1
|
Karampelias C, Liu KC, Tengholm A, Andersson O. Mechanistic insights and approaches for beta cell regeneration. Nat Chem Biol 2025:10.1038/s41589-024-01822-y. [PMID: 39881214 DOI: 10.1038/s41589-024-01822-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 12/09/2024] [Indexed: 01/31/2025]
Abstract
Diabetes is characterized by variable loss of insulin-producing beta cells, and new regenerative approaches to increasing the functional beta cell mass of patients hold promise for reversing disease progression. In this Review, we summarize recent chemical biology breakthroughs advancing our knowledge of beta cell regeneration. We present current chemical-based tools, sensors and mechanistic insights into pathways that can be targeted to enhance beta cell regeneration in model organisms. We group the pathways according to the cellular processes they affect, that is, proliferation, conversion of other mature cell types to beta cells and beta cell differentiation from progenitor-like populations. We also suggest assays for assessing the functionality of the regenerated beta cells. Although regeneration processes differ between animal models, such as zebrafish, mice and pigs, regenerative mechanisms identified in any one animal model may be translatable to humans. Overall, chemical biology-based approaches in beta cell regeneration give hope that specific molecular pathways can be targeted to enhance beta cell regeneration.
Collapse
Affiliation(s)
- Christos Karampelias
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany.
| | - Ka-Cheuk Liu
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden
| | - Anders Tengholm
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden
| | - Olov Andersson
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden.
| |
Collapse
|
2
|
Wu Y, Lin Z, Chen F, Zhang X, Liu Y, Sun H. Evaluation of aspartame effects at environmental concentration on early development of zebrafish: Morphology and transcriptome 1. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 361:124792. [PMID: 39182820 DOI: 10.1016/j.envpol.2024.124792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/13/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
The use of aspartame as an artificial sweetener is prevalent in a wide range of everyday food products, potentially leading to health complications such as obesity, diabetes mellitus, autism spectrum disorders, and neurodegeneration. Aspartame has also been detected in natural water bodies at a concentration of 0.49 μg/L, yet research on its ecotoxicological effects on aquatic life remains scarce. This study aimed to investigate the potential negative effects of environmentally relevant concentrations of aspartame on the development of various tissues and organs in zebrafish embryos. We used a zebrafish model to treat embryos with aspartame at environmental concentration and those higher than in the environment-up to 1000 times. We observed that after exposure to aspartame body length increased, pigmentation was delayed, and neutrophil production inhibited in zebrafish. Furthermore, transcriptome analysis revealed that early exposure of zebrafish embryos to aspartame affected the transcriptomics of various systems, primarily by downregulating genes related to immune cell production, eye and optic nerve development, nervous system development, and growth hormone-related transcription. Most of the genes associated with ferroptosis were upregulated. This study provides new insights into the ecotoxicological effects of aspartame on aquatic environments.
Collapse
Affiliation(s)
- Yitian Wu
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, 610041, China
| | - Ziyuan Lin
- SCU-CUHK Joint Laboratory for Reproductive Medicine, Zebrafish Research Platform, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, 610041, China
| | - Feng Chen
- SCU-CUHK Joint Laboratory for Reproductive Medicine, Zebrafish Research Platform, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, 610041, China
| | - Xuan Zhang
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, 610041, China
| | - Yanyan Liu
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, 610041, China.
| | - Huaqin Sun
- SCU-CUHK Joint Laboratory for Reproductive Medicine, Zebrafish Research Platform, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, 610041, China; Children's Medicine Key Laboratory of Sichuan Province, China.
| |
Collapse
|
3
|
Rosa F, Dray N, Bedu S, Bally-Cuif L. Non-apoptotic caspase events and Atf3 expression underlie direct neuronal differentiation of adult neural stem cells. Development 2024; 151:dev204381. [PMID: 39565097 DOI: 10.1242/dev.204381] [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: 09/09/2024] [Accepted: 10/08/2024] [Indexed: 11/21/2024]
Abstract
Neural stem cells (NSCs) generate neurons over a lifetime in adult vertebrate brains. In the adult zebrafish pallium, NSCs persist long term through balanced fate decisions. These decisions include direct neuronal conversions, i.e. delamination and neurogenesis without a division. To characterize this process, we reanalyze intravital imaging data of adult pallial NSCs, and observe shared delamination dynamics between NSCs and committed neuronal progenitors. Searching for mechanisms predicting direct NSC conversions, we build an NSC-specific genetic tracer of Caspase3/7 activation (Cas3*/Cas7*) in vivo. We show that non-apoptotic Cas3*/7* events occur in adult NSCs and are biased towards lineage termination under physiological conditions, with a predominant generation of single neurons. We further identify the transcription factor Atf3 as necessary for this bias. Finally, we show that the Cas3*/7* pathway is engaged by NSCs upon parenchymal lesion and correlates with NSCs more prone to lineage termination and neuron formation. These results provide evidence for non-apoptotic caspase events occurring in vertebrate adult NSCs and link these events with the NSC fate decision of direct conversion, which is important for long-term NSC population homeostasis.
Collapse
Affiliation(s)
- Frédéric Rosa
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, F-75015 Paris, France
| | - Nicolas Dray
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, F-75015 Paris, France
| | - Sébastien Bedu
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, F-75015 Paris, France
| | - Laure Bally-Cuif
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, F-75015 Paris, France
| |
Collapse
|
4
|
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
|
5
|
Chen Q, Leshkowitz D, Li H, van Impel A, Schulte-Merker S, Amit I, Rizzoti K, Levkowitz G. Neural plate progenitors give rise to both anterior and posterior pituitary cells. Dev Cell 2023; 58:2652-2665.e6. [PMID: 37683631 DOI: 10.1016/j.devcel.2023.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 05/14/2023] [Accepted: 08/09/2023] [Indexed: 09/10/2023]
Abstract
The pituitary is the master neuroendocrine gland, which regulates body homeostasis. It consists of the anterior pituitary/adenohypophysis harboring hormones producing cells and the posterior pituitary/neurohypophysis, which relays the passage of hormones from the brain to the periphery. It is accepted that the adenohypophysis originates from the oral ectoderm (Rathke's pouch), whereas the neural ectoderm contributes to the neurohypophysis. Single-cell transcriptomics of the zebrafish pituitary showed that cyp26b1-positive astroglial pituicytes of the neurohypophysis and prop1-positive adenohypophyseal progenitors expressed common markers implying lineage relatedness. Genetic tracing identifies that, in contrast to the prevailing dogma, neural plate precursors of zebrafish (her4.3+) and mouse (Sox1+) contribute to both neurohypophyseal and a subset of adenohypophyseal cells. Pituicyte-derived retinoic-acid-degrading enzyme Cyp26b1 fine-tunes differentiation of prop1+ progenitors into hormone-producing cells. These results challenge the notion that adenohypophyseal cells are exclusively derived from non-neural ectoderm and demonstrate that crosstalk between neuro- and adeno-hypophyseal cells affects differentiation of pituitary cells.
Collapse
Affiliation(s)
- Qiyu Chen
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Dena Leshkowitz
- Life Science Core Facilities, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Hanjie Li
- Department of Systems Immunology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel; Present address: CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Andreas van Impel
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, Cells-in-Motion Cluster of Excellence, WWU Münster, Münster, Germany
| | - Stefan Schulte-Merker
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, Cells-in-Motion Cluster of Excellence, WWU Münster, Münster, Germany
| | - Ido Amit
- Department of Systems Immunology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Karine Rizzoti
- Stem Cell Biology and Developmental Genetics Lab, The Francis Crick Institute, London, UK
| | - Gil Levkowitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel.
| |
Collapse
|
6
|
Matsuda H, Kubota Y. Zebrafish pancreatic β cell clusters undergo stepwise regeneration using Neurod1-expressing cells from different cell lineages. Cell Tissue Res 2023; 394:131-144. [PMID: 37474621 DOI: 10.1007/s00441-023-03805-2] [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: 12/02/2022] [Accepted: 06/30/2023] [Indexed: 07/22/2023]
Abstract
Pancreatic β cell clusters produce insulin and play a central role in glucose homeostasis. The regenerative capacity of mammalian β cells is limited and the loss of β cells causes diabetes. In contrast, zebrafish β cell clusters have a high regenerative capacity, making them an attractive model to study β cell cluster regeneration. How zebrafish β cell clusters regenerate, when the regeneration process is complete, and the identification of the cellular source of regeneration are fundamental questions that require investigation. Here, using larval and adult zebrafish, we demonstrate that pancreatic β cell clusters undergo a two-step regeneration process, regenerating functionality and then β cell numbers. Additionally, we found that all regenerating pancreatic β cells arose from Neurod1-expressing cells and that cells from different lineages contribute to both functional and β cell number recovery throughout their life. Furthermore, we found that during development and neogenesis, as well as regeneration, all β cells undergo Neurod1expression in zebrafish. Together, these results shed light on the fundamental cellular mechanisms underlying β cell cluster development, neogenesis, and regeneration.
Collapse
Affiliation(s)
- Hiroki Matsuda
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, 525-8577, Japan.
- R&D division, Repli-tech Co., Ltd., Shibuya-ku, 150-0012, Japan.
| | - Yukihiko Kubota
- Department of Bioinformatics, College of Life Sciences, Ritsumeikan University, Kusatsu, 525-8577, Japan
| |
Collapse
|
7
|
Mi J, Liu KC, Andersson O. Decoding pancreatic endocrine cell differentiation and β cell regeneration in zebrafish. SCIENCE ADVANCES 2023; 9:eadf5142. [PMID: 37595046 PMCID: PMC10438462 DOI: 10.1126/sciadv.adf5142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 07/20/2023] [Indexed: 08/20/2023]
Abstract
In contrast to mice, zebrafish have an exceptional yet elusive ability to replenish lost β cells in adulthood. Understanding this framework would provide mechanistic insights for β cell regeneration, which may be extrapolated to humans. Here, we characterize a krt4-expressing ductal cell type, which is distinct from the putative Notch-responsive cells, showing neogenic competence and giving rise to the majority of endocrine cells during postembryonic development. Furthermore, we demonstrate a marked ductal remodeling process featuring a Notch-responsive to krt4+ luminal duct transformation during late development, indicating several origins of krt4+ ductal cells displaying similar transcriptional patterns. Single-cell transcriptomics upon a series of time points during β cell regeneration unveil a previously unrecognized dlb+ transitional endocrine precursor cell, distinct regulons, and a differentiation trajectory involving cellular shuffling through differentiation and dedifferentiation dynamics. These results establish a model of zebrafish pancreatic endocrinogenesis and highlight key values of zebrafish for translational studies of β cell regeneration.
