluated. KEY FINDINGS The Nimbin analogs were not toxic to the L6 cells, and the analogs could scavenge ROS and suppress cellular damage induced due to high glucose. Enhanced glucose uptake was observed in N2, N5 and N7 compared to N8. The maximum activity of optimum concentration was found to be 100 μM. The N2, N5 and N7 showed an increase in IRTK, which is equivalent to insulin at a concentration of 100 µM. The IRTK inhibitor with Genistein (50 µM) confirmed the presence of IRTK-dependent glucose transport activation; it also supports the expression of key genes PI3K, Glut-4, GS and IRTK. As a result of PI3K activation, N2, N5, and N7 exhibited the insulin-mimetic effect by enhancing glucose uptake and glycogen conversion regulating glucose metabolism. SIGNIFICANCE N2, N5 and N7 could therapeutically benefit against insulin resistance by glucose metabolism modulation, insulin secretion, β-cell stimulation, inhibition of gluconeogenic enzymes and ROS protection.
Collapse
Affiliation(s)
- Gokul Sudhakaran
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur 603203, Tamil Nadu, India
| | - Ravi Rajesh
- Department of Chemistry, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur 603203, Tamil Nadu, India
| | - Bader O Almutairi
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, 11451 Riyadh, Saudi Arabia
| | - Selvaraj Arokiyaraj
- Department of Food Science & Biotechnology, Sejong University, Seoul 05006, South Korea
| | - Pushparathinam Gopinath
- Department of Chemistry, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur 603203, Tamil Nadu, India.
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur 603203, Tamil Nadu, India.
| |
Collapse
|
8
|
Dong Z, Chen X, Zhuo R, Li Y, Zhou Z, Sun Y, Liu Y, Liu M. Efficient manipulation of gene expression using Natronobacterium gregoryi Argonaute in zebrafish. BMC Biol 2023; 21:95. [PMID: 37095525 PMCID: PMC10127001 DOI: 10.1186/s12915-023-01599-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/13/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND Natronobacterium gregoryi Argonaute (NgAgo) was found to reduce mRNA without generating detectable DNA double-strand breaks in a couple of endogenous genes in zebrafish, suggesting its potential as a tool for gene knockdown. However, little is known about how it interacts with nucleic acid molecules to interfere with gene expression. RESULTS In this study, we first confirmed that coinjection of NgAgo and gDNA downregulated target genes, generated gene-specific phenotypes and verified some factors (including 5' phosphorylation, GC ratio, and target positions) of gDNAs affecting gene downregulation. Therein, the sense and antisense gDNAs were equally effective, suggesting that NgAgo possibly binds to DNA. NgAgo-VP64 with gDNAs targeting promoters upregulated the target genes, further providing evidence that NgAgo interacts with genomic DNA and controls gene transcription. Finally, we explain the downregulation of NgAgo/gDNA target genes by interference with the process of gene transcription, which differs from that of morpholino oligonucleotides. CONCLUSIONS The present study provides conclusions that NgAgo may target genomic DNA and that target positions and the gDNA GC ratio influence its regulation efficiency.
Collapse
Affiliation(s)
- Zhangji Dong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong Jiangsu, 226001, China.
| | - Xu Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong Jiangsu, 226001, China
| | - Run Zhuo
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong Jiangsu, 226001, China
| | - Yuanyuan Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong Jiangsu, 226001, China
| | - Zhihao Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong Jiangsu, 226001, China
| | - Ying Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong Jiangsu, 226001, China
| | - Yan Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong Jiangsu, 226001, China
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong Jiangsu, 226001, China.
| |
Collapse
|
9
|
Sudhakaran G, Rajesh R, Guru A, Arasu MV, Gopinath P, Arockiaraj J. Nimbin analogs N5 and N7 regulate the expression of lipid metabolic genes and inhibit lipid accumulation in high-fat diet-induced zebrafish larvae: An antihyperlipidemic study. Tissue Cell 2023; 80:102000. [PMID: 36542946 DOI: 10.1016/j.tice.2022.102000] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/05/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Excess accumulation of lipids leads to obesity. Triterpenoids are a group of plant compounds which poses various biological activities. The biological activities of Nimbin analogs N5 and N7 were addressed in this study on inhibiting lipid aggregation and underlying the derivatives molecular mechanisms for a therapeutical approach. AIM This study aims to evaluate the anti-adipogenic activity of semi-natural Nimbin analogs, N5 and N7, on zebrafish larvae induced with oxidative stress due to a high-fat diet (HFD) and adipogenesis using specific fluorescent stains. MATERIALS AND METHODS Zebrafish at 4 days post fertilized (dpf) larvae were divided into groups for the HFD diet along with exposure to various concentrations of N5 and N7. HFD induced accumulation of neutral lipids and triglycerides (Oil Red O and Nile red staining, respectively) with weight gain, which generated intracellular ROS (DCFH-DA staining) and superoxide anion production (DHE staining) with depleted glutathione levels (NDA staining) were assayed. HFD exposure promoted the accumulation of inflammatory macrophages (Neutral red staining) and impaired glucose metabolism (2NBDG staining). The ability of N5 and N7 to reduce total regulating lipogenic specific genes C/EBP-α, SREBP-1 and FAS were evaluated using relative gene expression. KEY FINDINGS The Nimbin analogues N5 and N7 suppressed adipogenesis, forming intracellular ROS and superoxide anion while simultaneously restoring glutathione levels. The analogues significantly lowered total TC and TG levels, prevented inflammatory macrophage build-up and boosted glucose absorption. Also, N5 and N7 down-regulate the lipogenic-specific genes. SIGNIFICANCE Nimbin analogs N5 and N7 enhance lipolysis and inhibit adipogenesis in in-vivo zebrafish larvae model.
Collapse
Affiliation(s)
- Gokul Sudhakaran
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamil Nadu, India
| | - Ravi Rajesh
- Department of Chemistry, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamil Nadu, India
| | - Ajay Guru
- Department of Conservative Dentistry and Endodontics, Saveetha Dental College and Hospitals, SIMATS, Chennai 600077, Tamil Nadu, India
| | - Mariadhas Valan Arasu
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Pusparathinam Gopinath
- Department of Chemistry, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamil Nadu, India.
| | - Jesu Arockiaraj
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamil Nadu, India.
| |
Collapse
|
10
|
Okada R, Adachi S, Takiya Y, Iwasaki R, Hirota A, Kikuyama S. Involvement of glucose in freeze tolerance in the Japanese tree frog Hyla japonica. Dev Growth Differ 2022; 64:486-493. [PMID: 36193878 DOI: 10.1111/dgd.12814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/28/2022] [Accepted: 08/16/2022] [Indexed: 01/04/2023]
Abstract
Glycerol and aquaporin 9 (aquaglyceroporin) are known to be involved in freeze tolerance in the Japanese tree frog Hyla japonica. However, the regulatory mechanisms of freeze tolerance in this species have not been fully elucidated. In the present study, we focused on the inter- and intracellular dynamics of glucose to analyze the role of glucose and glucose-related proteins such as transporter and metabolic enzymes in freeze tolerance. Serum glucose concentrations were compared among the frogs that were nonhibernating, hibernating, and thawed after freezing at -4°C for 6 hr. Serum concentrations of glucose in thawed frogs were significantly higher than those in hibernating and nonhibernating, active frogs. Periodic acid-Schiff staining showed that the accumulation of glycogen in the hepatocytes increased before hibernation and decreased after freezing and thawing. Quantitative RT-PCR analysis using the liver showed that, compared with active frogs, the type 2 glucose transporter gene (glut2) was upregulated in frozen frogs, the liver glycogen phosphorylase gene (pygl) was upregulated in frozen or thawed frogs, and the type 2 glycogen synthase gene (gys2) was upregulated in hibernating frogs. Immunohistochemistry of liver sections showed that, compared with nonhibernating frogs, Glut2 proteins were clearly increased most likely on the plasma membrane of hepatocytes in hibernating frogs and further increased by freezing, then decreased after thawing. These results suggest the possibility that glucose acts as a cryoprotectant in H. japonica.
