1
|
Yan G, Wang Y, Yu L, Bo J, Fang H, Chen W, Zhang Y, Chen H, Hong Z. Depolymerization of oyster glycosaminoglycans for the enhancement of α-glucosidase inhibition and its application in hypoglycemic studies. Food Res Int 2025; 205:116008. [PMID: 40032484 DOI: 10.1016/j.foodres.2025.116008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 03/05/2025]
Abstract
Glycosaminoglycans are a class of naturally occurring macromolecular heteropolysaccharides with α-glucosidase inhibitory effects, which play an essential role in regulating postprandial hyperglycemia. However, the significant molecular weight hinders their inhibitory activity. In this study, oyster glycosaminoglycans were used for purification and depolymerization studies to obtain oyster glycosaminoglycan oligosaccharides (OGAG-Oli) and to study their inhibitory activity against α-glucosidase in vitro and hypoglycemic effect on hyperglycemic zebrafish in vivo. The results showed that the OGAG-Oli with optimal α-glucosidase inhibition obtained by the β-elimination mechanism of chondroitinase ABC had a molecular weight of about 2400 Da with five disaccharide repeating units, and their inhibitory effect on α-glucosidase was significantly enhanced with an IC50 value of 0.12 mg/mL, which was related to the strong binding capacity with α-glucosidase as evidenced by molecular docking. FT-IR and NMR results showed that the conformation of OGAG-Oli was predominantly in the α-configuration, which indicated that its inhibitory effect on α-glucosidase mainly depended on the α-glucosidic bond therein. The enzymatic depolymerization process of glycosaminoglycans was optimized, and the process conditions were determined as phosphate buffer solution pH 8.0, reaction time 3 h, reaction temperature 30 °C, chondroitin enzyme ABC activity 0.5 mU. The hypoglycemic ability of OGAG-Oli was evaluated in diabetic zebrafish, and the results showed that OGAG-Oli at a high-dose (0.12 mg/g, bw/d) could extremely significantly reduce fasting blood glucose in zebrafish, with an effect close to that of acarbose (p > 0.05), and exceptionally significantly (p < 0.01) reduced the elevation of TC, TG, and LDL-C caused by prolonged hyperglycemia, and extremely significantly (p < 0.01) increased the HDL-C level. This study showed that the structurally modified oyster glycosaminoglycans had intense α-glucosidase inhibitory activity, and exhibited biological activity of reducing glucose uptake and consequently improving body weight and lipid metabolism in diabetic zebrafish. As a bioactive ingredient of natural origin, this study provides a theoretical basis for the development and utilization of glycosaminoglycans with hypoglycemic effects in foods and drugs.
Collapse
Affiliation(s)
- Guangyu Yan
- Xiamen Ocean Vocational College, Xiamen 361022, China
| | - Yaru Wang
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, China; College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350001, China
| | - Lei Yu
- Xiamen Ocean Vocational College, Xiamen 361022, China
| | - Jun Bo
- Key Laboratory of Marine Ecological Conservation and Restoration, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, China
| | - Hua Fang
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, China; Xiamen Ocean Vocational College, Xiamen 361022, China
| | - Weizhu Chen
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, China; Xiamen Ocean Vocational College, Xiamen 361022, China
| | - Yiping Zhang
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, China; Xiamen Ocean Vocational College, Xiamen 361022, China
| | - Hui Chen
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, China; Xiamen Ocean Vocational College, Xiamen 361022, China.
| | - Zhuan Hong
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, China; Xiamen Ocean Vocational College, Xiamen 361022, China.
| |
Collapse
|
2
|
Mi J, Ren L, Andersson O. Leveraging zebrafish to investigate pancreatic development, regeneration, and diabetes. Trends Mol Med 2024; 30:932-949. [PMID: 38825440 DOI: 10.1016/j.molmed.2024.05.002] [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: 01/31/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 06/04/2024]
Abstract
The zebrafish has become an outstanding model for studying organ development and tissue regeneration, which is prominently leveraged for studies of pancreatic development, insulin-producing β-cells, and diabetes. Although studied for more than two decades, many aspects remain elusive and it has only recently been possible to investigate these due to technical advances in transcriptomics, chemical-genetics, genome editing, drug screening, and in vivo imaging. Here, we review recent findings on zebrafish pancreas development, β-cell regeneration, and how zebrafish can be used to provide novel insights into gene functions, disease mechanisms, and therapeutic targets in diabetes, inspiring further use of zebrafish for the development of novel therapies for diabetes.
Collapse
Affiliation(s)
- Jiarui Mi
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden; Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, China.
| | - Lipeng Ren
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden; Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden
| | - Olov Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden; Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden.
| |
Collapse
|
3
|
Tucker SK, Ghosal R, Swartz ME, Zhang S, Eberhart JK. Zebrafish raptor mutation inhibits the activity of mTORC1, inducing craniofacial defects due to autophagy-induced neural crest cell death. Development 2024; 151:dev202216. [PMID: 38512806 PMCID: PMC11006402 DOI: 10.1242/dev.202216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 01/26/2024] [Indexed: 03/23/2024]
Abstract
The mechanistic target of rapamycin (mTOR) coordinates metabolism and cell growth with environmental inputs. mTOR forms two functional complexes: mTORC1 and mTORC2. Proper development requires both complexes but mTORC1 has unique roles in numerous cellular processes, including cell growth, survival and autophagy. Here, we investigate the function of mTORC1 in craniofacial development. We created a zebrafish raptor mutant via CRISPR/Cas9, to specifically disrupt mTORC1. The entire craniofacial skeleton and eyes were reduced in size in mutants; however, overall body length and developmental timing were not affected. The craniofacial phenotype associates with decreased chondrocyte size and increased neural crest cell death. We found that autophagy is elevated in raptor mutants. Chemical inhibition of autophagy reduced cell death and improved craniofacial phenotypes in raptor mutants. Genetic inhibition of autophagy, via mutation of the autophagy gene atg7, improved facial phenotypes in atg7;raptor double mutants, relative to raptor single mutants. We conclude that finely regulated levels of autophagy, via mTORC1, are crucial for craniofacial development.