Collapse
Affiliation(s)
| | - Ka-Cheuk Liu
- Department of Cell and Molecular Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | | |
Collapse
|
8
|
Leonard ER, Marques ES, Roy MA, Conlin SM, Ranjan R, Timme-Laragy AR. Dietary exposure to the food preservative tert-Butylhydroquinone (tBHQ) impairs zebrafish (Danio rerio) survival, growth, organ development, and gene expression in Nrf2a-dependent and independent ways. Food Chem Toxicol 2023; 176:113788. [PMID: 37075880 PMCID: PMC10213143 DOI: 10.1016/j.fct.2023.113788] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/04/2023] [Accepted: 04/15/2023] [Indexed: 04/21/2023]
Abstract
Tert-Butylhydroquinone (tBHQ), a preservative used to prevent oxidative deterioration of oil, fat, and meat products, has been linked to both chemoprotective and adverse effects. This study investigates the impact of dietary tBHQ consumption on survival, growth parameters, organ development, and gene expression in zebrafish (Danio rerio). As tBHQ activates the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2a), a zebrafish line with a mutation in the DNA-binding domain of Nrf2a was used to identify Nrf2a-dependent vs independent effects. Homozygous Nrf2a wildtype (wt) and mutant (m) larvae were fed a diet containing 5% tBHQ or a control diet. Survival and growth parameters were assessed at 15 days and at 5 months, and samples were collected for RNA sequencing at 5 months. Dietary exposure to tBHQ throughout the larval and juvenile periods negatively impacted growth and survival. RNA-seq analysis found differentially expressed genes related to growth and development and upregulation of several immune system-related pathways. The findings herein demonstrate that dietary tBHQ exposure may impair growth and survival in both Nrf2a dependent and independent manners.
Collapse
Affiliation(s)
- Emily R Leonard
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Emily S Marques
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Monika A Roy
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA; Biotechnology Training Program, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Sarah M Conlin
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Ravi Ranjan
- Genomics Resource Laboratory, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Alicia R Timme-Laragy
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA.
| |
Collapse
|
9
|
Duque M, Amorim JP, Bessa J. Ptf1a function and transcriptional cis-regulation, a cornerstone in vertebrate pancreas development. FEBS J 2022; 289:5121-5136. [PMID: 34125483 PMCID: PMC9545688 DOI: 10.1111/febs.16075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/23/2021] [Accepted: 06/14/2021] [Indexed: 12/11/2022]
Abstract
Vertebrate pancreas organogenesis is a stepwise process regulated by a complex network of signaling and transcriptional events, progressively steering the early endoderm toward pancreatic fate. Many crucial players of this process have been identified, including signaling pathways, cis-regulatory elements, and transcription factors (TFs). Pancreas-associated transcription factor 1a (PTF1A) is one such TF, crucial for pancreas development. PTF1A mutations result in dramatic pancreatic phenotypes associated with severe complications, such as neonatal diabetes and impaired food digestion due to exocrine pancreatic insufficiency. Here, we present a brief overview of vertebrate pancreas development, centered on Ptf1a function and transcriptional regulation, covering similarities and divergences in three broadly studied organisms: human, mouse and zebrafish.
Collapse
Affiliation(s)
- Marta Duque
- Instituto de Biologia Molecular e Celular (IBMC)Universidade do PortoPortugal
- Instituto de Investigação e Inovação em Saúde (i3S)Universidade do PortoPortugal
- Doctoral program in Molecular and Cell Biology (MCbiology)Instituto de Ciências Biomédicas Abel Salazar (ICBAS)Universidade do PortoPortugal
| | - João Pedro Amorim
- Instituto de Biologia Molecular e Celular (IBMC)Universidade do PortoPortugal
- Instituto de Investigação e Inovação em Saúde (i3S)Universidade do PortoPortugal
- Doctoral program in Molecular and Cell Biology (MCbiology)Instituto de Ciências Biomédicas Abel Salazar (ICBAS)Universidade do PortoPortugal
| | - José Bessa
- Instituto de Biologia Molecular e Celular (IBMC)Universidade do PortoPortugal
- Instituto de Investigação e Inovação em Saúde (i3S)Universidade do PortoPortugal
| |
Collapse
|
10
|
Molecular profiling of enteric nervous system cell lineages. Nat Protoc 2022; 17:1789-1817. [PMID: 35676375 DOI: 10.1038/s41596-022-00697-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 03/04/2022] [Indexed: 11/08/2022]
Abstract
The enteric nervous system (ENS) is an extensive network of enteric neurons and glial cells that is intrinsic to the gut wall and regulates almost all aspects of intestinal physiology. While considerable advancement has been made in understanding the genetic programs regulating ENS development, there is limited understanding of the molecular pathways that control ENS function in adult stages. One of the limitations in advancing the molecular characterization of the adult ENS relates to technical difficulties in purifying healthy neurons and glia from adult intestinal tissues. To overcome this, we developed novel methods for performing transcriptomic analysis of enteric neurons and glia, which are based on the isolation of fluorescently labeled nuclei. Here we provide a step-by-step protocol for the labeling of adult mouse enteric neuronal nuclei using adeno-associated-virus-mediated gene transfer, isolation of the labeled nuclei by fluorimetric analysis, RNA purification and nuclear RNA sequencing. This protocol has also been adapted for the isolation of enteric neuron and glia nuclei from myenteric plexus preparations from adult zebrafish intestine. Finally, we describe a method for visualization and quantification of RNA in myenteric ganglia: Spatial Integration of Granular Nuclear Signals (SIGNS). By following this protocol, it takes ~3 d to generate RNA and create cDNA libraries for nuclear RNA sequencing and 4 d to carry out high-resolution RNA expression analysis on ENS tissues.
Collapse
|
11
|
Tel2 regulates redifferentiation of bipotential progenitor cells via Hhex during zebrafish liver regeneration. Cell Rep 2022; 39:110596. [PMID: 35385752 DOI: 10.1016/j.celrep.2022.110596] [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: 10/14/2021] [Revised: 01/27/2022] [Accepted: 03/09/2022] [Indexed: 02/07/2023] Open
Abstract
Upon extensive hepatocyte loss or impaired hepatocyte proliferation, liver regeneration occurs via biliary epithelial cell (BEC) transdifferentiation, which includes dedifferentiation of BECs into bipotential progenitor cells (BP-PCs) and then redifferentiation of BP-PCs to nascent hepatocytes and BECs. This BEC-driven liver regeneration involves reactivation of hepatoblast markers, but the underpinning mechanisms and their effects on liver regeneration remain largely unknown. Using a zebrafish extensive hepatocyte ablation model, we perform an N-ethyl-N-nitrosourea (ENU) forward genetic screen and identify a liver regeneration mutant, liver logan (lvl), in which the telomere maintenance 2 (tel2) gene is mutated. During liver regeneration, the tel2 mutation specifically inhibits transcriptional activation of a hepatoblast marker, hematopoietically expressed homeobox (hhex), in BEC-derived cells, which blocks BP-PC redifferentiation. Mechanistic studies show that Tel2 associates with the hhex promoter region and promotes hhex transcription. Our results reveal roles of Tel2 in the BP-PC redifferentiation process of liver regeneration by activating hhex.
Collapse
|
12
|
Carril Pardo CA, Massoz L, Dupont MA, Bergemann D, Bourdouxhe J, Lavergne A, Tarifeño-Saldivia E, Helker CSM, Stainier DYR, Peers B, Voz MM, Manfroid I. A δ-cell subpopulation with a pro-β-cell identity contributes to efficient age-independent recovery in a zebrafish model of diabetes. eLife 2022; 11:e67576. [PMID: 35060900 PMCID: PMC8820734 DOI: 10.7554/elife.67576] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 01/20/2022] [Indexed: 11/13/2022] Open
Abstract
Restoring damaged β-cells in diabetic patients by harnessing the plasticity of other pancreatic cells raises the questions of the efficiency of the process and of the functionality of the new Insulin-expressing cells. To overcome the weak regenerative capacity of mammals, we used regeneration-prone zebrafish to study β-cells arising following destruction. We show that most new insulin cells differ from the original β-cells as they coexpress Somatostatin and Insulin. These bihormonal cells are abundant, functional and able to normalize glycemia. Their formation in response to β-cell destruction is fast, efficient, and age-independent. Bihormonal cells are transcriptionally close to a subset of δ-cells that we identified in control islets and that are characterized by the expression of somatostatin 1.1 (sst1.1) and by genes essential for glucose-induced Insulin secretion in β-cells such as pdx1, slc2a2 and gck. We observed in vivo the conversion of monohormonal sst1.1-expressing cells to sst1.1+ ins + bihormonal cells following β-cell destruction. Our findings support the conclusion that sst1.1 δ-cells possess a pro-β identity enabling them to contribute to the neogenesis of Insulin-producing cells during regeneration. This work unveils that abundant and functional bihormonal cells benefit to diabetes recovery in zebrafish.