Collapse
Affiliation(s)
- Reiko Okada
- Department of Biological Science, Faculty of Science, Shizuoka University, Shizuoka, Japan.,Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka, Japan
| | - Shun Adachi
- Department of Biological Science, Faculty of Science, Shizuoka University, Shizuoka, Japan
| | - Yu Takiya
- Department of Biological Science, Faculty of Science, Shizuoka University, Shizuoka, Japan
| | - Ryohei Iwasaki
- Department of Biological Science, Faculty of Science, Shizuoka University, Shizuoka, Japan
| | - Atsushi Hirota
- Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka, Japan.,Bureau of Construction, Tokyo Metropolitan Government, Tokyo, Japan
| | - Sakae Kikuyama
- Department of Biology, Faculty of Education and Integrated Sciences, Center for Advanced Biomedical Sciences, Waseda University, Tokyo, Japan
| |
Collapse
|
11
|
Glucocorticoid receptor activation reduces food intake independent of hyperglycemia in zebrafish. Sci Rep 2022; 12:15677. [PMID: 36127383 PMCID: PMC9489701 DOI: 10.1038/s41598-022-19572-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 08/31/2022] [Indexed: 11/08/2022] Open
Abstract
Chronic cortisol exposure suppresses food intake in fish, but the central mechanism(s) involved in appetite regulation are unclear. Stress and the associated increase in cortisol levels increase hepatic gluconeogenesis, leading to hyperglycemia. As hyperglycemia causes a reduction in food intake, we tested the hypothesis that cortisol-induced hyperglycemia suppresses feeding in zebrafish (Danio rerio). We first established that stress-independent hyperglycemia suppressed food intake, and this corresponded with a reduction in the phosphorylation of the nutrient sensor, AMP-activated protein kinase (AMPK) in the brain. Chronic cortisol exposure also led to hyperglycemia and reduced food intake, but the mechanisms were distinct. In cortisol-exposed fish, there were no changes in brain glucose uptake or AMPK phosphorylation. Also, the phosphorylation of Akt and mTOR was reduced along with an increase in redd1, suggesting an enhanced capacity for proteolysis. Loss of the glucocorticoid receptor did not rescue cortisol-mediated feeding suppression but did increase glucose uptake and abolished the changes seen in mTOR phosphorylation and redd1 transcript abundance. Taken together, our results indicate that GR activation enhances brain proteolysis, and the associated amino acids levels, and not hyperglycemia, maybe a key mediator of the feeding suppression in response to chronic cortisol stimulation in zebrafish.
Collapse
|
12
|
Sudhakaran G, Rajesh R, Guru A, Haridevamuthu B, Murugan R, Bhuvanesh N, Wadaan MA, Mahboob S, Juliet A, Gopinath P, Arockiaraj J. Deacetylated nimbin analog N2 fortifies alloxan-induced pancreatic β-cell damage in insulin-resistant zebrafish larvae by upregulating phosphoenolpyruvate carboxykinase (PEPCK) and insulin levels. Toxicol Appl Pharmacol 2022; 454:116229. [PMID: 36089001 DOI: 10.1016/j.taap.2022.116229] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/24/2022]
Abstract
This study aims to evaluate the protective behaviour of N2, a semi-natural analog of nimbin, for its anti-diabetic efficacy against alloxan-induced oxidative damage and β-cell dysfunction in in-vivo zebrafish larvae. A 500 μM of alloxan was exposed to zebrafish larvae for 24 h to induce oxidative stress in the pancreatic β-cells and co-exposed with N2 to study the protection of N2 by inhibiting ROS by DCFH-DA, DHE and NDA staining along with Cellular damage, apoptosis and lipid peroxidation. The zebrafish was further exposed to 500 μM alloxan for 72 h to induce β-cell destruction along with depleted glucose uptake and co-exposed to N2 to study the protective mechanism. Glucose levels were estimated, and PCR was used to verify the mRNA expression of phosphoenolpyruvate carboxykinase (PEPCK) and insulin. Alloxan induced (24 h) oxidative stress in the pancreatic β-cells in which N2's co-exposure inhibited ROS by eliminating O-₂ radicals and restoring the glutathione levels, thus preventing cellular damage and lipid peroxidation. The zebrafish exposed to 500 μM alloxan for 72 h was observed with β-cell destruction along with depleted glucose uptake when stained with 2NBDG, wherein N2 was able to protect the pancreatic β-cells from oxidative damage, promoted high glucose uptake and reduced glucose levels. N2 stimulated insulin production and downregulated PEPCK by inhibiting gluconeogenesis, attenuating post-prandial hyperglycemia. N2 may contribute to anti-oxidant protection against alloxan-induced β-cell damage and anti-hyperglycemic activity, restoring insulin function and suppressing PEPCK expression.
Collapse
Affiliation(s)
- Gokul Sudhakaran
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, 603 203 Chennai, Tamil Nadu, India
| | - Ravi Rajesh
- Department of Chemistry, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 203 Chennai, Tamil Nadu, India
| | - Ajay Guru
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, 603 203 Chennai, Tamil Nadu, India
| | - B Haridevamuthu
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, 603 203 Chennai, Tamil Nadu, India
| | - Raghul Murugan
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, 603 203 Chennai, Tamil Nadu, India
| | - Nattamai Bhuvanesh
- Department of Chemistry, Texas A&M University, College Station, TX 77842, USA
| | - Mohammad Ahmad Wadaan
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Shalid Mahboob
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Annie Juliet
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, University Station A4800, Austin, TX 78712, USA
| | - Pushparathinam Gopinath
- Department of Chemistry, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 203 Chennai, Tamil Nadu, India.
| | - Jesu Arockiaraj
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, 603 203 Chennai, Tamil Nadu, India.
| |
Collapse
|
13
|
Kim D, Kim J, Yu YS, Kim YR, Baek SH, Won KJ. Systemic approaches using single cell transcriptome reveal that C/EBPγ regulates autophagy under amino acid starved condition. Nucleic Acids Res 2022; 50:7298-7309. [PMID: 35801910 PMCID: PMC9303372 DOI: 10.1093/nar/gkac593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 06/17/2022] [Accepted: 06/24/2022] [Indexed: 11/14/2022] Open
Abstract
Autophagy, a catabolic process to remove unnecessary or dysfunctional organelles, is triggered by various signals including nutrient starvation. Depending on the types of the nutrient deficiency, diverse sensing mechanisms and signaling pathways orchestrate for transcriptional and epigenetic regulation of autophagy. However, our knowledge about nutrient type-specific transcriptional regulation during autophagy is limited. To understand nutrient type-dependent transcriptional mechanisms during autophagy, we performed single cell RNA sequencing (scRNAseq) in the mouse embryonic fibroblasts (MEFs) with or without glucose starvation (GS) as well as amino acid starvation (AAS). Trajectory analysis using scRNAseq identified sequential induction of potential transcriptional regulators for each condition. Gene regulatory rules inferred using TENET newly identified CCAAT/enhancer binding protein γ (C/EBPγ) as a regulator of autophagy in AAS, but not GS, condition, and knockdown experiment confirmed the TENET result. Cell biological and biochemical studies validated that activating transcription factor 4 (ATF4) is responsible for conferring specificity to C/EBPγ for the activation of autophagy genes under AAS, but not under GS condition. Together, our data identified C/EBPγ as a previously unidentified key regulator under AAS-induced autophagy.
Collapse
Affiliation(s)
- Dongha Kim
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea.,Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Junil Kim
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark.,School of Systems Biomedical Science, Soongsil University, 369 Sangdo-Ro, Dongjak-Gu, Seoul 06978, Republic of Korea
| | - Young Suk Yu
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yong Ryoul Kim
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Sung Hee Baek
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyoung-Jae Won
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
14
|
Matsunaga Y, Takahashi K, Takahashi Y, Hatta H. Effects of glucose ingestion at different frequencies on glycogen recovery in mice during the early hours post exercise. J Int Soc Sports Nutr 2021; 18:69. [PMID: 34743706 PMCID: PMC8574022 DOI: 10.1186/s12970-021-00467-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 10/25/2021] [Indexed: 11/24/2022] Open
Abstract
Background When a high-carbohydrate diet is ingested, whether as small frequent snacks or as large meals, there is no difference between the two with respect to post-exercise glycogen storage for a period of 24 h. However, the effect of carbohydrate intake frequency on glycogen recovery a few hours after exercise is not clear. Athletes need to recover glycogen quickly after physical exercise as they sometimes exercise multiple times a day. The aim of this study was to determine the effect of carbohydrate intake at different frequencies on glycogen recovery during the first few hours after exercise. Methods After 120 min of fasting, 6-week-old male ICR mice were subjected to treadmill running exercise (20 m/min for 60 min) to decrease the levels of muscle and liver glycogen. Mice were then given glucose as a bolus (1.2 mg/g of body weight [BW], immediately after exercise) or as a pulse (1.2 mg/g of BW, every 15 min × 4 times). Following this, the blood, tissue, and exhaled gas samples were collected. Results In the bolus group, blood glucose concentration was significantly lower and plasma insulin concentration was significantly higher than those in the pulse group (p < 0.05). The plantaris muscle glycogen concentration in the bolus group was 25.3% higher than that in the pulse group at 60 min after glucose ingestion (p < 0.05). Liver glycogen concentration in the pulse group was significantly higher than that in the bolus group at 120 min after glucose ingestion (p < 0.05). Conclusions The present study showed that ingesting a large amount of glucose immediately after exercise increased insulin secretion and enhanced muscle glycogen recovery, whereas frequent and small amounts of glucose intake was shown to enhance liver glycogen recovery.