Collapse
Affiliation(s)
- Scott K. Tucker
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research and Institute for Neuroscience, University of Texas, Austin, TX 78712, USA
| | - Ritika Ghosal
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research and Institute for Neuroscience, University of Texas, Austin, TX 78712, USA
| | - Mary E. Swartz
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research and Institute for Neuroscience, University of Texas, Austin, TX 78712, USA
| | - Stephanie Zhang
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research and Institute for Neuroscience, University of Texas, Austin, TX 78712, USA
| | - Johann K. Eberhart
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research and Institute for Neuroscience, University of Texas, Austin, TX 78712, USA
| |
Collapse
|
4
|
Fish EW, Tucker SK, Peterson RL, Eberhart JK, Parnell SE. Loss of tumor protein 53 protects against alcohol-induced facial malformations in mice and zebrafish. Alcohol Clin Exp Res 2021; 45:1965-1979. [PMID: 34581462 DOI: 10.1111/acer.14688] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Alcohol exposure during the gastrulation stage of development causes the craniofacial and brain malformations that define fetal alcohol syndrome. These malformations, such as a deficient philtrum, are exemplified by a loss of midline tissue and correspond, at least in part, to regionally selective cell death in the embryo. The tumor suppressor protein Tp53 is an important mechanism for cell death, but the role of Tp53 in the consequences of alcohol exposure during the gastrulation stage has yet to be examined. The current studies used mice and zebrafish to test whether genetic loss of Tp53 is a conserved mechanism to protect against the effects of early developmental stage alcohol exposure. METHODS Female mice, heterozygous for a mutation in the Tp53 gene, were mated with Tp53 heterozygous males, and the resulting embryos were exposed during gastrulation on gestational day 7 (GD 7) to alcohol (two maternal injections of 2.9 g/kg, i.p., 4 h apart) or a vehicle control. Zebrafish mutants or heterozygotes for the tp53zdf1 M214K mutation and their wild-type controls were exposed to alcohol (1.5% or 2%) beginning 6 h postfertilization (hpf), the onset of gastrulation. RESULTS Examination of GD 17 mice revealed that eye defects were the most common phenotype among alcohol-exposed fetuses, occurring in nearly 75% of the alcohol-exposed wild-type fetuses. Tp53 gene deletion reduced the incidence of eye defects in both the heterozygous and mutant fetuses (to about 35% and 20% of fetuses, respectively) and completely protected against alcohol-induced facial malformations. Zebrafish (4 days postfertilization) also demonstrated alcohol-induced reductions of eye size and trabeculae length that were less common and less severe in tp53 mutants, indicating a protective effect of tp53 deletion. CONCLUSIONS These results identify an evolutionarily conserved role of Tp53 as a pathogenic mechanism for alcohol-induced teratogenesis.
Collapse
Affiliation(s)
- Eric W Fish
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Scott K Tucker
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research and Institute for Neuroscience, University of Texas, Austin, Texas, USA
| | - Rachel L Peterson
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Johann K Eberhart
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research and Institute for Neuroscience, University of Texas, Austin, Texas, USA
| | - Scott E Parnell
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina, USA.,Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA.,Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
5
|
de Boer P, Giepmans BN. State-of-the-art microscopy to understand islets of Langerhans: what to expect next? Immunol Cell Biol 2021; 99:509-520. [PMID: 33667022 PMCID: PMC8252556 DOI: 10.1111/imcb.12450] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/12/2022]
Abstract
The discovery of Langerhans and microscopic description of islets in the pancreas were crucial steps in the discovery of insulin. Over the past 150 years, many discoveries in islet biology and type 1 diabetes have been made using powerful microscopic techniques. In the past decade, combination of new probes, animal and tissue models, application of new biosensors and automation of light and electron microscopic methods and other (sub)cellular imaging modalities have proven their potential in understanding the beta cell under (patho)physiological conditions. The imaging evolution, from fluorescent jellyfish to real‐time intravital functional imaging, the revolution in automation and data handling and the increased resolving power of analytical imaging techniques are now converging. Here, we review innovative approaches that address islet biology from new angles by studying cells and molecules at high spatiotemporal resolution and in live models. Broad implementation of these cellular imaging techniques will shed new light on cause/consequence of (mal)function in islets of Langerhans in the years to come.