Collapse
Affiliation(s)
| | - Laura Massoz
- Zebrafish Development and Disease Models laboratory, GIGA-Stem Cells, University of LiègeLiègeBelgium
| | - Marie A Dupont
- Zebrafish Development and Disease Models laboratory, GIGA-Stem Cells, University of LiègeLiègeBelgium
| | - David Bergemann
- Zebrafish Development and Disease Models laboratory, GIGA-Stem Cells, University of LiègeLiègeBelgium
| | - Jordane Bourdouxhe
- Zebrafish Development and Disease Models laboratory, GIGA-Stem Cells, University of LiègeLiègeBelgium
| | - Arnaud Lavergne
- Zebrafish Development and Disease Models laboratory, GIGA-Stem Cells, University of LiègeLiègeBelgium
- GIGA-Genomics core facility, University of LiègeLiègeBelgium
| | - Estefania Tarifeño-Saldivia
- Zebrafish Development and Disease Models laboratory, GIGA-Stem Cells, University of LiègeLiègeBelgium
- Gene Expression and Regulation Laboratory, Department of Biochemistry and Molecular Biology, University of ConcepciónConcepciónChile
| | - Christian SM Helker
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Didier YR Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Bernard Peers
- Zebrafish Development and Disease Models laboratory, GIGA-Stem Cells, University of LiègeLiègeBelgium
| | - Marianne M Voz
- Zebrafish Development and Disease Models laboratory, GIGA-Stem Cells, University of LiègeLiègeBelgium
| | - Isabelle Manfroid
- Zebrafish Development and Disease Models laboratory, GIGA-Stem Cells, University of LiègeLiègeBelgium
| |
Collapse
|
13
|
Moortgat S, Manfroid I, Pendeville H, Freeman S, Bourdouxhe J, Benoit V, Merhi A, Philippe C, Faivre L, Maystadt I. Broadening the phenotypic spectrum and physiological insights related to EIF2S3 variants. Hum Mutat 2021; 42:827-834. [PMID: 33942450 DOI: 10.1002/humu.24215] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 04/09/2021] [Accepted: 04/27/2021] [Indexed: 01/20/2023]
Abstract
Mental deficiency, epilepsy, hypogonadism, microcephaly, and obesity syndrome is a severe X-linked syndrome caused by pathogenic variants in EIF2S3. The gene encodes the γ subunit of the eukaryotic translation initiation factor-2, eIF2, essential for protein translation. A recurrent frameshift variant is described in severely affected patients while missense variants usually cause a moderate phenotype. We identified a novel missense variant (c.433A>G, p.(Met145Val)) in EIF2S3 in a mildly affected patient. Studies on zebrafish confirm the pathogenicity of this novel variant and three previously published missense variants. CRISPR/Cas9 knockout of eif2s3 in zebrafish embryos recapitulate the human microcephaly and show increased neuronal cell death. Abnormal high glucose levels were identified in mutant embryos, caused by beta cell and pancreatic progenitor deficiency, not related to apoptosis. Additional studies in patient-derived fibroblasts did not reveal apoptosis. Our results provide new insights into disease physiopathology, suggesting tissue-dependent mechanisms.
Collapse
Affiliation(s)
- Stephanie Moortgat
- Center de Génétique Humaine, Institut de Pathologie et de Génétique, Gosselies, Belgium
| | - Isabelle Manfroid
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA-Research, Tour B34, Université de Liège, Liège (Sart-Tilman), Belgium
| | - Hélène Pendeville
- GIGA-Research, Zebrafish Platform, Tour B34, Université de Liège, Liège (Sart-Tilman), Belgium
| | - Stephen Freeman
- GIGA-Research, Imaging and Flow Cytometry Platform, Tour B34, Université de Liège, Liège (Sart-Tilman), Belgium
| | - Jordane Bourdouxhe
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA-Research, Tour B34, Université de Liège, Liège (Sart-Tilman), Belgium
| | - Valérie Benoit
- Center de Génétique Humaine, Institut de Pathologie et de Génétique, Gosselies, Belgium
| | - Ahmad Merhi
- Laboratory of Translational Oncology, Institut de Pathologie et de Génétique, Gosselies, Belgium.,IPG BioBank, Institut de Pathologie et de Génétique, 6041 Charleroi, Gosselies, Belgium
| | - Christophe Philippe
- Inserm UMR 1231 GAD, Genetics of Developmental disorders, Université de Bourgogne-Franche Comté, Dijon, France.,Unité Fonctionnelle « Innovation diagnostique dans les maladies rares », laboratoire de génétique moléculaire, plate-forme de biologie hospitalo-universitaire, CHU Dijon, Dijon, France
| | - Laurence Faivre
- Inserm UMR 1231 GAD, Genetics of Developmental disorders, Université de Bourgogne-Franche Comté, Dijon, France.,Center de Génétique et Center de Référence Maladies Rares « Anomalies du Développement et Syndromes Malformatifs de l'Interrégion Est », Hôpital d'Enfants, CHU, Dijon, France.,Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Centre Hospitalier Universitaire Dijon, Dijon, France
| | - Isabelle Maystadt
- Center de Génétique Humaine, Institut de Pathologie et de Génétique, Gosselies, Belgium.,Faculté de Médecine, URPhyM, UNamur, Namur, Belgium
| |
Collapse
|
14
|
Zhang W, Chen J, Ni R, Yang Q, Luo L, He J. Contributions of biliary epithelial cells to hepatocyte homeostasis and regeneration in zebrafish. iScience 2021; 24:102142. [PMID: 33665561 PMCID: PMC7900353 DOI: 10.1016/j.isci.2021.102142] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/03/2020] [Accepted: 01/29/2021] [Indexed: 12/27/2022] Open
Abstract
Whether transdifferentiation of the biliary epithelial cells (BECs) to hepatocytes occurs under physiological conditions and contributes to liver homeostasis remains under long-term debate. Similar questions have been raised under pathological circumstances if a fibrotic liver is suffered from severe injuries. To address these questions in zebrafish, we established a sensitive lineage tracing system specific for the detection of BEC-derived hepatocytes. The BEC-to-hepatocyte transdifferentiation occurred and became minor contributors to hepatocyte homeostasis in a portion of adult individuals. The BEC-derived hepatocytes distributed in clusters in the liver. When a fibrotic liver underwent extreme hepatocyte damages, BEC-to-hepatocyte transdifferentiation acted as the major origin of regenerating hepatocytes. In contrast, partial hepatectomy failed to induce the BEC-to-hepatocyte conversion. In conclusion, based on a sensitive lineage tracing system, our results suggest that BECs are able to transdifferentiate into hepatocytes and contribute to both physiological hepatocyte homeostasis and pathological regeneration. Developed sensitivity system to trace BECs derived hepatocytes in liver homeostasis BECs convert to hepatocytes in liver homeostasis but are individually heterogeneous BECs are the primary regeneration sources in the extreme injury of the fibrotic liver BECs fail to contribute to new hepatocytes after partial hepatectomy
Collapse
Affiliation(s)
- Wenfeng Zhang
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, 2 Tiansheng Road, Beibei, 400715 Chongqing, China
| | - Jingying Chen
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, 2 Tiansheng Road, Beibei, 400715 Chongqing, China.,University of Chinese Academy of Sciences (Chongqing), Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Beibei, 400714 Chongqing, China
| | - Rui Ni
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, 2 Tiansheng Road, Beibei, 400715 Chongqing, China
| | - Qifen Yang
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, 2 Tiansheng Road, Beibei, 400715 Chongqing, China
| | - Lingfei Luo
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, 2 Tiansheng Road, Beibei, 400715 Chongqing, China
| | - Jianbo He
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, 2 Tiansheng Road, Beibei, 400715 Chongqing, China
| |
Collapse
|
15
|
Freudenblum J, Meyer D, Kimmel RA. Inducible Mosaic Cell Labeling Provides Insights Into Pancreatic Islet Morphogenesis. Front Cell Dev Biol 2020; 8:586651. [PMID: 33102488 PMCID: PMC7546031 DOI: 10.3389/fcell.2020.586651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/02/2020] [Indexed: 11/13/2022] Open
Abstract
Pancreatic islets, discrete microorgans embedded within the exocrine pancreas, contain beta cells which are critical for glucose homeostasis. Loss or dysfunction of beta cells leads to diabetes, a disease with expanding global prevalence, and for which regenerative therapies are actively being pursued. Recent efforts have focused on producing mature beta cells in vitro, but it is increasingly recognized that achieving a faithful three-dimensional islet structure is crucial for generating fully functional beta cells. Our current understanding of islet morphogenesis is far from complete, due to the deep internal location of the pancreas in mammalian models, which hampers direct visualization. Zebrafish is a model system well suited for studies of pancreas morphogenesis due to its transparency and the accessible location of the larval pancreas. In order to further clarify the cellular mechanisms of islet formation, we have developed new tools for in vivo visualization of single-cell dynamics. Our results show that clustering islet cells make contact and interconnect through dynamic actin-rich processes, move together while remaining in close proximity to the duct, and maintain high protrusive motility after forming clusters. Quantitative analyses of cell morphology and motility in 3-dimensions lays the groundwork to define therapeutically applicable factors responsible for orchestrating the morphogenic behaviors of coalescing endocrine cells.
Collapse
Affiliation(s)
- Julia Freudenblum
- Institute of Molecular Biology/CMBI, University of Innsbruck, Innsbruck, Austria
| | - Dirk Meyer
- Institute of Molecular Biology/CMBI, University of Innsbruck, Innsbruck, Austria
| | - Robin A Kimmel
- Institute of Molecular Biology/CMBI, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
16
|
Bloch S, Hagio H, Thomas M, Heuzé A, Hermel JM, Lasserre E, Colin I, Saka K, Affaticati P, Jenett A, Kawakami K, Yamamoto N, Yamamoto K. Non-thalamic origin of zebrafish sensory nuclei implies convergent evolution of visual pathways in amniotes and teleosts. eLife 2020; 9:e54945. [PMID: 32896272 PMCID: PMC7478893 DOI: 10.7554/elife.54945] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 08/14/2020] [Indexed: 12/17/2022] Open
Abstract
Ascending visual projections similar to the mammalian thalamocortical pathway are found in a wide range of vertebrate species, but their homology is debated. To get better insights into their evolutionary origin, we examined the developmental origin of a thalamic-like sensory structure of teleosts, the preglomerular complex (PG), focusing on the visual projection neurons. Similarly to the tectofugal thalamic nuclei in amniotes, the lateral nucleus of PG receives tectal information and projects to the pallium. However, our cell lineage study in zebrafish reveals that the majority of PG cells are derived from the midbrain, unlike the amniote thalamus. We also demonstrate that the PG projection neurons develop gradually until late juvenile stages. Our data suggest that teleost PG, as a whole, is not homologous to the amniote thalamus. Thus, the thalamocortical-like projections evolved from a non-forebrain cell population, which indicates a surprising degree of variation in the vertebrate sensory systems.