Collapse
Affiliation(s)
- Yutaka Matsunaga
- Department of Sports Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan.
| | - Kenya Takahashi
- Department of Sports Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan
| | - Yumiko Takahashi
- Department of Sports Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan
| | - Hideo Hatta
- Department of Sports Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan
| |
Collapse
|
15
|
Dakic T, Lakic I, Zec M, Takic M, Stojiljkovic M, Jevdjovic T. Fructose-rich diet and walnut supplementation differently regulate rat hypothalamic and hippocampal glucose transporters expression. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2021; 101:5984-5991. [PMID: 33856052 DOI: 10.1002/jsfa.11252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/06/2021] [Accepted: 04/15/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Nutritional modulations may be considered a strategy to protect mental health. Neuronal homeostasis is highly dependent on the availability of glucose, which represents the primary energy source for the brain. In this study, we evaluated the effects of walnut intake and fructose-rich diet on the expression of glucose transporters (GLUTs) in two rat brain regions: hypothalamus and hippocampus. RESULTS Our results show that walnut supplementation of fructose-fed animals restored the hypothalamic content of GLUT1 and GLUT3 protein. Furthermore, walnut intake did not affect increased hypothalamic GLUT2 content upon fructose consumption. These effects were accompanied by distinctive alterations of hippocampal GLUTs levels. Specifically, walnut intake increased GLUT1 content, whereas GLUT2 protein was decreased within the rat hippocampus after both individual and combined treatments. CONCLUSION Overall, our study suggests that walnut supplementation exerted modulatory effects on the glucose transporters within specific brain regions in the presence of developed metabolic disorder. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Tamara Dakic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry 'Ivan Djaja', Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Iva Lakic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry 'Ivan Djaja', Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Manja Zec
- Centre of Excellence for Nutrition and Metabolism Research, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Marija Takic
- Centre of Excellence for Nutrition and Metabolism Research, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Mojca Stojiljkovic
- Department for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Tanja Jevdjovic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry 'Ivan Djaja', Faculty of Biology, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
16
|
Cooman T, Bergeron SA, Coltogirone R, Horstick E, Arroyo L. Evaluation of fentanyl toxicity and metabolism using a zebrafish model. J Appl Toxicol 2021; 42:706-714. [PMID: 34647333 DOI: 10.1002/jat.4253] [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: 08/29/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 01/02/2023]
Abstract
The increased abuse of novel drugs has created a critical need for cheap and rapid in vivo models to understand whole organism drug-induced toxicity and metabolic impacts. One such model is zebrafish, which share many similarities to human. Assays have been developed for behavioral, toxicity, and metabolism elucidation following chemical exposure. The zebrafish model provides the advantage of assessing these parameters within a single study. Previous zebrafish studies have evaluated the behavioral effects of fentanyl, but not developmental toxicity and its relation to metabolism. In this study, we evaluate the effects of fentanyl on the development of wild-type (TL strain) zebrafish and its metabolism over 4 days. Fertilized eggs were exposed to six concentrations of fentanyl (0.01, 0.1, 1, 10, 50, and 100 μM) through embryo media incubated at 28-29°C. Observations included egg coagulation, somite formation, heartbeat, tail and yolk morphology, pericardial formation, and swim bladder inflation. The incubation media was analyzed for the presence of metabolites using a targeted metabolomics approach. Fentanyl concentration caused significant effects on survival and development, with notable defects to the tail, yolk, and pericardium at 50 and 100 μM. Despropionyl fentanyl (4-ANPP), β-hydroxy fentanyl, and norfentanyl were detected in zebrafish larvae. We present a single in vivo model to assess toxicity and metabolism of fentanyl exposure in a vertebrate model system. Our findings provide a foundation for further investigations into fentanyl's mechanism of action and translation to human drug exposure.
Collapse
Affiliation(s)
- Travon Cooman
- Department of Forensic and Investigative Science, West Virginia University, Morgantown, West Virginia, USA
| | - Sadie A Bergeron
- Department of Biology, West Virginia University, Morgantown, West Virginia, USA
| | - Rebecca Coltogirone
- Department of Biology, West Virginia University, Morgantown, West Virginia, USA
| | - Eric Horstick
- Department of Biology, West Virginia University, Morgantown, West Virginia, USA
| | - Luis Arroyo
- Department of Forensic and Investigative Science, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
17
|
Matsunaga Y, Koyama S, Takahashi K, Takahashi Y, Shinya T, Yoshida H, Hatta H. Effects of post-exercise glucose ingestion at different solution temperatures on glycogen repletion in mice. Physiol Rep 2021; 9:e15041. [PMID: 34553503 PMCID: PMC8459029 DOI: 10.14814/phy2.15041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 11/24/2022] Open
Abstract
Carbohydrate ingestion is essential for glycogen recovery after exercise. Although studies have investigated methods for enhancement of glycogen repletion with regard to nutrients and their amounts, no studies have examined the effect of temperature of the ingested solution on glycogen recovery. Therefore, this study aimed to investigate the effect of the temperature of glucose solution ingested after exercise on glycogen recovery. Seven-week-old male ICR mice were fasted for 16 h and subjected to treadmill running exercise (20 m/min for 60 min) to decrease glycogen storage. Then, the mice were administered glucose (1.5 mg/g body weight) at three different solution temperatures: 4°C, cold solution group (Cold); 37°C, mild solution group (Mild); and 55°C, hot solution group (Hot). Our results revealed that blood glucose, plasma insulin, and muscle glycogen concentrations did not differ among the three groups. In contrast, liver glycogen concentration in the Hot group was significantly higher than that in the post-exercise and Cold groups (p < 0.05). Furthermore, portal glucose concentration was significantly higher in the Hot group than in the Cold group (p < 0.01). These observations suggest that postexercise muscle glycogen repletion occurs regardless of glucose solution temperature, and that ingesting hot glucose solution after exercise can be an effective means for liver glycogen repletion compared with cold glucose solution ingestion.
Collapse
Affiliation(s)
| | - Sho Koyama
- Department of Sports SciencesThe University of TokyoTokyoJapan
| | - Kenya Takahashi
- Department of Sports SciencesThe University of TokyoTokyoJapan
| | | | - Terunaga Shinya
- Department of Sports SciencesThe University of TokyoTokyoJapan
| | - Hiroki Yoshida
- Department of Sports SciencesThe University of TokyoTokyoJapan
| | - Hideo Hatta
- Department of Sports SciencesThe University of TokyoTokyoJapan
| |
Collapse
|
18
|
A Great Catch for Investigating Inborn Errors of Metabolism-Insights Obtained from Zebrafish. Biomolecules 2020; 10:biom10091352. [PMID: 32971894 PMCID: PMC7564250 DOI: 10.3390/biom10091352] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/18/2020] [Accepted: 09/19/2020] [Indexed: 12/14/2022] Open
Abstract
Inborn errors of metabolism cause abnormal synthesis, recycling, or breakdown of amino acids, neurotransmitters, and other various metabolites. This aberrant homeostasis commonly causes the accumulation of toxic compounds or depletion of vital metabolites, which has detrimental consequences for the patients. Efficient and rapid intervention is often key to survival. Therefore, it requires useful animal models to understand the pathomechanisms and identify promising therapeutic drug targets. Zebrafish are an effective tool to investigate developmental mechanisms and understanding the pathophysiology of disorders. In the past decades, zebrafish have proven their efficiency for studying genetic disorders owing to the high degree of conservation between human and zebrafish genes. Subsequently, several rare inherited metabolic disorders have been successfully investigated in zebrafish revealing underlying mechanisms and identifying novel therapeutic targets, including methylmalonic acidemia, Gaucher’s disease, maple urine disorder, hyperammonemia, TRAPPC11-CDGs, and others. This review summarizes the recent impact zebrafish have made in the field of inborn errors of metabolism.