Collapse
Affiliation(s)
- Pascal de Boer
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ben Ng Giepmans
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
6
|
Yang L, Webb SE, Jin N, Lee HM, Chan TF, Xu G, Chan JC, Miller AL, Ma RC. Investigating the role of dachshund b in the development of the pancreatic islet in zebrafish. J Diabetes Investig 2021; 12:710-727. [PMID: 33449448 PMCID: PMC8089008 DOI: 10.1111/jdi.13503] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/28/2020] [Accepted: 01/01/2021] [Indexed: 12/29/2022] Open
Abstract
Aims/Introduction β‐Cell dysfunction is a hallmark of type 2 diabetes. In a previous pilot study, we identified an association between genetic variants within the human DACH1 gene and young‐onset type 2 diabetes. Here, we characterized the function of dachb, the only dach homologue to be expressed in the pancreas, in developing zebrafish embryos. Materials and Methods We injected one‐cell stage embryos with a dachb‐morpholino (MO) or with the dachb‐MO and dachb messenger ribonucleic acid, and determined the effect on the development of the pancreatic islet. We also carried out quantitative polymerase chain reaction and ribonucleic acid sequencing on the dachb‐MO group to determine the effect of dachb knockdown on gene expression. Results MO‐mediated dachb knockdown resulted in impaired islet cell development, with a significant decrease in both the β‐cell and islet cell numbers. This islet developmental defect was rescued when embryos were co‐injected with dachb‐MO and dachb messenger ribonucleic acid. Knockdown of dachb was associated with a significant downregulation of the β‐cell specific marker gene, insa, and the somatostatin cell marker, sst2, as well as regulators of pancreas development, ptf1a, neuroD, pax6a and nkx6.1, and the cell cycle gene, insm1a. Furthermore, ribonucleic sequencing analysis showed an upregulation of genes enriched in the forkhead box O and mitogen‐activated protein kinase signaling pathways in the dachb‐MO group, when compared with the control groups. Conclusions Together, our results suggest the possible role of dachb in islet development in zebrafish.
Collapse
Affiliation(s)
- Lingling Yang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong
| | - Sarah E Webb
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Nana Jin
- School of Life Sciences and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Heung Man Lee
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong
| | - Ting Fung Chan
- School of Life Sciences and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Gang Xu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.,Teaching and Research Division, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Juliana Cn Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.,Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Andrew L Miller
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Ronald Cw Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.,Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
7
|
Han B, Zhang Y, Bi X, Zhou Y, Krueger CJ, Hu X, Zhu Z, Tong X, Zhang B. Bi-FoRe: an efficient bidirectional knockin strategy to generate pairwise conditional alleles with fluorescent indicators. Protein Cell 2021; 12:39-56. [PMID: 32681448 PMCID: PMC7815861 DOI: 10.1007/s13238-020-00747-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/22/2020] [Indexed: 12/11/2022] Open
Abstract
Gene expression labeling and conditional manipulation of gene function are important for elaborate dissection of gene function. However, contemporary generation of pairwise dual-function knockin alleles to achieve both conditional and geno-tagging effects with a single donor has not been reported. Here we first developed a strategy based on a flipping donor named FoRe to generate conditional knockout alleles coupled with fluorescent allele-labeling through NHEJ-mediated unidirectional targeted insertion in zebrafish facilitated by the CRISPR/Cas system. We demonstrated the feasibility of this strategy at sox10 and isl1 loci, and successfully achieved Cre-induced conditional knockout of target gene function and simultaneous switch of the fluorescent reporter, allowing generation of genetic mosaics for lineage tracing. We then improved the donor design enabling efficient one-step bidirectional knockin to generate paired positive and negative conditional alleles, both tagged with two different fluorescent reporters. By introducing Cre recombinase, these alleles could be used to achieve both conditional knockout and conditional gene restoration in parallel; furthermore, differential fluorescent labeling of the positive and negative alleles enables simple, early and efficient real-time discrimination of individual live embryos bearing different genotypes prior to the emergence of morphologically visible phenotypes. We named our improved donor as Bi-FoRe and demonstrated its feasibility at the sox10 locus. Furthermore, we eliminated the undesirable bacterial backbone in the donor using minicircle DNA technology. Our system could easily be expanded for other applications or to other organisms, and coupling fluorescent labeling of gene expression and conditional manipulation of gene function will provide unique opportunities to fully reveal the power of emerging single-cell sequencing technologies.
Collapse
Affiliation(s)
- Bingzhou Han
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, Peking University Genome Editing Research Center, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Yage Zhang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, Peking University Genome Editing Research Center, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Xuetong Bi
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, Peking University Genome Editing Research Center, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Yang Zhou
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Christopher J Krueger
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 100871, China
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory, Atlanta, GA, 33032, USA
| | - Xinli Hu
- Institute of Molecular Medicine, Peking University, Beijing, 100871, China
| | - Zuoyan Zhu
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, Peking University Genome Editing Research Center, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Xiangjun Tong
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, Peking University Genome Editing Research Center, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Bo Zhang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, Peking University Genome Editing Research Center, College of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
8
|
β-Cell specific transcription factors in the context of diabetes mellitus and β-cell regeneration. Mech Dev 2020; 163:103634. [PMID: 32711047 DOI: 10.1016/j.mod.2020.103634] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 07/15/2020] [Accepted: 07/15/2020] [Indexed: 02/06/2023]
Abstract
All pancreatic cell populations arise from the standard gut endoderm layer in developing embryos, requiring a regulatory gene network to originate and maintain endocrine lineages and endocrine function. The pancreatic organogenesis is regulated by the temporal expression of transcription factors and plays a diverse role in the specification, development, differentiation, maturation, and functional maintenance. Altered expression and activity of these transcription factors are often associated with diabetes mellitus. Recent advancements in the stem cells and invitro derived islets to treat diabetes mellitus has attracted a great deal of interest in the understanding of factors regulating the development, differentiation, and functions of islets including transcription factors. This review discusses the myriad of transcription factors regulating the development of the pancreas, differentiation of β-islets, and how these factors regulated in normal and disease states. Exploring these factors in such critical context and exogenous or endogenous expression of development and differentiation-specific transcription factors with improved epigenetic plasticity/signaling axis in diabetic milieu would useful for the development of β-cells from other cell sources.