Collapse
Affiliation(s)
- Solal Bloch
- Paris-Saclay Institute of Neuroscience (Neuro-PSI), Université Paris-Saclay, CNRSGif-sur-YvetteFrance
| | - Hanako Hagio
- Laboratory of Fish Biology, Graduate School of Bioagricultural Sciences, Nagoya UniversityNagoyaJapan
- Institute for Advanced Research, Nagoya UniversityNagoyaJapan
| | - Manon Thomas
- Paris-Saclay Institute of Neuroscience (Neuro-PSI), Université Paris-Saclay, CNRSGif-sur-YvetteFrance
| | - Aurélie Heuzé
- Paris-Saclay Institute of Neuroscience (Neuro-PSI), Université Paris-Saclay, CNRSGif-sur-YvetteFrance
| | - Jean-Michel Hermel
- Paris-Saclay Institute of Neuroscience (Neuro-PSI), Université Paris-Saclay, CNRSGif-sur-YvetteFrance
| | - Elodie Lasserre
- Paris-Saclay Institute of Neuroscience (Neuro-PSI), Université Paris-Saclay, CNRSGif-sur-YvetteFrance
| | - Ingrid Colin
- Paris-Saclay Institute of Neuroscience (Neuro-PSI), Université Paris-Saclay, CNRSGif-sur-YvetteFrance
| | - Kimiko Saka
- Laboratory of Molecular and Developmental Biology, National Institute of GeneticsMishimaJapan
| | - Pierre Affaticati
- TEFOR Paris-Saclay, CNRS UMS2010, INRA UMS1451, Université Paris-SaclayGif-sur-YvetteFrance
| | - Arnim Jenett
- TEFOR Paris-Saclay, CNRS UMS2010, INRA UMS1451, Université Paris-SaclayGif-sur-YvetteFrance
| | - Koichi Kawakami
- Laboratory of Molecular and Developmental Biology, National Institute of GeneticsMishimaJapan
- Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies)MishimaJapan
| | - Naoyuki Yamamoto
- Laboratory of Fish Biology, Graduate School of Bioagricultural Sciences, Nagoya UniversityNagoyaJapan
| | - Kei Yamamoto
- Paris-Saclay Institute of Neuroscience (Neuro-PSI), Université Paris-Saclay, CNRSGif-sur-YvetteFrance
| |
Collapse
|
17
|
McCallum S, Obata Y, Fourli E, Boeing S, Peddie CJ, Xu Q, Horswell S, Kelsh RN, Collinson L, Wilkinson D, Pin C, Pachnis V, Heanue TA. Enteric glia as a source of neural progenitors in adult zebrafish. eLife 2020; 9:e56086. [PMID: 32851974 PMCID: PMC7521928 DOI: 10.7554/elife.56086] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 08/26/2020] [Indexed: 12/23/2022] Open
Abstract
The presence and identity of neural progenitors in the enteric nervous system (ENS) of vertebrates is a matter of intense debate. Here, we demonstrate that the non-neuronal ENS cell compartment of teleosts shares molecular and morphological characteristics with mammalian enteric glia but cannot be identified by the expression of canonical glial markers. However, unlike their mammalian counterparts, which are generally quiescent and do not undergo neuronal differentiation during homeostasis, we show that a relatively high proportion of zebrafish enteric glia proliferate under physiological conditions giving rise to progeny that differentiate into enteric neurons. We also provide evidence that, similar to brain neural stem cells, the activation and neuronal differentiation of enteric glia are regulated by Notch signalling. Our experiments reveal remarkable similarities between enteric glia and brain neural stem cells in teleosts and open new possibilities for use of mammalian enteric glia as a potential source of neurons to restore the activity of intestinal neural circuits compromised by injury or disease.
Collapse
Affiliation(s)
- Sarah McCallum
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Yuuki Obata
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Evangelia Fourli
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Stefan Boeing
- Bionformatics & Biostatistics Science Technology Platform, The Francis Crick InstituteLondonUnited Kingdom
| | - Christopher J Peddie
- Electron Microscopy Science Technology Platform, The Francis Crick InstituteLondonUnited Kingdom
| | - Qiling Xu
- Neural Development Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Stuart Horswell
- Bionformatics & Biostatistics Science Technology Platform, The Francis Crick InstituteLondonUnited Kingdom
| | - Robert N Kelsh
- Department of Biology and Biochemistry, University of BathBathUnited Kingdom
| | - Lucy Collinson
- Electron Microscopy Science Technology Platform, The Francis Crick InstituteLondonUnited Kingdom
| | - David Wilkinson
- Neural Development Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Carmen Pin
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZenecaCambridgeUnited Kingdom
| | - Vassilis Pachnis
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Tiffany A Heanue
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| |
Collapse
|
18
|
Okuda KS, Hogan BM. Endothelial Cell Dynamics in Vascular Development: Insights From Live-Imaging in Zebrafish. Front Physiol 2020; 11:842. [PMID: 32792978 PMCID: PMC7387577 DOI: 10.3389/fphys.2020.00842] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/23/2020] [Indexed: 01/16/2023] Open
Abstract
The formation of the vertebrate vasculature involves the acquisition of endothelial cell identities, sprouting, migration, remodeling and maturation of functional vessel networks. To understand the cellular and molecular processes that drive vascular development, live-imaging of dynamic cellular events in the zebrafish embryo have proven highly informative. This review focusses on recent advances, new tools and new insights from imaging studies in vascular cell biology using zebrafish as a model system.
Collapse
Affiliation(s)
- Kazuhide S Okuda
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Benjamin M Hogan
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
19
|
Yang B, Covington BA, Chen W. In vivo generation and regeneration of β cells in zebrafish. CELL REGENERATION (LONDON, ENGLAND) 2020; 9:9. [PMID: 32613468 PMCID: PMC7329966 DOI: 10.1186/s13619-020-00052-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023]
Abstract
The pathological feature of diabetes, hyperglycemia, is a result of an inadequate number and/or function of insulin producing β cells. Replenishing functional β cells is a strategy to cure the disease. Although β-cell regeneration occurs in animal models under certain conditions, human β cells are refractory to proliferation. A better understanding of both the positive and the negative regulatory mechanisms of β-cell regeneration in animal models is essential to develop novel strategies capable of inducing functional β cells in patients. Zebrafish are an attractive model system for studying β-cell regeneration due to the ease to which genetic and chemical-genetic approaches can be used as well as their high regenerative capacity. Here, we highlight the current state of β-cell regeneration studies in zebrafish with an emphasis on cell signaling mechanisms.
Collapse
Affiliation(s)
- Bingyuan Yang
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN, 37232, USA
| | - Brittney A Covington
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN, 37232, USA
| | - Wenbiao Chen
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN, 37232, USA.
| |
Collapse
|
20
|
Wang H, Wei X, Shi W, He J, Luo L. Key Developmental Regulators Suggest Multiple Origins of Pancreatic Beta Cell Regeneration. Zebrafish 2020; 17:187-195. [PMID: 32460659 DOI: 10.1089/zeb.2019.1777] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Extensive efforts have been done to try to restore the lost β cell mass for the cure of diabetes. Animal models have been established to provide evidences of cellular origins and contextual regulators of β cell regeneration. Here, we used a zebrafish β cell ablation and regeneration model to investigate β cell neogenesis in the first few days after a near-total β cell loss. Regeneration of β cells first occurred within 7 h post-treatment. Developmental regulators such as neurod, pdx1, mnx1, and nkx2.2a were active in the regenerating β cells, while at the same time suggesting different subpopulations of regenerative cellular origins. Using Cre/loxP-based lineage tracing, we showed that intrapancreatic ductal cells resisted to give rise to regenerating β cells. Given that transdifferentiation of α cell and δ cell can regenerate β cell, here we have provided further molecular evidence highly suggesting that the regenerating β cells originate from multiple cellular origins.
Collapse
Affiliation(s)
- Hao Wang
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, Chongqing, China
| | - Xiangyong Wei
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, Chongqing, China
| | - Wenchao Shi
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, Chongqing, China
| | - Jianbo He
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, Chongqing, China
| | - Lingfei Luo
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, Chongqing, China
| |
Collapse
|
21
|
Chawla P, Delgadillo Silva LF, Ninov N. Insights on β-cell regeneration from the zebrafish shoal: from generation of cells to functional integration. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2019.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
22
|
Li J, Dedloff MR, Stevens K, Maney L, Prochaska M, Hongay CF, Wallace KN. A novel group of secretory cells regulates development of the immature intestinal stem cell niche through repression of the main signaling pathways driving proliferation. Dev Biol 2019; 456:47-62. [PMID: 31398318 DOI: 10.1016/j.ydbio.2019.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 07/23/2019] [Accepted: 08/05/2019] [Indexed: 12/12/2022]
Abstract
The intestinal epithelium has constant turnover throughout the life of the organ, with apoptosis of cells at the tips of folds or villi releasing cells into the lumen. Due to constant turnover, epithelial cells need to be constantly replaced. Epithelial cells are supplied by stem cell niches that form at the base of the interfold space (zebrafish) and crypts (birds and mammals). Within the adult stem cell niche of mammals, secretory cells such as Paneth and goblet cells play a role in modulation of proliferation and stem cell activity, producing asymmetric divisions. Progeny of asymmetric divisions move up the fold or villi, giving rise to all of the epithelial cell types. Although much is known about function and organization of the adult intestinal stem cell niche, less is understood about regulation within the immature stem cell compartment. Following smooth muscle formation, the intestinal epithelium folds and proliferation becomes restricted to the interfold base. Symmetric divisions continue in the developing interfold niche until stem cell progeny begin asymmetric divisions, producing progeny that migrate up the developing folds. Proliferative progeny from the developing stem cell niche begin migrating out of the niche during the third week post-embryogenesis (zebrafish) or during the postnatal period (mammals). Regulation and organization of epithelial proliferation in the immature stem cell niche may be regulated by signals comparable to the adult niche. Here we identify a novel subset of secretory cells associated with the developing stem cell niche that receive Notch signaling (referred to as NRSCs). Inhibition of the embryonic NRSCs between 74 hpf to 120 hpf increases epithelial proliferation as well as EGF and IGF signaling. Inhibition of post-embryonic NRSCs (6 hpf to 12 dpf) also increases epithelial proliferation and expression level of Wnt target genes. We conclude that NRSCs play a role in modulation of epithelial proliferation through repression of signaling pathways that drive proliferation during both embryogenesis and the post embryonic period.