Collapse
|
19
|
Glucose transporters in brain in health and disease. Pflugers Arch 2020; 472:1299-1343. [PMID: 32789766 PMCID: PMC7462931 DOI: 10.1007/s00424-020-02441-x] [Citation(s) in RCA: 254] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/20/2020] [Accepted: 07/24/2020] [Indexed: 12/15/2022]
Abstract
Energy demand of neurons in brain that is covered by glucose supply from the blood is ensured by glucose transporters in capillaries and brain cells. In brain, the facilitative diffusion glucose transporters GLUT1-6 and GLUT8, and the Na+-d-glucose cotransporters SGLT1 are expressed. The glucose transporters mediate uptake of d-glucose across the blood-brain barrier and delivery of d-glucose to astrocytes and neurons. They are critically involved in regulatory adaptations to varying energy demands in response to differing neuronal activities and glucose supply. In this review, a comprehensive overview about verified and proposed roles of cerebral glucose transporters during health and diseases is presented. Our current knowledge is mainly based on experiments performed in rodents. First, the functional properties of human glucose transporters expressed in brain and their cerebral locations are described. Thereafter, proposed physiological functions of GLUT1, GLUT2, GLUT3, GLUT4, and SGLT1 for energy supply to neurons, glucose sensing, central regulation of glucohomeostasis, and feeding behavior are compiled, and their roles in learning and memory formation are discussed. In addition, diseases are described in which functional changes of cerebral glucose transporters are relevant. These are GLUT1 deficiency syndrome (GLUT1-SD), diabetes mellitus, Alzheimer’s disease (AD), stroke, and traumatic brain injury (TBI). GLUT1-SD is caused by defect mutations in GLUT1. Diabetes and AD are associated with changed expression of glucose transporters in brain, and transporter-related energy deficiency of neurons may contribute to pathogenesis of AD. Stroke and TBI are associated with changes of glucose transporter expression that influence clinical outcome.
Collapse
|
20
|
Chou LY, Chao YM, Peng YC, Lin HC, Wu YL. Glucosamine Enhancement of BDNF Expression and Animal Cognitive Function. Molecules 2020; 25:molecules25163667. [PMID: 32806562 PMCID: PMC7465318 DOI: 10.3390/molecules25163667] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/19/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is an important factor for memory consolidation and cognitive function. Protein kinase A (PKA) signaling interacts significantly with BDNF-provoked downstream signaling. Glucosamine (GLN), a common dietary supplement, has been demonstrated to perform a variety of beneficial physiological functions. In the current study, an in vivo model of 7-week-old C57BL/6 mice receiving daily intraperitoneal injection of GLN (0, 3, 10 and 30 mg/animal) was subjected to the novel object recognition test in order to determine cognitive performance. GLN significantly increased cognitive function. In the hippocampus GLN elevated tissue cAMP concentrations and CREB phosphorylation, and upregulated the expression of BDNF, CREB5 and the BDNF receptor TrkB, but it reduced PDE4B expression. With the in vitro model in the HT22 hippocampal cell line, GLN exposure significantly increased protein and mRNA levels of BDNF and CREB5 and induced cAMP responsive element (CRE) reporter activity; the GLN-mediated BDNF expression and CRE reporter induction were suppressed by PKA inhibitor H89. Our current findings suggest that GLN can exert a cognition-enhancing function and this may act at least in part by upregulating the BDNF levels via a cAMP/PKA/CREB-dependent pathway.
Collapse
Affiliation(s)
- Lien-Yu Chou
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan; (L.-Y.C.), (Y.-M.C.); (H.-C.L.)
| | - Yu-Ming Chao
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan; (L.-Y.C.), (Y.-M.C.); (H.-C.L.)
| | - Yen-Chun Peng
- Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan;
| | - Hui-Ching Lin
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan; (L.-Y.C.), (Y.-M.C.); (H.-C.L.)
| | - Yuh-Lin Wu
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan; (L.-Y.C.), (Y.-M.C.); (H.-C.L.)
- Correspondence: ; Tel.: +886-2-2826-7081; Fax: +886-2-2826-4049
| |
Collapse
|
21
|
Wang X, Yang XL, Liu KC, Sheng WL, Xia Q, Wang RC, Chen XQ, Zhang Y. Effects of streptozotocin on pancreatic islet β-cell apoptosis and glucose metabolism in zebrafish larvae. FISH PHYSIOLOGY AND BIOCHEMISTRY 2020; 46:1025-1038. [PMID: 31993854 DOI: 10.1007/s10695-020-00769-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/21/2020] [Indexed: 06/10/2023]
Abstract
Type 1 diabetes is characterized by an increase in blood glucose levels resulting from damage to β cells in pancreatic islets and the consequent absolute insufficiency of insulin. Animal models of type 1 diabetes were usually established using drugs toxic to β cells, such as streptozotocin (STZ). To assess the application of zebrafish larvae in diabetes research, we explore the effects of STZ on pancreatic islets and glucose metabolism in zebrafish larvae. STZ was microinjected into the pericardial cavity of zebrafish larvae on alternate days for three times. At 2 days after the whole series of STZ injection (12 dpf), free-glucose level in larvae tissue shows a significant increase, and the fluorescence signal in immunohistochemistry, which indicates the insulin expression, was significantly weaker compared with the solution-injected control. Obvious apoptosis signals were also observed in the location of pancreatic islet, and insulin content decreased to be undetectable in STZ-injected larvae. Gene expression level of ins decreased to half of the solution injection control and that of casp3a was upregulated by 2.20-fold. Expression level of glut2 and gck decreased to 0.312-fold and 0.093-fold, respectively. pck1 was upregulated by 2.533-fold in STZ-injected larvae. By tracking detection, we found the free-glucose level in STZ-injected larvae gradually approached the level of the solution injection control and the insulin content recovered at 6 days post-STZ injection (16 dpf). Consistent with the change of the glucose level, the regeneration rate of the caudal fin in the STZ-injected group decreased initially, but recovered and accelerated gradually finally at 8 days post-amputation (20 dpf). These results indicate the generation of a transient hyperglycemia model due to β-cell apoptosis caused by STZ, which is abated by the vigorous regeneration ability of β cells in zebrafish larvae.
Collapse
Affiliation(s)
- Xue Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan, 250103, Shandong Province, People's Republic of China
- Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, Jinan, 250103, Shandong Province, China
| | - Xue-Liang Yang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan, 250103, Shandong Province, People's Republic of China
- Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, Jinan, 250103, Shandong Province, China
| | - Ke-Chun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan, 250103, Shandong Province, People's Republic of China
- Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, Jinan, 250103, Shandong Province, China
| | - Wen-Long Sheng
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan, 250103, Shandong Province, People's Republic of China
- Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, Jinan, 250103, Shandong Province, China
| | - Qing Xia
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan, 250103, Shandong Province, People's Republic of China
- Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, Jinan, 250103, Shandong Province, China
| | - Rong-Chun Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan, 250103, Shandong Province, People's Republic of China
- Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, Jinan, 250103, Shandong Province, China
| | - Xi-Qiang Chen
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan, 250103, Shandong Province, People's Republic of China
- Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, Jinan, 250103, Shandong Province, China
| | - Yun Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan, 250103, Shandong Province, People's Republic of China.
- Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, Jinan, 250103, Shandong Province, China.
| |
Collapse
|
22
|
Brun NR, van Hage P, Hunting ER, Haramis APG, Vink SC, Vijver MG, Schaaf MJM, Tudorache C. Polystyrene nanoplastics disrupt glucose metabolism and cortisol levels with a possible link to behavioural changes in larval zebrafish. Commun Biol 2019; 2:382. [PMID: 31646185 PMCID: PMC6802380 DOI: 10.1038/s42003-019-0629-6] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 09/23/2019] [Indexed: 01/05/2023] Open
Abstract
Plastic nanoparticles originating from weathering plastic waste are emerging contaminants in aquatic environments, with unknown modes of action in aquatic organisms. Recent studies suggest that internalised nanoplastics may disrupt processes related to energy metabolism. Such disruption can be crucial for organisms during development and may ultimately lead to changes in behaviour. Here, we investigated the link between polystyrene nanoplastic (PSNP)-induced signalling events and behavioural changes. Larval zebrafish exhibited PSNP accumulation in the pancreas, which coincided with a decreased glucose level. By using hyperglycemic and glucocorticoid receptor (Gr) mutant larvae, we demonstrate that the PSNP-induced disruption in glucose homoeostasis coincided with increased cortisol secretion and hyperactivity in challenge phases. Our work sheds new light on a potential mechanism underlying nanoplastics toxicity in fish, suggesting that the adverse effect of PSNPs are at least in part mediated by Gr activation in response to disrupted glucose homeostasis, ultimately leading to aberrant locomotor activity.
Collapse
Affiliation(s)
- Nadja R. Brun
- Institute of Environmental Sciences (CML), Leiden University, Leiden, The Netherlands
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, MA USA
| | - Patrick van Hage
- Institute of Environmental Sciences (CML), Leiden University, Leiden, The Netherlands
| | | | | | - Suzanne C. Vink
- Institute of Environmental Sciences (CML), Leiden University, Leiden, The Netherlands
| | - Martina G. Vijver
- Institute of Environmental Sciences (CML), Leiden University, Leiden, The Netherlands
| | | | | |
Collapse
|
23
|
Lechermeier CG, Zimmer F, Lüffe TM, Lesch KP, Romanos M, Lillesaar C, Drepper C. Transcript Analysis of Zebrafish GLUT3 Genes, slc2a3a and slc2a3b, Define Overlapping as Well as Distinct Expression Domains in the Zebrafish ( Danio rerio) Central Nervous System. Front Mol Neurosci 2019; 12:199. [PMID: 31507372 PMCID: PMC6718831 DOI: 10.3389/fnmol.2019.00199] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/02/2019] [Indexed: 01/05/2023] Open
Abstract
The transport of glucose across the cell plasma membrane is vital to most mammalian cells. The glucose transporter (GLUT; also called SLC2A) family of transmembrane solute carriers is responsible for this function in vivo. GLUT proteins encompass 14 different isoforms in humans with different cell type-specific expression patterns and activities. Central to glucose utilization and delivery in the brain is the neuronally expressed GLUT3. Recent research has shown an involvement of GLUT3 genetic variation or altered expression in several different brain disorders, including Huntington's and Alzheimer's diseases. Furthermore, GLUT3 was identified as a potential risk gene for multiple psychiatric disorders. To study the role of GLUT3 in brain function and disease a more detailed knowledge of its expression in model organisms is needed. Zebrafish (Danio rerio) has in recent years gained popularity as a model organism for brain research and is now well-established for modeling psychiatric disorders. Here, we have analyzed the sequence of GLUT3 orthologs and identified two paralogous genes in the zebrafish, slc2a3a and slc2a3b. Interestingly, the Glut3b protein sequence contains a unique stretch of amino acids, which may be important for functional regulation. The slc2a3a transcript is detectable in the central nervous system including distinct cellular populations in telencephalon, diencephalon, mesencephalon and rhombencephalon at embryonic and larval stages. Conversely, the slc2a3b transcript shows a rather diffuse expression pattern at different embryonic stages and brain regions. Expression of slc2a3a is maintained in the adult brain and is found in the telencephalon, diencephalon, mesencephalon, cerebellum and medulla oblongata. The slc2a3b transcripts are present in overlapping as well as distinct regions compared to slc2a3a. Double in situ hybridizations were used to demonstrate that slc2a3a is expressed by some GABAergic neurons at embryonic stages. This detailed description of zebrafish slc2a3a and slc2a3b expression at developmental and adult stages paves the way for further investigations of normal GLUT3 function and its role in brain disorders.
Collapse
Affiliation(s)
- Carina G Lechermeier
- Child and Adolescent Psychiatry, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany.,Department of Physiological Chemistry, Biocenter, Am Hubland, University of Würzburg, Würzburg, Germany
| | - Frederic Zimmer
- Department of Physiological Chemistry, Biocenter, Am Hubland, University of Würzburg, Würzburg, Germany
| | - Teresa M Lüffe
- Child and Adolescent Psychiatry, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany.,Department of Physiological Chemistry, Biocenter, Am Hubland, University of Würzburg, Würzburg, Germany
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany.,Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.,Department of Neuroscience, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, Netherlands
| | - Marcel Romanos
- Child and Adolescent Psychiatry, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Christina Lillesaar
- Child and Adolescent Psychiatry, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany.,Department of Physiological Chemistry, Biocenter, Am Hubland, University of Würzburg, Würzburg, Germany
| | - Carsten Drepper
- Child and Adolescent Psychiatry, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
24
|
Benson S, Fernandez A, Barth ND, de Moliner F, Horrocks MH, Herrington CS, Abad JL, Delgado A, Kelly L, Chang Z, Feng Y, Nishiura M, Hori Y, Kikuchi K, Vendrell M. SCOTfluors: Small, Conjugatable, Orthogonal, and Tunable Fluorophores for In Vivo Imaging of Cell Metabolism. Angew Chem Int Ed Engl 2019; 58:6911-6915. [PMID: 30924239 PMCID: PMC6563150 DOI: 10.1002/anie.201900465] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Indexed: 12/11/2022]
Abstract
The transport and trafficking of metabolites are critical for the correct functioning of live cells. However, in situ metabolic imaging studies are hampered by the lack of fluorescent chemical structures that allow direct monitoring of small metabolites under physiological conditions with high spatial and temporal resolution. Herein, we describe SCOTfluors as novel small-sized multi-colored fluorophores for real-time tracking of essential metabolites in live cells and in vivo and for the acquisition of metabolic profiles from human cancer cells of variable origin.