Collapse
|
9
|
Lu CJ, Fan XY, Guo YF, Cheng ZC, Dong J, Chen JZ, Li LY, Wang MW, Wu ZK, Wang F, Tong XJ, Luo LF, Tang FC, Zhu ZY, Zhang B. Single-cell analyses identify distinct and intermediate states of zebrafish pancreatic islet development. J Mol Cell Biol 2020; 11:435-447. [PMID: 30407522 PMCID: PMC6604604 DOI: 10.1093/jmcb/mjy064] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 10/31/2018] [Accepted: 11/08/2018] [Indexed: 12/13/2022] Open
Abstract
Pancreatic endocrine islets are vital for glucose homeostasis. However, the islet developmental trajectory and its regulatory network are not well understood. To define the features of these specification and differentiation processes, we isolated individual islet cells from TgBAC(neurod1:EGFP) transgenic zebrafish and analyzed islet developmental dynamics across four different embryonic stages using a single-cell RNA-seq strategy. We identified proliferative endocrine progenitors, which could be further categorized by different cell cycle phases with the G1/S subpopulation displaying a distinct differentiation potential. We identified endocrine precursors, a heterogeneous intermediate-state population consisting of lineage-primed alpha, beta and delta cells that were characterized by the expression of lineage-specific transcription factors and relatively low expression of terminally differentiation markers. The terminally differentiated alpha, beta, and delta cells displayed stage-dependent differentiation states, which were related to their functional maturation. Our data unveiled distinct states, events and molecular features during the islet developmental transition, and provided resources to comprehensively understand the lineage hierarchy of islet development at the single-cell level.
Collapse
Affiliation(s)
- Chong-Jian Lu
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Xiao-Ying Fan
- Beijing Advanced Innovation Center for Genomics (ICG), College of Life Sciences, Peking University, Beijing, China
| | - Yue-Feng Guo
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Zhen-Chao Cheng
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Ji Dong
- Beijing Advanced Innovation Center for Genomics (ICG), College of Life Sciences, Peking University, Beijing, China
| | - Jin-Zi Chen
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Chongqing, China
| | - Lian-Yan Li
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Mei-Wen Wang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Ze-Kai Wu
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Fei Wang
- National Center for Protein Sciences, Peking University, Beijing, China
| | - Xiang-Jun Tong
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Ling-Fei Luo
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Chongqing, China
| | - Fu-Chou Tang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China.,Beijing Advanced Innovation Center for Genomics (ICG), College of Life Sciences, Peking University, Beijing, China
| | - Zuo-Yan Zhu
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Bo Zhang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| |
Collapse
|
10
|
Li LC, Qiu WL, Zhang YW, Xu ZR, Xiao YN, Hou C, Lamaoqiezhong, Yu P, Cheng X, Xu CR. Single-cell transcriptomic analyses reveal distinct dorsal/ventral pancreatic programs. EMBO Rep 2018; 19:embr.201846148. [PMID: 30065074 DOI: 10.15252/embr.201846148] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 06/30/2018] [Accepted: 07/09/2018] [Indexed: 11/09/2022] Open
Abstract
The pancreas of vertebrates is separately derived from both the dorsal and ventral endodermal domains. However, the difference between these two programs has been unclear. Here, using a pancreatic determination gene, Pdx1, driven GFP transgenic mouse strain, we identified Pdx1-GFP highly expressing cells (Pdx1high) and Pdx1-GFP lowly expressing cells (Pdx1low) in both embryonic dorsal Pdx1-expressing region (DPR) and ventral Pdx1-expressing region (VPR). We analyzed the transcriptomes of single Pdx1low and Pdx1high cells from the DPR and VPR. In the VPR, Pdx1low cells have an intermediate progenitor identity and can generate hepatoblasts, extrahepatobiliary cells, and Pdx1high pancreatic progenitor cells. In the DPR, Pdx1high cells are directly specified as pancreatic progenitors, whereas Pdx1low cells are precocious endocrine cells. Therefore, our study defines distinct road maps for dorsal and ventral pancreatic progenitor specification. The findings provide guidance for optimization of current β-cell induction protocols by following the in vivo dorsal pancreatic specification program.