Collapse
Affiliation(s)
- Jianlong Li
- Department of Biology, Clarkson University, Potsdam, NY, USA
| | | | - Katrina Stevens
- Department of Biology, Clarkson University, Potsdam, NY, USA
| | - Lea Maney
- Department of Biology, Clarkson University, Potsdam, NY, USA
| | | | - Cintia F Hongay
- Department of Biology, Clarkson University, Potsdam, NY, USA
| | | |
Collapse
|
23
|
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
|
24
|
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
|
25
|
Kleinert M, Clemmensen C, Hofmann SM, Moore MC, Renner S, Woods SC, Huypens P, Beckers J, de Angelis MH, Schürmann A, Bakhti M, Klingenspor M, Heiman M, Cherrington AD, Ristow M, Lickert H, Wolf E, Havel PJ, Müller TD, Tschöp MH. Animal models of obesity and diabetes mellitus. Nat Rev Endocrinol 2018; 14:140-162. [PMID: 29348476 DOI: 10.1038/nrendo.2017.161] [Citation(s) in RCA: 569] [Impact Index Per Article: 81.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
More than one-third of the worldwide population is overweight or obese and therefore at risk of developing type 2 diabetes mellitus. In order to mitigate this pandemic, safer and more potent therapeutics are urgently required. This necessitates the continued use of animal models to discover, validate and optimize novel therapeutics for their safe use in humans. In order to improve the transition from bench to bedside, researchers must not only carefully select the appropriate model but also draw the right conclusions. In this Review, we consolidate the key information on the currently available animal models of obesity and diabetes and highlight the advantages, limitations and important caveats of each of these models.
Collapse
Affiliation(s)
- Maximilian Kleinert
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, D-80333 Munich, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Christoffer Clemmensen
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, D-80333 Munich, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Susanna M Hofmann
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Institute for Diabetes and Regeneration Research, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Ziemssenstr. 1, D-80336 Munich, Germany
| | - Mary C Moore
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee 37212, USA
| | - Simone Renner
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilan University München, Feodor-Lynen-Str. 25, D-81377 Munich, Germany
| | - Stephen C Woods
- University of Cincinnati College of Medicine, Department of Psychiatry and Behavioral Neuroscience, Metabolic Diseases Institute, 2170 East Galbraith Road, Cincinnati, Ohio 45237, USA
| | - Peter Huypens
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Johannes Beckers
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Technische Universität München, Chair of Experimental Genetics, D-85354 Freising, Germany
| | - Martin Hrabe de Angelis
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Technische Universität München, Chair of Experimental Genetics, D-85354 Freising, Germany
| | - Annette Schürmann
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Department of Experimental Diabetology, German Institute of Human Nutrition (DIfE), Arthur-Scheunert-Allee 114-116, D-14558 Nuthetal, Germany
| | - Mostafa Bakhti
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Institute for Diabetes and Regeneration Research, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Martin Klingenspor
- Chair of Molecular Nutritional Medicine, Technische Universität München, TUM School of Life Sciences Weihenstephan, Gregor-Mendel-Str. 2, D-85354 Freising, Germany
- Else Kröner-Fresenius Center for Nutritional Medicine, Technische Universität München, D-85354 Freising, Germany
- Institute for Food & Health, Technische Universität München, D-85354 Freising, Germany
| | - Mark Heiman
- MicroBiome Therapeutics, 1316 Jefferson Ave, New Orleans, Louisiana 70115, USA
| | - Alan D Cherrington
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee 37212, USA
| | - Michael Ristow
- Energy Metabolism Laboratory, Institute of Translational Medicine, Swiss Federal Institute of Technology (ETH) Zurich, CH-8603 Zurich-Schwerzenbach, Switzerland
| | - Heiko Lickert
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Institute for Diabetes and Regeneration Research, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Eckhard Wolf
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilan University München, Feodor-Lynen-Str. 25, D-81377 Munich, Germany
| | - Peter J Havel
- Department of Molecular Biosciences, School of Veterinary Medicine and Department of Nutrition, 3135 Meyer Hall, University of California, Davis, California 95616-5270, USA
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, D-80333 Munich, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, D-80333 Munich, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| |
Collapse
|
26
|
Gao C, Zhu Z, Gao Y, Lo LJ, Chen J, Luo L, Peng J. Hepatocytes in a normal adult liver are derived solely from the embryonic hepatocytes. J Genet Genomics 2018; 45:173-175. [PMID: 29574114 DOI: 10.1016/j.jgg.2017.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 12/14/2017] [Accepted: 12/28/2017] [Indexed: 01/13/2023]
Affiliation(s)
- Ce Gao
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhihui Zhu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuqi Gao
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Li Jan Lo
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jun Chen
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lingfei Luo
- College of Life Sciences, Southwest University, Chongqing 400715, China
| | - Jinrong Peng
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
27
|
Carney TJ, Mosimann C. Switch and Trace: Recombinase Genetics in Zebrafish. Trends Genet 2018; 34:362-378. [PMID: 29429760 DOI: 10.1016/j.tig.2018.01.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/04/2018] [Accepted: 01/08/2018] [Indexed: 01/04/2023]
Abstract
Transgenic approaches are instrumental for labeling and manipulating cells and cellular machineries in vivo. Transgenes have traditionally been static entities that remained unaltered following genome integration, limiting their versatility. The development of DNA recombinase-based methods to modify, excise, or rearrange transgene cassettes has introduced versatile control of transgene activity and function. In particular, recombinase-controlled transgenes enable regulation of exogenous gene expression, conditional mutagenesis, and genetic lineage tracing. In zebrafish, transgenesis-based recombinase genetics using Cre/lox, Flp/FRT, and ΦC31 are increasingly applied to study development and homeostasis, and to generate disease models. Intersected with the versatile imaging capacity of the zebrafish model and recent breakthroughs in genome editing, we review and discuss past, current, and potential future approaches and resources for recombinase-based techniques in zebrafish.
Collapse
Affiliation(s)
- Tom J Carney
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology, and Research (A*STAR), Singapore.
| | - Christian Mosimann
- Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
28
|
Freudenblum J, Iglesias JA, Hermann M, Walsen T, Wilfinger A, Meyer D, Kimmel RA. In vivo imaging of emerging endocrine cells reveals a requirement for PI3K-regulated motility in pancreatic islet morphogenesis. Development 2018; 145:dev158477. [PMID: 29386244 PMCID: PMC5818004 DOI: 10.1242/dev.158477] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 01/10/2018] [Indexed: 01/03/2023]
Abstract
The three-dimensional architecture of the pancreatic islet is integral to beta cell function, but the process of islet formation remains poorly understood due to the difficulties of imaging internal organs with cellular resolution. Within transparent zebrafish larvae, the developing pancreas is relatively superficial and thus amenable to live imaging approaches. We performed in vivo time-lapse and longitudinal imaging studies to follow islet development, visualizing both naturally occurring islet cells and cells arising with an accelerated timecourse following an induction approach. These studies revealed previously unappreciated fine dynamic protrusions projecting between neighboring and distant endocrine cells. Using pharmacological compound and toxin interference approaches, and single-cell analysis of morphology and cell dynamics, we determined that endocrine cell motility is regulated by phosphoinositide 3-kinase (PI3K) and G-protein-coupled receptor (GPCR) signaling. Linking cell dynamics to islet formation, perturbation of protrusion formation disrupted endocrine cell coalescence, and correlated with decreased islet cell differentiation. These studies identified novel cell behaviors contributing to islet morphogenesis, and suggest a model in which dynamic exploratory filopodia establish cell-cell contacts that subsequently promote cell clustering.