Collapse
Affiliation(s)
- Sam Benson
- Centre for Inflammation ResearchThe University of EdinburghEH16 4TJEdinburghUK
| | - Antonio Fernandez
- Centre for Inflammation ResearchThe University of EdinburghEH16 4TJEdinburghUK
| | - Nicole D. Barth
- Centre for Inflammation ResearchThe University of EdinburghEH16 4TJEdinburghUK
| | - Fabio de Moliner
- Centre for Inflammation ResearchThe University of EdinburghEH16 4TJEdinburghUK
| | - Mathew H. Horrocks
- UK Dementia Research Institute and EaStCHEM School of ChemistryThe University of EdinburghEH9 3FJEdinburghUK
| | | | - Jose Luis Abad
- Research Unit on Bioactive MoleculesInstitute for Advanced Chemistry of Catalonia08034BarcelonaSpain
- University of BarcelonaFaculty of Pharmacy, Unit of Pharmaceutical Chemistry (CSIC Associated Unit)BarcelonaSpain
| | - Antonio Delgado
- Research Unit on Bioactive MoleculesInstitute for Advanced Chemistry of Catalonia08034BarcelonaSpain
- University of BarcelonaFaculty of Pharmacy, Unit of Pharmaceutical Chemistry (CSIC Associated Unit)BarcelonaSpain
| | - Lisa Kelly
- Centre for Inflammation ResearchThe University of EdinburghEH16 4TJEdinburghUK
| | - Ziyuan Chang
- Centre for Inflammation ResearchThe University of EdinburghEH16 4TJEdinburghUK
| | - Yi Feng
- Centre for Inflammation ResearchThe University of EdinburghEH16 4TJEdinburghUK
| | | | - Yuichiro Hori
- Graduate School of EngineeringOsaka UniversitySuitaJapan
| | - Kazuya Kikuchi
- Graduate School of EngineeringOsaka UniversitySuitaJapan
| | - Marc Vendrell
- Centre for Inflammation ResearchThe University of EdinburghEH16 4TJEdinburghUK
| |
Collapse
|
25
|
Benson S, Fernandez A, Barth ND, de Moliner F, Horrocks MH, Herrington CS, Abad JL, Delgado A, Kelly L, Chang Z, Feng Y, Nishiura M, Hori Y, Kikuchi K, Vendrell M. SCOTfluors: Small, Conjugatable, Orthogonal, and Tunable Fluorophores for In Vivo Imaging of Cell Metabolism. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201900465] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Sam Benson
- Centre for Inflammation ResearchThe University of Edinburgh EH16 4TJ Edinburgh UK
| | - Antonio Fernandez
- Centre for Inflammation ResearchThe University of Edinburgh EH16 4TJ Edinburgh UK
| | - Nicole D. Barth
- Centre for Inflammation ResearchThe University of Edinburgh EH16 4TJ Edinburgh UK
| | - Fabio de Moliner
- Centre for Inflammation ResearchThe University of Edinburgh EH16 4TJ Edinburgh UK
| | - Mathew H. Horrocks
- UK Dementia Research Institute and EaStCHEM School of ChemistryThe University of Edinburgh EH9 3FJ Edinburgh UK
| | | | - Jose Luis Abad
- Research Unit on Bioactive MoleculesInstitute for Advanced Chemistry of Catalonia 08034 Barcelona Spain
- University of BarcelonaFaculty of Pharmacy, Unit of Pharmaceutical Chemistry (CSIC Associated Unit) Barcelona Spain
| | - Antonio Delgado
- Research Unit on Bioactive MoleculesInstitute for Advanced Chemistry of Catalonia 08034 Barcelona Spain
- University of BarcelonaFaculty of Pharmacy, Unit of Pharmaceutical Chemistry (CSIC Associated Unit) Barcelona Spain
| | - Lisa Kelly
- Centre for Inflammation ResearchThe University of Edinburgh EH16 4TJ Edinburgh UK
| | - Ziyuan Chang
- Centre for Inflammation ResearchThe University of Edinburgh EH16 4TJ Edinburgh UK
| | - Yi Feng
- Centre for Inflammation ResearchThe University of Edinburgh EH16 4TJ Edinburgh UK
| | | | - Yuichiro Hori
- Graduate School of EngineeringOsaka University Suita Japan
| | - Kazuya Kikuchi
- Graduate School of EngineeringOsaka University Suita Japan
| | - Marc Vendrell
- Centre for Inflammation ResearchThe University of Edinburgh EH16 4TJ Edinburgh UK
| |
Collapse
|
26
|
Sant KE, Timme-Laragy AR. Zebrafish as a Model for Toxicological Perturbation of Yolk and Nutrition in the Early Embryo. Curr Environ Health Rep 2019; 5:125-133. [PMID: 29417450 DOI: 10.1007/s40572-018-0183-2] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Developmental toxicity assessments often focus on structural outcomes and overlook subtle metabolic differences which occur during the early embryonic period. Deviant embryonic nutrition can result in later-life disease, including diabetes, obesity, and cardiovascular disease. Prior to placenta-mediated nutrient exchange, the human embryo requires maternally supplied nutritional substrates for growth, called yolk. Here, we compare the biology of the human and zebrafish yolk and review examples of toxicant-mediated perturbation of yolk defects, composition, and utilization. RECENT FINDINGS Zebrafish embryos, like human embryos, have a protruding yolk sac that serves as a nutritional cache. Aberrant yolk morphology is a common qualitative finding in fish embryotoxicity studies, but quantitative assessment and characterization provides an opportunity to uncover mechanistic targets of toxicant effects on embryonic nutrition. The zebrafish and the study of its yolk sac is an excellent model for uncovering toxicant disruptions to early embryonic nutrition and has potential to discover mechanistic insights into the developmental origins of health and disease.
Collapse
Affiliation(s)
- Karilyn E Sant
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Goessman 171, 686 N Pleasant St, Amherst, MA, 01003, USA
| | - Alicia R Timme-Laragy
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Goessman 171, 686 N Pleasant St, Amherst, MA, 01003, USA.
| |
Collapse
|
27
|
Dickmeis T, Feng Y, Mione MC, Ninov N, Santoro M, Spaink HP, Gut P. Nano-Sampling and Reporter Tools to Study Metabolic Regulation in Zebrafish. Front Cell Dev Biol 2019; 7:15. [PMID: 30873407 PMCID: PMC6401643 DOI: 10.3389/fcell.2019.00015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 01/31/2019] [Indexed: 01/09/2023] Open
Abstract
In the past years, evidence has emerged that hallmarks of human metabolic disorders can be recapitulated in zebrafish using genetic, pharmacological or dietary interventions. An advantage of modeling metabolic diseases in zebrafish compared to other "lower organisms" is the presence of a vertebrate body plan providing the possibility to study the tissue-intrinsic processes preceding the loss of metabolic homeostasis. While the small size of zebrafish is advantageous in many aspects, it also has shortcomings such as the difficulty to obtain sufficient amounts for biochemical analyses in response to metabolic challenges. A workshop at the European Zebrafish Principal Investigator meeting in Trento, Italy, was dedicated to discuss the advantages and disadvantages of zebrafish to study metabolic disorders. This perspective article by the participants highlights strategies to achieve improved tissue-resolution for read-outs using "nano-sampling" approaches for metabolomics as well as live imaging of zebrafish expressing fluorescent reporter tools that inform on cellular or subcellular metabolic processes. We provide several examples, including the use of reporter tools to study the heterogeneity of pancreatic beta-cells within their tissue environment. While limitations exist, we believe that with the advent of new technologies and more labs developing methods that can be applied to minimal amounts of tissue or single cells, zebrafish will further increase their utility to study energy metabolism.
Collapse
Affiliation(s)
- Thomas Dickmeis
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Yi Feng
- Centre for Inflammation Research, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland
| | | | - Nikolay Ninov
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | | | - Herman P. Spaink
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Philipp Gut
- Nestlé Research, EPFL Innovation Park, Lausanne, Switzerland
| |
Collapse
|
28
|
Dissecting metabolism using zebrafish models of disease. Biochem Soc Trans 2019; 47:305-315. [PMID: 30700500 DOI: 10.1042/bst20180335] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/18/2018] [Accepted: 01/02/2019] [Indexed: 02/07/2023]
Abstract
Zebrafish (Danio rerio) are becoming an increasingly powerful model organism to study the role of metabolism in disease. Since its inception, the zebrafish model has relied on unique attributes such as the transparency of embryos, high fecundity and conservation with higher vertebrates, to perform phenotype-driven chemical and genetic screens. In this review, we describe how zebrafish have been used to reveal novel mechanisms by which metabolism regulates embryonic development, obesity, fatty liver disease and cancer. In addition, we will highlight how new approaches in advanced microscopy, transcriptomics and metabolomics using zebrafish as a model system have yielded fundamental insights into the mechanistic underpinnings of disease.
Collapse
|
29
|
Sposato V, Canu N, Fico E, Fusco S, Bolasco G, Ciotti MT, Spinelli M, Mercanti D, Grassi C, Triaca V, Calissano P. The Medial Septum Is Insulin Resistant in the AD Presymptomatic Phase: Rescue by Nerve Growth Factor-Driven IRS 1 Activation. Mol Neurobiol 2019; 56:535-552. [PMID: 29736736 PMCID: PMC6334735 DOI: 10.1007/s12035-018-1038-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 03/23/2018] [Indexed: 12/15/2022]
Abstract
Basal forebrain cholinergic neurons (BFCN) are key modulators of learning and memory and are high energy-demanding neurons. Impaired neuronal metabolism and reduced insulin signaling, known as insulin resistance, has been reported in the early phase of Alzheimer's disease (AD), which has been suggested to be "Type 3 Diabetes." We hypothesized that BFCN may develop insulin resistance and their consequent failure represents one of the earliest event in AD. We found that a condition reminiscent of insulin resistance occurs in the medial septum of 3 months old 3×Tg-AD mice, reported to develop typical AD histopathology and cognitive deficits in adulthood. Further, we obtained insulin resistant BFCN by culturing them with high insulin concentrations. By means of these paradigms, we observed that nerve growth factor (NGF) reduces insulin resistance in vitro and in vivo. NGF activates the insulin receptor substrate 1 (IRS1) and rescues c-Fos expression and glucose metabolism. This effect involves binding of activated IRS1 to the NGF receptor TrkA, and is lost in presence of the specific IRS inhibitor NT157. Overall, our findings indicate that, in a well-established animal model of AD, the medial septum develops insulin resistance several months before it is detectable in the neocortex and hippocampus. Remarkably, NGF counteracts molecular alterations downstream of insulin-resistant receptor and its nasal administration restores insulin signaling in 3×Tg-AD mice by TrkA/IRS1 activation. The cross-talk between NGF and insulin pathways downstream the insulin receptor suggests novel potential therapeutic targets to slow cognitive decline in AD and diabetes-related brain insulin resistance.