Collapse
Affiliation(s)
- Lin-Chen Li
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Wei-Lin Qiu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Beijing, China.,PKU-Tsinghua-NIBS Graduate Program, Peking University, Beijing, China
| | - Yu-Wei Zhang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Zi-Ran Xu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Beijing, China.,PKU-Tsinghua-NIBS Graduate Program, Peking University, Beijing, China
| | - Yi-Ni Xiao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Caiying Hou
- General Hospital of PLA Rocket Force, Beijing, China
| | - Lamaoqiezhong
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Peng Yu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Xin Cheng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Cheng-Ran Xu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Beijing, China
| |
Collapse
|
11
|
Sant KE, Jacobs HM, Borofski KA, Moss JB, Timme-Laragy AR. Embryonic exposures to perfluorooctanesulfonic acid (PFOS) disrupt pancreatic organogenesis in the zebrafish, Danio rerio. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2017; 220:807-817. [PMID: 27810111 PMCID: PMC5140685 DOI: 10.1016/j.envpol.2016.10.057] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/23/2016] [Accepted: 10/19/2016] [Indexed: 05/18/2023]
Abstract
Perfluorooctanesulfonic acid (PFOS) is a ubiquitous environmental contaminant, previously utilized as a non-stick application for consumer products and firefighting foam. It can cross the placenta, and has been repeatedly associated with increased risk for diabetes in epidemiological studies. Here, we sought to establish the hazard posed by embryonic PFOS exposures on the developing pancreas in a model vertebrate embryo, and develop criteria for an adverse outcome pathway (AOP) framework to study the developmental origins of metabolic dysfunction. Zebrafish (Danio rerio) embryos were exposed to 16, 32, or 64 μM PFOS beginning at the mid-blastula transition. We assessed embryo health, size, and islet morphology in Tg(insulin-GFP) embryos at 48, 96 and 168 hpf, and pancreas length in Tg(ptf1a-GFP) embryos at 96 and 168 hpf. QPCR was used to measure gene expression of endocrine and exocrine hormones, digestive peptides, and transcription factors to determine whether these could be used as a predictive measure in an AOP. Embryos exposed to PFOS showed anomalous islet morphology and decreased islet size and pancreas length in a U-shaped dose-response curve, which resemble congenital defects associated with increased risk for diabetes in humans. Expression of genes encoding islet hormones and exocrine digestive peptides followed a similar pattern, as did total larval growth. Our results demonstrate that embryonic PFOS exposures can disrupt pancreatic organogenesis in ways that mimic human congenital defects known to predispose individuals to diabetes; however, future study of the association between these defects and metabolic dysfunction are needed to establish an improved AOP framework.
Collapse
Affiliation(s)
- Karilyn E Sant
- Department of Environmental Health Sciences, University of Massachusetts Amherst, Amherst, MA 01003, United States
| | - Haydee M Jacobs
- Department of Environmental Health Sciences, University of Massachusetts Amherst, Amherst, MA 01003, United States
| | - Katrina A Borofski
- Department of Environmental Health Sciences, University of Massachusetts Amherst, Amherst, MA 01003, United States
| | - Jennifer B Moss
- Duke Molecular Physiology Institute, Endocrine Division, Duke University Medical Center, Durham, NC 27701, United States
| | - Alicia R Timme-Laragy
- Department of Environmental Health Sciences, University of Massachusetts Amherst, Amherst, MA 01003, United States.
| |
Collapse
|
12
|
Wang W, Shi Q, Guo T, Yang Z, Jia Z, Chen P, Zhou C. PDX1 and ISL1 differentially coordinate with epigenetic modifications to regulate insulin gene expression in varied glucose concentrations. Mol Cell Endocrinol 2016; 428:38-48. [PMID: 26994512 DOI: 10.1016/j.mce.2016.03.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 02/26/2016] [Accepted: 03/15/2016] [Indexed: 01/01/2023]
Abstract
The mechanism of insulin gene transcription control in response to glucose concentration is poorly defined. The islet-restricted transcription factors PDX1 and ISL1 interact with BETA2, activating insulin gene expression. However, their contribution and hierarchical organization in insulin expression control based on glucose concentration remain unknown. We investigated PDX1 and ISL1 regulation of insulin gene expression in pancreatic β cells cultured in normal (5 mM/L) and high (25 mM/L) glucose conditions. ISL1 interacted with BETA2 to maintain basic insulin gene transcriptional activity under normal glucose. The ISL1-recruited cofactors SET9 and JMJD3 facilitated insulin gene histone modifications under normal glucose. In high-glucose concentrations, PDX1 formed a complex with BETA2 to enhance insulin gene expression. PDX1 also recruited SET9 and JMJD3 to promote the activation of histone modulation on the insulin promoter. This is the first evidence transcription factors orchestrate epigenetic modifications to control insulin gene expression based on glucose concentration.
Collapse
Affiliation(s)
- Weiping Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education of China, Peking University, 38 Xue Yuan Road, Beijing 100191, China
| | - Qiong Shi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education of China, Peking University, 38 Xue Yuan Road, Beijing 100191, China
| | - Ting Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education of China, Peking University, 38 Xue Yuan Road, Beijing 100191, China
| | - Zhe Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education of China, Peking University, 38 Xue Yuan Road, Beijing 100191, China
| | - Zhuqing Jia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education of China, Peking University, 38 Xue Yuan Road, Beijing 100191, China
| | - Ping Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education of China, Peking University, 38 Xue Yuan Road, Beijing 100191, China
| | - Chunyan Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education of China, Peking University, 38 Xue Yuan Road, Beijing 100191, China.
| |
Collapse
|
13
|
The LIM-homeodomain transcription factor Islet2a promotes angioblast migration. Dev Biol 2016; 414:181-92. [PMID: 27126199 DOI: 10.1016/j.ydbio.2016.04.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 03/25/2016] [Accepted: 04/24/2016] [Indexed: 02/04/2023]
Abstract
Angioblasts of the developing vascular system require many signaling inputs to initiate their migration, proliferation and differentiation into endothelial cells. What is less studied is which intrinsic cell factors interpret these extrinsic signals. Here, we show the Lim homeodomain transcription factor islet2a (isl2a) is expressed in the lateral posterior mesoderm prior to angioblast migration. isl2a deficient angioblasts show disorganized migration to the midline to form axial vessels and fail to spread around the tailbud of the embryo. Isl2a morphants have fewer vein cells and decreased vein marker expression. We demonstrate that isl2a is required cell autonomously in angioblasts to promote their incorporation into the vein, and is permissive for vein identity. Knockout of isl2a results in decreased migration and proliferation of angioblasts during intersegmental artery growth. Since Notch signaling controls both artery-vein identity and tip-stalk cell formation, we explored the interaction of isl2a and Notch. We find that isl2a expression is negatively regulated by Notch activity, and that isl2a positively regulates flt4, a VEGF-C receptor repressed by Notch during angiogenesis. Thus Isl2a may act as an intermediate between Notch signaling and genetic programs controlling angioblast number and migration, placing it as a novel transcriptional regulator of early angiogenesis.