Collapse
Affiliation(s)
- Julia Freudenblum
- Institute of Molecular Biology/CMBI, University of Innsbruck, Technikerstrasse 25, A-6020 Innsbruck, Austria
| | - José A Iglesias
- Johann Radon Institute for Computational and Applied Mathematics (RICAM), Austrian Academy of Sciences, Altenbergerstrasse 69, A-4040 Linz, Austria
| | - Martin Hermann
- Department of Anaesthesiology and Critical Care Medicine, Innsbruck Medical University, Innrain 66, 6020 Innsbruck, Austria
| | - Tanja Walsen
- Department of Neurosurgery, Medical University of Innsbruck, 6020 Innsbruck Austria
| | - Armin Wilfinger
- Institute of Molecular Biology/CMBI, University of Innsbruck, Technikerstrasse 25, A-6020 Innsbruck, Austria
| | - Dirk Meyer
- Institute of Molecular Biology/CMBI, University of Innsbruck, Technikerstrasse 25, A-6020 Innsbruck, Austria
| | - Robin A Kimmel
- Institute of Molecular Biology/CMBI, University of Innsbruck, Technikerstrasse 25, A-6020 Innsbruck, Austria
| |
Collapse
|
29
|
Furlan G, Cuccioli V, Vuillemin N, Dirian L, Muntasell AJ, Coolen M, Dray N, Bedu S, Houart C, Beaurepaire E, Foucher I, Bally-Cuif L. Life-Long Neurogenic Activity of Individual Neural Stem Cells and Continuous Growth Establish an Outside-In Architecture in the Teleost Pallium. Curr Biol 2017; 27:3288-3301.e3. [PMID: 29107546 PMCID: PMC5678050 DOI: 10.1016/j.cub.2017.09.052] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 08/14/2017] [Accepted: 09/25/2017] [Indexed: 01/08/2023]
Abstract
Spatiotemporal variations of neurogenesis are thought to account for the evolution of brain shape. In the dorsal telencephalon (pallium) of vertebrates, it remains unresolved which ancestral neurogenesis mode prefigures the highly divergent cytoarchitectures that are seen in extant species. To gain insight into this question, we developed genetic tools to generate here the first 4-dimensional (3D + birthdating time) map of pallium construction in the adult teleost zebrafish. Using a Tet-On-based genetic birthdating strategy, we identify a “sequential stacking” construction mode where neurons derived from the zebrafish pallial germinal zone arrange in outside-in, age-related layers from a central core generated during embryogenesis. We obtained no evidence for overt radial or tangential neuronal migrations. Cre-lox-mediated tracing, which included following Brainbow clones, further demonstrates that this process is sustained by the persistent neurogenic activity of individual pallial neural stem cells (NSCs) from embryo to adult. Together, these data demonstrate that the spatiotemporal control of NSC activity is an important driver of the macroarchitecture of the zebrafish adult pallium. This simple mode of pallium construction shares distinct traits with pallial genesis in mammals and non-mammalian amniotes such as birds or reptiles, suggesting that it may exemplify the basal layout from which vertebrate pallial architectures were elaborated. Neurons of the teleost pallium are arranged in concentric age-dependent layers Neurons of the central pallial domain, Dc, are born during embryogenesis Most pallial neurons are generated from ventricular her4-positive radial glia The majority of individual pallial radial glia are neurogenic throughout life
Collapse
Affiliation(s)
- Giacomo Furlan
- Team Zebrafish Neurogenetics, Paris-Saclay Institute for Neuroscience (Neuro-PSI), UMR 9197, CNRS-Université Paris-Sud, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France
| | - Valentina Cuccioli
- Team Zebrafish Neurogenetics, Paris-Saclay Institute for Neuroscience (Neuro-PSI), UMR 9197, CNRS-Université Paris-Sud, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France; Unit Zebrafish Neurogenetics, Developmental and Stem Cell Biology Department, Institut Pasteur, 25 Rue du Dr Roux, 75015 Paris, France; CNRS UMR 3738, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Nelly Vuillemin
- Laboratory for Optics and Biosciences, École Polytechnique, CNRS UMR 7645 and INSERM U1182, 91128 Palaiseau, France
| | - Lara Dirian
- Team Zebrafish Neurogenetics, Paris-Saclay Institute for Neuroscience (Neuro-PSI), UMR 9197, CNRS-Université Paris-Sud, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France
| | - Anna Janue Muntasell
- Centre for Developmental Neurobiology and MRC Centre for Neurodevelopmental Disorders, IoPPN, King's College London, London SE1 1UL, UK
| | - Marion Coolen
- Team Zebrafish Neurogenetics, Paris-Saclay Institute for Neuroscience (Neuro-PSI), UMR 9197, CNRS-Université Paris-Sud, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France; Unit Zebrafish Neurogenetics, Developmental and Stem Cell Biology Department, Institut Pasteur, 25 Rue du Dr Roux, 75015 Paris, France; CNRS UMR 3738, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Nicolas Dray
- Team Zebrafish Neurogenetics, Paris-Saclay Institute for Neuroscience (Neuro-PSI), UMR 9197, CNRS-Université Paris-Sud, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France; Unit Zebrafish Neurogenetics, Developmental and Stem Cell Biology Department, Institut Pasteur, 25 Rue du Dr Roux, 75015 Paris, France; CNRS UMR 3738, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Sébastien Bedu
- Team Zebrafish Neurogenetics, Paris-Saclay Institute for Neuroscience (Neuro-PSI), UMR 9197, CNRS-Université Paris-Sud, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France; Unit Zebrafish Neurogenetics, Developmental and Stem Cell Biology Department, Institut Pasteur, 25 Rue du Dr Roux, 75015 Paris, France; CNRS UMR 3738, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Corinne Houart
- Centre for Developmental Neurobiology and MRC Centre for Neurodevelopmental Disorders, IoPPN, King's College London, London SE1 1UL, UK
| | - Emmanuel Beaurepaire
- Laboratory for Optics and Biosciences, École Polytechnique, CNRS UMR 7645 and INSERM U1182, 91128 Palaiseau, France
| | - Isabelle Foucher
- Team Zebrafish Neurogenetics, Paris-Saclay Institute for Neuroscience (Neuro-PSI), UMR 9197, CNRS-Université Paris-Sud, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France; Unit Zebrafish Neurogenetics, Developmental and Stem Cell Biology Department, Institut Pasteur, 25 Rue du Dr Roux, 75015 Paris, France; CNRS UMR 3738, 25 Rue du Dr. Roux, 75015 Paris, France.
| | - Laure Bally-Cuif
- Team Zebrafish Neurogenetics, Paris-Saclay Institute for Neuroscience (Neuro-PSI), UMR 9197, CNRS-Université Paris-Sud, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France; Unit Zebrafish Neurogenetics, Developmental and Stem Cell Biology Department, Institut Pasteur, 25 Rue du Dr Roux, 75015 Paris, France; CNRS UMR 3738, 25 Rue du Dr. Roux, 75015 Paris, France.
| |
Collapse
|
30
|
Zhang D, Gates KP, Barske L, Wang G, Lancman JJ, Zeng XXI, Groff M, Wang K, Parsons MJ, Crump JG, Dong PDS. Endoderm Jagged induces liver and pancreas duct lineage in zebrafish. Nat Commun 2017; 8:769. [PMID: 28974684 PMCID: PMC5626745 DOI: 10.1038/s41467-017-00666-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 07/15/2017] [Indexed: 12/17/2022] Open
Abstract
Liver duct paucity is characteristic of children born with Alagille Syndrome (ALGS), a disease associated with JAGGED1 mutations. Here, we report that zebrafish embryos with compound homozygous mutations in two Notch ligand genes, jagged1b (jag1b) and jagged2b (jag2b) exhibit a complete loss of canonical Notch activity and duct cells within the liver and exocrine pancreas, whereas hepatocyte and acinar pancreas development is not affected. Further, animal chimera studies demonstrate that wild-type endoderm cells within the liver and pancreas can rescue Notch activity and duct lineage specification in adjacent cells lacking jag1b and jag2b expression. We conclude that these two Notch ligands are directly and solely responsible for all duct lineage specification in these organs in zebrafish. Our study uncovers genes required for lineage specification of the intrahepatopancreatic duct cells, challenges the role of duct cells as progenitors, and suggests a genetic mechanism for ALGS ductal paucity.The hepatopancreatic duct cells connect liver hepatocytes and pancreatic acinar cells to the intestine, but the mechanism for their lineage specification is unclear. Here, the authors reveal that Notch ligands Jagged1b and Jagged2b induce duct cell lineage in the liver and pancreas of the zebrafish.
Collapse
Affiliation(s)
- Danhua Zhang
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
- Graduate School of Biomedical, Science, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Keith P Gates
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Lindsey Barske
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA, 90033, USA
| | - Guangliang Wang
- Department of Surgery, and McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University School of Medicine, 733N. Broadway, Baltimore, MD, 21205, USA
| | - Joseph J Lancman
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Xin-Xin I Zeng
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Megan Groff
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA, 90033, USA
| | - Kasper Wang
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA, 90033, USA
| | - Michael J Parsons
- Department of Surgery, and McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University School of Medicine, 733N. Broadway, Baltimore, MD, 21205, USA
| | - J Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA, 90033, USA
| | - P Duc Si Dong
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA.
- Graduate School of Biomedical, Science, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA.
| |
Collapse
|
31
|
Different developmental histories of beta-cells generate functional and proliferative heterogeneity during islet growth. Nat Commun 2017; 8:664. [PMID: 28939870 PMCID: PMC5610262 DOI: 10.1038/s41467-017-00461-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 06/30/2017] [Indexed: 12/11/2022] Open
Abstract
The proliferative and functional heterogeneity among seemingly uniform cells is a universal phenomenon. Identifying the underlying factors requires single-cell analysis of function and proliferation. Here we show that the pancreatic beta-cells in zebrafish exhibit different growth-promoting and functional properties, which in part reflect differences in the time elapsed since birth of the cells. Calcium imaging shows that the beta-cells in the embryonic islet become functional during early zebrafish development. At later stages, younger beta-cells join the islet following differentiation from post-embryonic progenitors. Notably, the older and younger beta-cells occupy different regions within the islet, which generates topological asymmetries in glucose responsiveness and proliferation. Specifically, the older beta-cells exhibit robust glucose responsiveness, whereas younger beta-cells are more proliferative but less functional. As the islet approaches its mature state, heterogeneity diminishes and beta-cells synchronize function and proliferation. Our work illustrates a dynamic model of heterogeneity based on evolving proliferative and functional beta-cell states. Βeta-cells have recently been shown to be heterogeneous with regard to morphology and function. Here, the authors show that β-cells in zebrafish switch from proliferative to functional states with increasing time since β-cell birth, leading to functional and proliferative heterogeneity.