Collapse
Affiliation(s)
- Valentina Sposato
- European Brain Research Institute (EBRI) Rita Levi-Montalcini Foundation, Viale Regina Elena 295, Rome, Italy
| | - Nadia Canu
- National Research Council (CNR), Institute of Cell Biology and Neurobiology, Via del Fosso di Fiorano 64, Rome, Italy
- Department of System Medicine, Section of Physiology, University of Rome “TorVergata”, Rome, Italy
| | - Elena Fico
- National Research Council (CNR), Institute of Cell Biology and Neurobiology, Via del Fosso di Fiorano 64, Rome, Italy
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Salvatore Fusco
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giulia Bolasco
- European Molecular Biology Laboratory (EMBL), Monterotondo Outstation, Rome, Italy
| | - Maria Teresa Ciotti
- European Brain Research Institute (EBRI) Rita Levi-Montalcini Foundation, Viale Regina Elena 295, Rome, Italy
| | - Matteo Spinelli
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Delio Mercanti
- National Research Council (CNR), Institute of Cell Biology and Neurobiology, Via del Fosso di Fiorano 64, Rome, Italy
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy
| | - Viviana Triaca
- National Research Council (CNR), Institute of Cell Biology and Neurobiology, Via del Fosso di Fiorano 64, Rome, Italy
| | - Pietro Calissano
- European Brain Research Institute (EBRI) Rita Levi-Montalcini Foundation, Viale Regina Elena 295, Rome, Italy
- National Research Council (CNR), Institute of Cell Biology and Neurobiology, Via del Fosso di Fiorano 64, Rome, Italy
| |
Collapse
|
30
|
Ahmed QU, Sarian MN, Mat So'ad SZ, Latip J, Arief Ichwan SJ, Hussein NN, Taher M, Alhassan AM, Hamidon H, Fakurazi S. Methylation and Acetylation Enhanced the Antidiabetic Activity of Some Selected Flavonoids: In Vitro, Molecular Modelling and Structure Activity Relationship-Based Study. Biomolecules 2018; 8:E149. [PMID: 30445784 PMCID: PMC6316872 DOI: 10.3390/biom8040149] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 11/09/2018] [Indexed: 12/14/2022] Open
Abstract
Flavonoids have been reported to exert antihyperglycemic effects and have potential to enhance the current therapy options against type 2 diabetes mellitus. However, the structure activity relationships (SAR) studies of flavonoids against this disease have not been thoroughly comprehended. Hence, in the present study, 14 structurally related flavonoids viz. wogonin, techtochrysin, norwogonin, isoscutellarein, hypolaetin, kaempferol, quercetin, methyl ether of wogonin, acetate of wogonin, acetate of norwogonin, 8-hydroxy-7-methoxyflavone, chrysin, (+)-catechin and (-)-epicatechin were taken into account for in vitro antidiabetic evaluation. Cell viability of RIN-5F pancreatic cells and 3T3-L1 pre-adipocyte cells was initially tested, then an insulin secretion assay of RIN-5F as well as adipogenesis and glucose uptake measurements of adipocyte were investigated. Subsequently, protein expressions study through adipokines measurement (leptin, adiponectin, TNF-α, RBP-4) via enzyme-linked immunosorbent assay (ELISA) kit, Western blotting analysis against GLUT4 and C/EBP-α as well as molecular docking against GLUT1 were analyzed. The results from cell culture antidiabetic assays (insulin secretion, adipogenesis, and glucose uptake), protein expressions and molecular docking pointed that the methoxy group at position C-8 is responsible for antidiabetic property of selected flavonoids via glucose uptake mechanism indicated by up regulation of GLUT4 and C/EBP-α expressions. The mechanism could be enhanced by the addition of an acetate group at C-5 and C-7 of the flavone skeleton.
Collapse
Affiliation(s)
- Qamar Uddin Ahmed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, International Islamic University Malaysia, Kuantan 25200, Pahang DM, Malaysia.
| | - Murni Nazira Sarian
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, International Islamic University Malaysia, Kuantan 25200, Pahang DM, Malaysia.
- Laboratory of Vaccines and Immunotherapeutics, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Siti Zaiton Mat So'ad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, International Islamic University Malaysia, Kuantan 25200, Pahang DM, Malaysia.
| | - Jalifah Latip
- School of Chemical Sciences and Food Technology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia.
| | | | - Nurlaili Najmie Hussein
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, International Islamic University Malaysia, Kuantan 25200, Pahang DM, Malaysia.
| | - Muhammad Taher
- Department of Pharmaceutical Technology, Faculty of Pharmacy, International Islamic University Malaysia, Kuantan 25200, Pahang DM, Malaysia.
| | - Alhassan Muhammad Alhassan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, International Islamic University Malaysia, Kuantan 25200, Pahang DM, Malaysia.
| | - Hanisuhana Hamidon
- Department of Pharmaceutical Technology, Faculty of Pharmacy, International Islamic University Malaysia, Kuantan 25200, Pahang DM, Malaysia.
| | - Sharida Fakurazi
- Laboratory of Vaccines and Immunotherapeutics, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| |
Collapse
|
31
|
Matsuda H. Zebrafish as a model for studying functional pancreatic β cells development and regeneration. Dev Growth Differ 2018; 60:393-399. [DOI: 10.1111/dgd.12565] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/06/2018] [Accepted: 07/06/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Hiroki Matsuda
- Department of Developmental Biology and Neurosciences; Graduate School of Life Sciences; Tohoku University; Sendai Japan
- Department of Biomedical Sciences; College of Life Sciences; Ritsumeikan University; Kusatsu Japan
| |
Collapse
|
32
|
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: 6] [Impact Index Per Article: 0.9] [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
|
33
|
Abstract
Intrauterine growth restriction (IUGR) has been defined in several ways, but in general describes a condition in which the fetus exhibits poor growth in utero. This complication of pregnancy poses a significant public health burden as well as increased morbidity and mortality for the offspring. In human IUGR, alteration in fetal glucose and insulin homeostasis occurs in an effort to conserve energy and survive at the expense of fetal growth in an environment of inadequate nutrient provision. Several animal models of IUGR have been utilized to study the effects of IUGR on fetal glucose handling, as well as the postnatal reprogramming of energy metabolite handling, which may be unmasked in adulthood as a maladaptive propensity for cardiometabolic disease. This developmental programming may be mediated in part by epigenetic modification of essential regulators of glucose homeostasis. Several pharmacological therapies and nonpharmacological lifestyle modifications have shown early promise in mitigating the risk for or severity of adult metabolic phenotypes but still require further study of unanticipated and/or untoward side effects.
Collapse
Affiliation(s)
- Sherin U Devaskar
- Department of Pediatrics, Division of Neonatology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Alison Chu
- Department of Pediatrics, Division of Neonatology, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
34
|
Maddison LA, Chen W. Modeling Pancreatic Endocrine Cell Adaptation and Diabetes in the Zebrafish. Front Endocrinol (Lausanne) 2017; 8:9. [PMID: 28184214 PMCID: PMC5266698 DOI: 10.3389/fendo.2017.00009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 01/11/2017] [Indexed: 12/13/2022] Open
Abstract
Glucose homeostasis is an important element of energy balance and is conserved in organisms from fruit fly to mammals. Central to the control of circulating glucose levels in vertebrates are the endocrine cells of the pancreas, particularly the insulin-producing β-cells and the glucagon producing α-cells. A feature of α- and β-cells is their plasticity, an ability to adapt, in function and number as a response to physiological and pathophysiological conditions of increased hormone demand. The molecular mechanisms underlying these adaptive responses that maintain glucose homeostasis are incompletely defined. The zebrafish is an attractive model due to the low cost, high fecundity, and amenability to genetic and compound screens, and mechanisms governing the development of the pancreatic endocrine cells are conserved between zebrafish and mammals. Post development, both β- and α-cells of zebrafish display plasticity as in mammals. Here, we summarize the studies of pancreatic endocrine cell adaptation in zebrafish. We further explore the utility of the zebrafish as a model for diabetes, a relevant topic considering the increase in diabetes in the human population.