Collapse
|
14
|
|
15
|
Sjakste T, Paramonova N, Osina K, Dokane K, Sokolovska J, Sjakste N. Genetic variations in the PSMA3, PSMA6 and PSMC6 genes are associated with type 1 diabetes in Latvians and with expression level of number of UPS-related and T1DM-susceptible genes in HapMap individuals. Mol Genet Genomics 2015; 291:891-903. [PMID: 26661414 DOI: 10.1007/s00438-015-1153-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/28/2015] [Indexed: 01/04/2023]
Abstract
The ubiquitin-proteasome system (UPS), a key player of proteostasis network in the body, was implicated in type 1 diabetes mellitus (T1DM) pathogenesis. Polymorphisms in genes encoding proteasome subunits may potentially affect system efficiency. However, data in this field are still limited. To fulfil this gap, single nucleotide polymorphisms in the PSMB5 (rs11543947), PSMA6 (rs2277460, rs1048990), PSMC6 (rs2295826, rs2295827) and PSMA3 (rs2348071) genes were genotyped on susceptibility to T1DM in Latvians. The rs11543947 was found to be neutral and other loci manifested disease susceptibility, with rs1048990 and rs2348071 being the most significantly associated (P < 0.001; OR 2.042 [1.376-3.032] and OR 2.096 [1.415-3.107], respectively). Risk effect was associated with female phenotype for rs2277460 and family history for rs2277460, rs2295826 and rs2295827. Five-locus genotypes being at risk simultaneously at any two or more loci showed strong (P < 0.0001) T1DM association. The T1DM protective effects (P < 0.001) were shown for five-locus genotype and haplotype homozygous on common alleles and composed of common alleles, respectively. Using SNPexp data set, correlations have been revealed between the rs1048990, rs2295826, rs2295827 and rs2348071 T1DM risk genotypes and expression levels of 14 genes related to the UPS and 42 T1DM-susceptible genes encoding proteins involved in innate and adaptive immunity, antiviral response, insulin signalling, glucose-energy metabolism and other pathways implicated in T1DM pathogenesis. Genotype-phenotype and genotype-genotype clusterings support genotyping results. Our results provide evidence on new T1DM-susceptible loci in the PSMA3, PSMA6 and PSMC6 proteasome genes and give a new insight into the T1DM pathogenesis.
Collapse
Affiliation(s)
- Tatjana Sjakste
- Genomics and Bioinformatics, Institute of Biology of the University of Latvia, Salaspils, Latvia.
| | - Natalia Paramonova
- Genomics and Bioinformatics, Institute of Biology of the University of Latvia, Salaspils, Latvia
| | - Kristine Osina
- Genomics and Bioinformatics, Institute of Biology of the University of Latvia, Salaspils, Latvia
| | - Kristine Dokane
- Genomics and Bioinformatics, Institute of Biology of the University of Latvia, Salaspils, Latvia
| | | | | |
Collapse
|
16
|
Timme-Laragy AR, Sant KE, Rousseau ME, diIorio PJ. Deviant development of pancreatic beta cells from embryonic exposure to PCB-126 in zebrafish. Comp Biochem Physiol C Toxicol Pharmacol 2015; 178:25-32. [PMID: 26393762 DOI: 10.1016/j.cbpc.2015.08.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/17/2015] [Accepted: 08/28/2015] [Indexed: 10/23/2022]
Abstract
Exposures to co-planar PCBs and dioxins have been associated with diabetes in epidemiologic studies. Individuals may be predisposed to diseases such as diabetes as a result of exposure to environmental contaminants during early life, resulting in dysmorphic pancreatic islets or metabolically fragile β-cells. We tested the hypothesis that embryonic exposure to a model Ahr-ligand, PCB-126 would cause structural and/or functional alterations to the developing primary pancreatic islet in the zebrafish (Danio rerio). To assess β-cell development, transgenic zebrafish embryos (Tg(ins:GFP) and Tg(ins:mcherry) were exposed to nominal concentrations of 2 or 5nM PCB-126 or DMSO from 24-48h post fertilization (hpf), and imaged via time-lapse microscopy from 80-102hpf. We identified defects including hypomorphic islets, altered islet migration, islet fragmentation, and formation of ectopic β-cells. As we recently showed the transcription factor Nrf2a is protective in PCB-126 embryotoxicity, we then assessed the transcriptional function of the islets in wildtype and nrf2a(fh318/fh318) mutant embryos. We measured gene expression of preproinsulin a, somatostatin2, pdx1, ghrelin, and glucagon. Expression of preproinsulin a increased with PCB treatment in wildtype embryos, while expression of all measured pancreas genes was altered by the nrf2a mutant genotype, suggesting misregulation of the glucose homeostasis axis in those embryos, independent of PCB treatment. This study shows that embryonic exposure to PCB-126 can result in deviant development of the pancreatic islet and suggests that Nrf2a plays a role in regulating glucose homeostasis during development.