Collapse
|
32
|
Ahmed Alfar E, Kirova D, Konantz J, Birke S, Mansfeld J, Ninov N. Distinct Levels of Reactive Oxygen Species Coordinate Metabolic Activity with Beta-cell Mass Plasticity. Sci Rep 2017; 7:3994. [PMID: 28652605 PMCID: PMC5484671 DOI: 10.1038/s41598-017-03873-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 05/08/2017] [Indexed: 11/13/2022] Open
Abstract
The pancreatic beta-cells control glucose homeostasis by secreting insulin in response to nutrient intake. The number of beta-cells is under tight metabolic control, as this number increases with higher nutrient intake. However, the signaling pathways matching nutrition with beta-cell mass plasticity remain poorly defined. By applying pharmacological and genetic manipulations, we show that reactive oxygen species (ROS) regulate dose-dependently beta-cell proliferation in vivo and in vitro. In particular, reducing ROS levels in beta-cells blocks their proliferation in response to nutrients. Using a non-invasive genetic sensor of intracellular hydrogen peroxide (H2O2), we reveal that glucose can directly increase the levels of H2O2. Furthermore, a moderate increase in H2O2 levels can stimulate beta-cell proliferation. Interestingly, while high H2O2 levels are inhibitory to beta-cell proliferation, they expand beta-cell mass in vivo by inducing rapid beta-cell neogenesis. Our study thus reveals a ROS-level-dependent mechanism linking nutrients with beta-cell mass plasticity. Hence, given the requirement of ROS for beta-cell mass expansion, antioxidant therapies should be applied with caution in diabetes.
Collapse
Affiliation(s)
- Ezzaldin Ahmed Alfar
- DFG Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany.,Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Hospital and Faculty of Medicine Carl Gustav Carus of Technische Universität Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Neuherberg, Germany.,Department of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Dilyana Kirova
- Cell Cycle, Biotechnology Center, Technische Universität Dresden, 01307, Dresden, Germany
| | - Judith Konantz
- DFG Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Sarah Birke
- DFG Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Jörg Mansfeld
- Cell Cycle, Biotechnology Center, Technische Universität Dresden, 01307, Dresden, Germany.
| | - Nikolay Ninov
- DFG Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany. .,Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Hospital and Faculty of Medicine Carl Gustav Carus of Technische Universität Dresden, Dresden, Germany. .,German Center for Diabetes Research (DZD e.V.), Neuherberg, Neuherberg, Germany.
| |
Collapse
|
33
|
Liu Z, Chen O, Wall JBJ, Zheng M, Zhou Y, Wang L, Vaseghi HR, Qian L, Liu J. Systematic comparison of 2A peptides for cloning multi-genes in a polycistronic vector. Sci Rep 2017; 7:2193. [PMID: 28526819 PMCID: PMC5438344 DOI: 10.1038/s41598-017-02460-2] [Citation(s) in RCA: 438] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 04/12/2017] [Indexed: 11/09/2022] Open
Abstract
Cloning of multiple genes in a single vector has greatly facilitated both basic and translational studies that require co-expression of multiple factors or multi-units of complex protein. Many strategies have been adopted, among which 2A “self-cleaving” peptides have garnered increased interest for their polycistronic nature, small size and high “cleavage” efficiency. However, broad application of 2 A peptides is limited by the lack of systematic comparison of different 2As alone or in combination. Here we characterized the effect of varying gene position and 2As on the expression of proteins encoded in bi-, tri-, or quad-cistronic constructs. Using direct cardiac reprogramming as an example, we further determined the effect of varied 2As on the efficiency of fluorescent cell labeling and cell fate conversion. We found that the expression of fluorophores decreased as it was moved towards the end of the construct while reprogramming was most efficient with the fluorophore at the second position. Moreover, quad-cistronic TPE2A constructs resulted in more efficient reprogramming than 3P2A or PTE2A constructs. We expect that the bi-, tri-, and quad-cistronic vectors constructed here and our results on protein expression ratios from different 2A constructs could serve to guide future utilization of 2A peptides in basic research and clinical applications.
Collapse
Affiliation(s)
- Ziqing Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA.,McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Olivia Chen
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA.,McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - J Blake Joseph Wall
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA.,McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Michael Zheng
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA.,McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Yang Zhou
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA.,McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Li Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA.,McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Haley Ruth Vaseghi
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA.,McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA. .,McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA. .,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA.
| | - Jiandong Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA. .,McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA. .,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
34
|
Schmitner N, Kohno K, Meyer D. ptf1a+ , ela3l- cells are developmentally maintained progenitors for exocrine regeneration following extreme loss of acinar cells in zebrafish larvae. Dis Model Mech 2017; 10:307-321. [PMID: 28138096 PMCID: PMC5374315 DOI: 10.1242/dmm.026633] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 01/23/2017] [Indexed: 12/12/2022] Open
Abstract
The exocrine pancreas displays a significant capacity for regeneration and renewal. In humans and mammalian model systems, the partial loss of exocrine tissue, such as after acute pancreatitis or partial pancreatectomy induces rapid recovery via expansion of surviving acinar cells. In mouse it was further found that an almost complete removal of acinar cells initiates regeneration from a currently not well-defined progenitor pool. Here, we used the zebrafish as an alternative model to study cellular mechanisms of exocrine regeneration following an almost complete removal of acinar cells. We introduced and validated two novel transgenic approaches for genetically encoded conditional cell ablation in the zebrafish, either by caspase-8-induced apoptosis or by rendering cells sensitive to diphtheria toxin. By using the ela3l promoter for exocrine-specific expression, we show that both approaches allowed cell-type-specific removal of >95% of acinar tissue in larval and adult zebrafish without causing any signs of unspecific side effects. We find that zebrafish larvae are able to recover from a virtually complete acinar tissue ablation within 2 weeks. Using short-term lineage-tracing experiments and EdU incorporation assays, we exclude duct-associated Notch-responsive cells as the source of regeneration. Rather, a rare population of slowly dividing ela3l-negative cells expressing ptf1a and CPA was identified as the origin of the newly forming exocrine cells. Cells are actively maintained, as revealed by a constant number of these cells at different larval stages and after repeated cell ablation. These cells establish ela3l expression about 4-6 days after ablation without signs of increased proliferation in between. With onset of ela3l expression, cells initiate rapid proliferation, leading to fast expansion of the ela3l-positive population. Finally, we show that this proliferation is blocked by overexpression of the Wnt-signaling antagonist dkk1b In conclusion, we show a conserved requirement for Wnt signaling in exocrine tissue expansion and reveal a potential novel progenitor or stem cell population as a source for exocrine neogenesis after complete loss of acinar cells.
Collapse
Affiliation(s)
- Nicole Schmitner
- Institute for Molecular Biology, CMBI, University of Innsbruck, 6020 Innsbruck Austria
| | - Kenji Kohno
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0101, Japan
| | - Dirk Meyer
- Institute for Molecular Biology, CMBI, University of Innsbruck, 6020 Innsbruck Austria
| |
Collapse
|
35
|
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
|
36
|
Abstract
The zebrafish pancreas shares its basic organization and cell types with the mammalian pancreas. In addition, the developmental pathways that lead to the establishment of the pancreatic islets of Langherhans are generally conserved from fish to mammals. Zebrafish provides a powerful tool to probe the mechanisms controlling establishment of the pancreatic endocrine cell types from early embryonic progenitor cells, as well as the regeneration of endocrine cells after damage. This knowledge is, in turn, applicable to refining protocols to generate renewable sources of human pancreatic islet cells that are critical for regulation of blood sugar levels. Here, we review how previous and ongoing studies in zebrafish and beyond are influencing the understanding of molecular mechanisms underlying various forms of diabetes and efforts to develop cell-based approaches to cure this increasingly widespread disease.
Collapse
|
37
|
Hill JH, Franzosa EA, Huttenhower C, Guillemin K. A conserved bacterial protein induces pancreatic beta cell expansion during zebrafish development. eLife 2016; 5:e20145. [PMID: 27960075 PMCID: PMC5154760 DOI: 10.7554/elife.20145] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 11/03/2016] [Indexed: 12/13/2022] Open
Abstract
Resident microbes play important roles in the development of the gastrointestinal tract, but their influence on other digestive organs is less well explored. Using the gnotobiotic zebrafish, we discovered that the normal expansion of the pancreatic β cell population during early larval development requires the intestinal microbiota and that specific bacterial members can restore normal β cell numbers. These bacteria share a gene that encodes a previously undescribed protein, named herein BefA (β Cell Expansion Factor A), which is sufficient to induce β cell proliferation in developing zebrafish larvae. Homologs of BefA are present in several human-associated bacterial species, and we show that they have conserved capacity to stimulate β cell proliferation in larval zebrafish. Our findings highlight a role for the microbiota in early pancreatic β cell development and suggest a possible basis for the association between low diversity childhood fecal microbiota and increased diabetes risk.