Collapse
Affiliation(s)
- Lisette A. Maddison
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Wenbiao Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
35
|
Kersten S, Arjona FJ. Ion transport in the zebrafish kidney from a human disease angle: possibilities, considerations, and future perspectives. Am J Physiol Renal Physiol 2017; 312:F172-F189. [DOI: 10.1152/ajprenal.00425.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 11/11/2016] [Accepted: 11/14/2016] [Indexed: 12/31/2022] Open
Abstract
Unique experimental advantages, such as its embryonic/larval transparency, high-throughput nature, and ease of genetic modification, underpin the rapid emergence of the zebrafish ( Danio rerio) as a preeminent model in biomedical research. Particularly in the field of nephrology, the zebrafish provides a promising model for studying the physiological implications of human solute transport processes along consecutive nephron segments. However, although the zebrafish might be considered a valuable model for numerous renal ion transport diseases and functional studies of many channels and transporters, not all human renal electrolyte transport mechanisms and human diseases can be modeled in the zebrafish. With this review, we explore the ontogeny of zebrafish renal ion transport, its nephron structure and function, and thereby demonstrate the clinical translational value of this model. By critical assessment of genomic and amino acid conservation of human proteins involved in renal ion handling (channels, transporters, and claudins), kidney and nephron segment conservation, and renal electrolyte transport physiology in the zebrafish, we provide researchers and nephrologists with an indication of the possibilities and considerations of the zebrafish as a model for human renal ion transport. Combined with advanced techniques envisioned for the future, implementation of the zebrafish might expand beyond unraveling pathophysiological mechanisms that underlie distinct genetic or environmentally, i.e., pharmacological and lifestyle, induced renal transport deficits. Specifically, the ease of drug administration and the exploitation of improved genetic approaches might argue for the adoption of the zebrafish as a model for preclinical personalized medicine for distinct renal diseases and renal electrolyte transport proteins.
Collapse
Affiliation(s)
- Simone Kersten
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands; and
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Francisco J. Arjona
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands; and
| |
Collapse
|
36
|
Jung JH, Iwabuchi K, Yang Z, Loeken MR. Embryonic Stem Cell Proliferation Stimulated By Altered Anabolic Metabolism From Glucose Transporter 2-Transported Glucosamine. Sci Rep 2016; 6:28452. [PMID: 27311888 PMCID: PMC4911601 DOI: 10.1038/srep28452] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 06/02/2016] [Indexed: 01/19/2023] Open
Abstract
The hexose transporter, GLUT2 (SLC2A2), which is expressed by mouse embryos, is important for survival before embryonic day 10.5, but its function in embryos is unknown. GLUT2 can transport the amino sugar glucosamine (GlcN), which could increase substrate for the hexosamine biosynthetic pathway (HBSP) that produces UDP-N-acetylglucosamine for O-linked N-acetylglucosamine modification (O-GlcNAcylation) of proteins. To understand this, we employed a novel murine embryonic stem cell (ESC) line that, like mouse embryos, expresses functional GLUT2 transporters. GlcN stimulated ESC proliferation in a GLUT2-dependent fashion but did not regulate pluripotency. Stimulation of proliferation was not due to increased O-GlcNAcylation. Instead, GlcN decreased dependence of the HBSP on fructose-6-PO4 and glutamine. Consequently, glycolytic- and glutamine-derived intermediates that are needed for anabolic metabolism were increased. Thus, maternally obtained GlcN may increase substrates for biomass accumulation by embryos, as exogenous GlcN does for GLUT2-expressing ESC, and may explain the need for GLUT2 expression by embryos.
Collapse
Affiliation(s)
- Jin Hyuk Jung
- Section on Islet Cell and Regenerative Biology Joslin Diabetes Center, Boston, MA 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Kumiko Iwabuchi
- Section on Islet Cell and Regenerative Biology Joslin Diabetes Center, Boston, MA 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Zhihong Yang
- Section on Islet Cell and Regenerative Biology Joslin Diabetes Center, Boston, MA 02215, USA.,Section on Vascular Cell Biology Harvard Medical School, Boston, MA 02115, USA
| | - Mary R Loeken
- Section on Islet Cell and Regenerative Biology Joslin Diabetes Center, Boston, MA 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
37
|
Glucose Transporters at the Blood-Brain Barrier: Function, Regulation and Gateways for Drug Delivery. Mol Neurobiol 2016; 54:1046-1077. [PMID: 26801191 DOI: 10.1007/s12035-015-9672-6] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/17/2015] [Indexed: 12/31/2022]
Abstract
Glucose transporters (GLUTs) at the blood-brain barrier maintain the continuous high glucose and energy demands of the brain. They also act as therapeutic targets and provide routes of entry for drug delivery to the brain and central nervous system for treatment of neurological and neurovascular conditions and brain tumours. This article first describes the distribution, function and regulation of glucose transporters at the blood-brain barrier, the major ones being the sodium-independent facilitative transporters GLUT1 and GLUT3. Other GLUTs and sodium-dependent transporters (SGLTs) have also been identified at lower levels and under various physiological conditions. It then considers the effects on glucose transporter expression and distribution of hypoglycemia and hyperglycemia associated with diabetes and oxygen/glucose deprivation associated with cerebral ischemia. A reduction in glucose transporters at the blood-brain barrier that occurs before the onset of the main pathophysiological changes and symptoms of Alzheimer's disease is a potential causative effect in the vascular hypothesis of the disease. Mutations in glucose transporters, notably those identified in GLUT1 deficiency syndrome, and some recreational drug compounds also alter the expression and/or activity of glucose transporters at the blood-brain barrier. Approaches for drug delivery across the blood-brain barrier include the pro-drug strategy whereby drug molecules are conjugated to glucose transporter substrates or encapsulated in nano-enabled delivery systems (e.g. liposomes, micelles, nanoparticles) that are functionalised to target glucose transporters. Finally, the continuous development of blood-brain barrier in vitro models is important for studying glucose transporter function, effects of disease conditions and interactions with drugs and xenobiotics.
Collapse
|
38
|
Ruggiero JE, Northrup H, Au KS. Association of facilitated glucose transporter 2 gene variants with the myelomeningocele phenotype. ACTA ACUST UNITED AC 2015; 103:479-87. [PMID: 25776730 DOI: 10.1002/bdra.23358] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Neural tube defects (NTDs) remain the second most common cause of congenital malformations. Myelomeningocele (MM), the most common NTD compatible with survival, results from genetic and environmental factors. Epidemiologic studies and murine models support the hypotheses that obesity, diabetes and hyperglycemia confer increased risk of NTDs. Presence of wild-type facilitated glucose transporter, Glut2, in mouse embryos has been shown to increase risk for NTDs in hyperglycemic pregnancy. METHODS The GLUT2 gene of 96 MM patients was amplified, sequenced and compared with the reference sequence (NM_000340). Variants previously unreported in the single nucleotide polymorphisms (SNP) database were considered novel. Allele frequencies of reported SNPs were compared with reference populations using Fisher's exact test. RESULTS Analysis revealed three novel variants: a substitution in the core promoter region (c.-331c>t), a substitution (c.-182g>a) in the 5'-untranslated region, and a single base pair deletion (c.1441delT) in the coding sequences. Polymorphic alleles for 10 SNPs were also identified. Seven SNPs are significantly associated with MM in the Mexican American patients tested (p < 0.05) and two of the seven remained significant after Bonferroni correction. CONCLUSION We identified three novel variants and seven SNPs associated with MM. The novel variants in the core promoter and in the 5'-untranslated region could affect GLUT2 mRNA transcription and stability and translation efficiency. The c.1441delT variant is predicted to alter the reading frame and prematurely terminate translation of the GLUT2 protein at the C-terminus, affecting GLUT2 protein function. Presence of GLUT2 variants may disrupt GLUT2 activity and influence MM susceptibility.
Collapse
Affiliation(s)
- Jaclyn E Ruggiero
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of Texas Medical School at Houston, Houston, Texas
| | - Hope Northrup
- Division of Medical Genetics, Department of Pediatrics, The University of Texas Medical School at Houston, Houston, Texas.,Shriners Hospitals for Children, Houston, Texas
| | - Kit Sing Au
- Division of Medical Genetics, Department of Pediatrics, The University of Texas Medical School at Houston, Houston, Texas
| |
Collapse
|