Collapse
Affiliation(s)
- Alicia R Timme-Laragy
- Department of Environmental Health Science, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA.
| | - Karilyn E Sant
- Department of Environmental Health Science, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA.
| | - Michelle E Rousseau
- Department of Environmental Health Science, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA.
| | - Philip J diIorio
- Department of Environmental Health Science, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| |
Collapse
|
17
|
Mastracci TL, Robertson MA, Mirmira RG, Anderson RM. Polyamine biosynthesis is critical for growth and differentiation of the pancreas. Sci Rep 2015; 5:13269. [PMID: 26299433 PMCID: PMC4547391 DOI: 10.1038/srep13269] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 06/30/2015] [Indexed: 02/03/2023] Open
Abstract
The pancreas, in most studied vertebrates, is a compound organ with both exocrine and endocrine functions. The exocrine compartment makes and secretes digestive enzymes, while the endocrine compartment, organized into islets of Langerhans, produces hormones that regulate blood glucose. High concentrations of polyamines, which are aliphatic amines, are reported in exocrine and endocrine cells, with insulin-producing β cells showing the highest concentrations. We utilized zebrafish as a model organism, together with pharmacological inhibition or genetic manipulation, to determine how polyamine biosynthesis functions in pancreatic organogenesis. We identified that inhibition of polyamine biosynthesis reduces exocrine pancreas and β cell mass, and that these reductions are at the level of differentiation. Moreover, we demonstrate that inhibition of ornithine decarboxylase (ODC), the rate-limiting enzyme in polyamine biosynthesis, phenocopies inhibition or knockdown of the enzyme deoxyhypusine synthase (DHS). These data identify that the pancreatic requirement for polyamine biosynthesis is largely mediated through a requirement for spermidine for the downstream posttranslational modification of eIF5A by its enzymatic activator DHS, which in turn impacts mRNA translation. Altogether, we have uncovered a role for polyamine biosynthesis in pancreatic organogenesis and identified that it may be possible to exploit polyamine biosynthesis to manipulate pancreatic cell differentiation.
Collapse
Affiliation(s)
- Teresa L Mastracci
- Department of Pediatrics, Indiana University School of Medicine, USA.,Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, USA
| | - Morgan A Robertson
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, USA
| | - Raghavendra G Mirmira
- Department of Pediatrics, Indiana University School of Medicine, USA.,Department of Physiology, Indiana University School of Medicine, USA.,Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, USA
| | - Ryan M Anderson
- Department of Pediatrics, Indiana University School of Medicine, USA.,Department of Physiology, Indiana University School of Medicine, USA.,Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, USA
| |
Collapse
|
18
|
Ye L, Robertson MA, Hesselson D, Stainier DYR, Anderson RM. Glucagon is essential for alpha cell transdifferentiation and beta cell neogenesis. Development 2015; 142:1407-17. [PMID: 25852199 DOI: 10.1242/dev.117911] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The interconversion of cell lineages via transdifferentiation is an adaptive mode of tissue regeneration and an appealing therapeutic target. However, its clinical exploitation is contingent upon the discovery of contextual regulators of cell fate acquisition and maintenance. In murine models of diabetes, glucagon-secreting alpha cells transdifferentiate into insulin-secreting beta cells following targeted beta cell depletion, regenerating the form and function of the pancreatic islet. However, the molecular triggers of this mode of regeneration are unknown. Here, using lineage-tracing assays in a transgenic zebrafish model of beta cell ablation, we demonstrate conserved plasticity of alpha cells during islet regeneration. In addition, we show that glucagon expression is upregulated after injury. Through gene knockdown and rescue approaches, we also find that peptides derived from the glucagon gene are necessary for alpha-to-beta cell fate switching. Importantly, whereas beta cell neogenesis was stimulated by glucose, alpha-to-beta cell conversion was not, suggesting that transdifferentiation is not mediated by glucagon/GLP-1 control of hepatic glucose production. Overall, this study supports the hypothesis that alpha cells are an endogenous reservoir of potential new beta cells. It further reveals that glucagon plays an important role in maintaining endocrine cell homeostasis through feedback mechanisms that govern cell fate stability.
Collapse
Affiliation(s)
- Lihua Ye
- Wells Center for Pediatric Research and Department of Cellular and Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Van Nuys Medical Sciences Building MS2053, Indianapolis, IN 46202, USA
| | - Morgan A Robertson
- Wells Center for Pediatric Research and Department of Cellular and Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Van Nuys Medical Sciences Building MS2053, Indianapolis, IN 46202, USA
| | - Daniel Hesselson
- Department of Biochemistry and Biophysics, 1550 4th Street Rock Hall Room 381, University of California San Francisco, San Francisco, CA 94143, USA
| | - Didier Y R Stainier
- Department of Biochemistry and Biophysics, 1550 4th Street Rock Hall Room 381, University of California San Francisco, San Francisco, CA 94143, USA
| | - Ryan M Anderson
- Wells Center for Pediatric Research and Department of Cellular and Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Van Nuys Medical Sciences Building MS2053, Indianapolis, IN 46202, USA
| |
Collapse
|
19
|
Nam YH, Hong BN, Rodriguez I, Ji MG, Kim K, Kim UJ, Kang TH. Synergistic Potentials of Coffee on Injured Pancreatic Islets and Insulin Action via KATP Channel Blocking in Zebrafish. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:5612-21. [PMID: 25993315 DOI: 10.1021/acs.jafc.5b00027] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Pancreatic islets (PIs) are damaged under diabetic conditions, resulting in decreased PI size. This study examined the regenerative effects of coffee and its components (caffeine, CFI; trigonelline, TRG; chlorogenic acid, CGA) on zebrafish larval PIs and β-cells damaged by administration of alloxan (AX). In addition, the influence of coffee and its active components on KATP channels was investigated using diazoxide (DZ) as a KATP channel activator. PI size and fluorescence intensity were significantly increased in the coffee-treated group relative to the no-treatment group (P < 0.0001). In addition, coffee exerted significant regenerative effects on pancreatic β-cells (p = 0.006). Treatment with TRG and CGA rescued PI damage, and the combination of TRG/CGA had a synergistic effect. In conclusion, the results indicate that coffee has beneficial effects on AX-damaged PIs and may also be useful as a blocker of pancreatic β-cell K(+) channels.
Collapse
Affiliation(s)
| | - Bin Na Hong
- §Department of Audiology, Nambu University, Gwangju 506-824, Republic of Korea
| | | | | | | | | | | |
Collapse
|
20
|
Dalgin G, Prince VE. Differential levels of Neurod establish zebrafish endocrine pancreas cell fates. Dev Biol 2015; 402:81-97. [PMID: 25797153 DOI: 10.1016/j.ydbio.2015.03.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Revised: 03/02/2015] [Accepted: 03/10/2015] [Indexed: 11/27/2022]
Abstract
During development a network of transcription factors functions to differentiate foregut cells into pancreatic endocrine cells. Differentiation of appropriate numbers of each hormone-expressing endocrine cell type is essential for the normal development of the pancreas and ultimately for effective maintenance of blood glucose levels. A fuller understanding of the details of endocrine cell differentiation may contribute to development of cell replacement therapies to treat diabetes. In this study, by using morpholino and gRNA/Cas9 mediated knockdown we establish that differential levels of the basic-helix loop helix (bHLH) transcription factor Neurod are required for the differentiation of distinct endocrine cell types in developing zebrafish. While Neurod plays a role in the differentiation of all endocrine cells, we find that differentiation of glucagon-expressing alpha cells is disrupted by a minor reduction in Neurod levels, whereas differentiation of insulin-expressing beta cells is less sensitive to Neurod depletion. The endocrine cells that arise during embryonic stages to produce the primary islet, and those that arise subsequently during larval stages from the intra-pancreatic duct (IPD) to ultimately contribute to the secondary islets, show similar dependence on differential Neurod levels. Intriguingly, Neurod-deficiency triggers premature formation of endocrine precursors from the IPD during early larval stages. However, the Neurod-deficient endocrine precursors fail to differentiate appropriately, and the larvae are unable to maintain normal glucose levels. In summary, differential levels of Neurod are required to generate endocrine pancreas subtypes from precursors during both embryonic and larval stages, and Neurod function is in turn critical to endocrine function.
Collapse
Affiliation(s)
- Gökhan Dalgin
- Department of Organismal Biology and Anatomy, University of Chicago, Chicago, IL 60637, USA.
| | - Victoria E Prince
- Department of Organismal Biology and Anatomy, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
21
|
Zhang Q, Yang Z, Jia Z, Liu C, Guo C, Lu H, Chen P, Ma K, Wang W, Zhou C. ISL-1 is overexpressed in non-Hodgkin lymphoma and promotes lymphoma cell proliferation by forming a p-STAT3/p-c-Jun/ISL-1 complex. Mol Cancer 2014; 13:181. [PMID: 25070240 PMCID: PMC4125377 DOI: 10.1186/1476-4598-13-181] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 07/22/2014] [Indexed: 12/20/2022] Open
Abstract
Background Insulin enhancer binding protein-1 (ISL-1), a LIM-homeodomain transcription factor, is essential for the heart, motor neuron and pancreas development. Recently, ISL-1 has been found in some types of human cancers. However, how ISL-1 exerts the role in tumor development is not clear. Methods and results The expression of ISL-1 was assessed in 211 human lymphoma samples and 23 normal lymph node samples. Immunohistochemistry results demonstrated a markedly higher expression of ISL-1 in 75% of non-Hodgkin lymphoma (NHL) samples compared with that in normal lymph nodes or Hodgkin lymphoma (HL) samples. CCK-8 analysis, cell cycle assay and xenograft model were performed to characterize the association between ISL-1 expression level and biological functions in NHL. The results showed that ISL-1 overexpression obviously promoted NHL cells proliferation, changed the cell cycle distribution in vitro and significantly enhanced xenografted lymphoma development in vivo. Real-time PCR, Western blot, luciferase assay and ChIP assay were used to explore the potential regulatory targets of ISL-1 and the results demonstrated that ISL-1 activated the c-Myc expression in NHL by direct binding to a conserved binding site on the c-Myc enhancer. Further results revealed that ISL-1 could be positively regulated by the c-Jun N-terminal kinase (JNK) and the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathways. Both the JNK and JAK/STAT signaling inhibitors could significantly suppressed the growth of NHL cells through the down-regulation of ISL-1 as demonstrated by CCK-8 and Western blot assays. Bioinformatic analysis and luciferase assay exhibited that ISL-1 was a novel target of p-STAT3 and p-c-jun. ChIP, Co-IP and ChIP-re-IP analysis revealed that ISL-1 could participate with p-STAT3 and p-c-Jun to form a p-STAT3/p-c-Jun/ISL-1 transcriptional complex that binds directly on the ISL-1 promoter, demonstrating a positive feedback regulatory mechanism for ISL-1 expression in NHL. Conclusions Our results provide the first evidence that ISL-1 is tightly linked to NHL proliferation and development by promoting c-Myc transcription, and its aberrant expression was regulated by p-STAT3/p-c-Jun/ISL-1 complex activation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Weiping Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education of China, Peking University, 38 Xueyuan Road, 100191 Beijing, China.
| | | |
Collapse
|