Collapse
Affiliation(s)
| | - Eric A Franzosa
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, United States
- The Broad Institute, Cambridge, United States
| | - Curtis Huttenhower
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, United States
- The Broad Institute, Cambridge, United States
| | - Karen Guillemin
- Institute of Molecular Biology, University of Oregon, Eugene, United States
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, Canada
| |
Collapse
|
38
|
Galant S, Furlan G, Coolen M, Dirian L, Foucher I, Bally-Cuif L. Embryonic origin and lineage hierarchies of the neural progenitor subtypes building the zebrafish adult midbrain. Dev Biol 2016; 420:120-135. [PMID: 27693369 PMCID: PMC5156517 DOI: 10.1016/j.ydbio.2016.09.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 08/31/2016] [Accepted: 09/26/2016] [Indexed: 01/11/2023]
Abstract
Neurogenesis in the post-embryonic vertebrate brain varies in extent and efficiency between species and brain territories. Distinct neurogenesis modes may account for this diversity, and several neural progenitor subtypes, radial glial cells (RG) and neuroepithelial progenitors (NE), have been identified in the adult zebrafish brain. The neurogenic sequences issued from these progenitors, and their contribution to brain construction, remain incompletely understood. Here we use genetic tracing techniques based on conditional Cre recombination and Tet-On neuronal birthdating to unravel the neurogenic sequence operating from NE progenitors in the zebrafish post-embryonic optic tectum. We reveal that a subpopulation of her5-positive NE cells of the posterior midbrain layer stands at the top of a neurogenic hierarchy involving, in order, the amplification pool of the tectal proliferation zone (TPZ), followed by her4-positive RG cells with transient neurogenic activity. We further demonstrate that the adult her5-positive NE pool is issued in lineage from an identically located NE pool expressing the same gene in the embryonic neural tube. Finally, we show that these features are reminiscent of the neurogenic sequence and embryonic origin of the her9-positive progenitor NE pool involved in the construction of the lateral pallium at post-embryonic stages. Together, our results highlight the shared recruitment of an identical neurogenic strategy by two remote brain territories, where long-lasting NE pools serve both as a growth zone and as the life-long source of young neurogenic RG cells. Zebrafish post-embryonic tectal neurogenesis is driven by neuroepithelial progenitors. The neuroepithelial progenitor pool is long-lasting and expresses Her5 life long. Tectal radial glia originate from the her5-positive pool and are transiently neurogenic. The post-embryonic neurogenic sequences of the tectum and lateral pallium are similar.
Collapse
Affiliation(s)
- Sonya Galant
- Paris-Saclay Institute for Neuroscience, CNRS UMR9197 - Université Paris-Sud, Team Zebrafish Neurogenetics, Avenue de la Terrasse, Bldg 5, F-91198 Gif-sur-Yvette, France
| | - Giacomo Furlan
- Paris-Saclay Institute for Neuroscience, CNRS UMR9197 - Université Paris-Sud, Team Zebrafish Neurogenetics, Avenue de la Terrasse, Bldg 5, F-91198 Gif-sur-Yvette, France
| | - Marion Coolen
- Paris-Saclay Institute for Neuroscience, CNRS UMR9197 - Université Paris-Sud, Team Zebrafish Neurogenetics, Avenue de la Terrasse, Bldg 5, F-91198 Gif-sur-Yvette, France; Department of Developmental and Stem Cell Biology and CNRS UMR 3738, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France
| | - Lara Dirian
- Paris-Saclay Institute for Neuroscience, CNRS UMR9197 - Université Paris-Sud, Team Zebrafish Neurogenetics, Avenue de la Terrasse, Bldg 5, F-91198 Gif-sur-Yvette, France
| | - Isabelle Foucher
- Paris-Saclay Institute for Neuroscience, CNRS UMR9197 - Université Paris-Sud, Team Zebrafish Neurogenetics, Avenue de la Terrasse, Bldg 5, F-91198 Gif-sur-Yvette, France; Department of Developmental and Stem Cell Biology and CNRS UMR 3738, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France.
| | - Laure Bally-Cuif
- Paris-Saclay Institute for Neuroscience, CNRS UMR9197 - Université Paris-Sud, Team Zebrafish Neurogenetics, Avenue de la Terrasse, Bldg 5, F-91198 Gif-sur-Yvette, France; Department of Developmental and Stem Cell Biology and CNRS UMR 3738, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France.
| |
Collapse
|
39
|
Sant KE, Jacobs HM, Xu J, Borofski KA, Moss LG, Moss JB, Timme-Laragy AR. Assessment of Toxicological Perturbations and Variants of Pancreatic Islet Development in the Zebrafish Model. TOXICS 2016; 4. [PMID: 28393070 PMCID: PMC5380372 DOI: 10.3390/toxics4030020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The pancreatic islets, largely comprised of insulin-producing beta cells, play a critical role in endocrine signaling and glucose homeostasis. Because they have low levels of antioxidant defenses and a high perfusion rate, the endocrine islets may be a highly susceptible target tissue of chemical exposures. However, this endpoint, as well as the integrity of the surrounding exocrine pancreas, is often overlooked in studies of developmental toxicology. Disruption of development by toxicants can alter cell fate and migration, resulting in structural alterations that are difficult to detect in mammalian embryo systems, but that are easily observed in the zebrafish embryo model (Danio rerio). Using endogenously expressed fluorescent protein markers for developing zebrafish beta cells and exocrine pancreas tissue, we documented differences in islet area and incidence rates of islet morphological variants in zebrafish embryos between 48 and 96 h post fertilization (hpf), raised under control conditions commonly used in embryotoxicity assays. We identified critical windows for chemical exposures during which increased incidences of endocrine pancreas abnormalities were observed following exposure to cyclopamine (2–12 hpf), Mono-2-ethylhexyl phthalate (MEHP) (3–48 hpf), and Perfluorooctanesulfonic acid (PFOS) (3–48 hpf). Both islet area and length of the exocrine pancreas were sensitive to oxidative stress from exposure to the oxidant tert-butyl hydroperoxide during a highly proliferative critical window (72 hpf). Finally, pancreatic dysmorphogenesis following developmental exposures is discussed with respect to human disease.
Collapse
Affiliation(s)
- Karilyn E. Sant
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA; (K.E.S.); (H.M.J.); (J.X.); (K.A.B.)
| | - Haydee M. Jacobs
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA; (K.E.S.); (H.M.J.); (J.X.); (K.A.B.)
| | - Jiali Xu
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA; (K.E.S.); (H.M.J.); (J.X.); (K.A.B.)
| | - Katrina A. Borofski
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA; (K.E.S.); (H.M.J.); (J.X.); (K.A.B.)
| | - Larry G. Moss
- Duke Molecular Physiology Institute, Endocrine Division, Duke University Medical Center, Durham, NC 27701, USA; (L.G.M.); (J.B.M.)
| | - Jennifer B. Moss
- Duke Molecular Physiology Institute, Endocrine Division, Duke University Medical Center, Durham, NC 27701, USA; (L.G.M.); (J.B.M.)
| | - Alicia R. Timme-Laragy
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA; (K.E.S.); (H.M.J.); (J.X.); (K.A.B.)
- Correspondence: ; Tel.: +1-413-545-7423
| |
Collapse
|
40
|
Huang W, Beer RL, Delaspre F, Wang G, Edelman HE, Park H, Azuma M, Parsons MJ. Sox9b is a mediator of retinoic acid signaling restricting endocrine progenitor differentiation. Dev Biol 2016; 418:28-39. [PMID: 27565026 DOI: 10.1016/j.ydbio.2016.08.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 08/16/2016] [Accepted: 08/17/2016] [Indexed: 10/21/2022]
Abstract
Centroacinar cells (CACs) are ductal Notch-responsive progenitors that in the larval zebrafish pancreas differentiate to form new islets and ultimately contribute to the majority of the adult endocrine mass. Uncovering the mechanisms regulating CAC differentiation will facilitate understanding how insulin-producing β cells are formed. Previously we reported retinoic acid (RA) signaling and Notch signaling both regulate larval CAC differentiation, suggesting a shared downstream intermediate. Sox9b is a transcription factor important for islet formation whose expression is upregulated by Notch signaling in larval CACs. Here we report that sox9b expression in larval CACs is also regulated by RA signaling. Therefore, we hypothesized that Sox9b is an intermediate between both RA- and Notch-signaling pathways. In order to study the role of Sox9b in larval CACs, we generated two cre/lox based transgenic tools, which allowed us to express full-length or truncated Sox9b in larval CACs. In this way we were able to perform spatiotemporal-controlled Sox9b gain- and loss-of-function studies and observe the subsequent effect on progenitor differentiation. Our results are consistent with Sox9b regulating CAC differentiation by being a downstream intermediate of both RA- and Notch-signaling pathways. We also demonstrate that adult zebrafish with only one functional allele of sox9b undergo accelerated β-cell regeneration, an observation consistent with sox9b regulating CAC differentiation in adults.
Collapse
Affiliation(s)
- Wei Huang
- McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University School of Medicine, 733 N. Broadway, 470 Miller Research Building, Baltimore, MD 21205, USA
| | - Rebecca L Beer
- McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University School of Medicine, 733 N. Broadway, 470 Miller Research Building, Baltimore, MD 21205, USA
| | - Fabien Delaspre
- McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University School of Medicine, 733 N. Broadway, 470 Miller Research Building, Baltimore, MD 21205, USA
| | - Guangliang Wang
- McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University School of Medicine, 733 N. Broadway, 470 Miller Research Building, Baltimore, MD 21205, USA
| | - Hannah E Edelman
- McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University School of Medicine, 733 N. Broadway, 470 Miller Research Building, Baltimore, MD 21205, USA
| | - Hyewon Park
- Department of Molecular Biosciences, University of Kansas, 1200 Sunnyside Avenue, Lawrence, KS 66045, USA
| | - Mizuki Azuma
- Department of Molecular Biosciences, University of Kansas, 1200 Sunnyside Avenue, Lawrence, KS 66045, USA
| | - Michael J Parsons
- McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University School of Medicine, 733 N. Broadway, 470 Miller Research Building, Baltimore, MD 21205, USA; Department of Surgery, Johns Hopkins University School of Medicine, 733 N. Broadway, 470 Miller Research Building, Baltimore, MD 21205, USA
| |
Collapse
|
41
|
Hanovice NJ, McMains E, Gross JM. A GAL4-inducible transgenic tool kit for the in vivo modulation of Rho GTPase activity in zebrafish. Dev Dyn 2016; 245:844-53. [PMID: 27105927 PMCID: PMC4946996 DOI: 10.1002/dvdy.24412] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 03/21/2016] [Accepted: 03/29/2016] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Rho GTPases are small monomeric G-proteins that play key roles in many cellular processes. Due to Rho GTPases' widespread expression and broad functions, analyses of their function during late development require tissue-specific modulation of activity. The GAL4/UAS system provides an excellent tool for investigating the function of Rho GTPases in vivo. With this in mind, we created a transgenic tool kit enabling spatial and temporal modulation of Rho GTPase activity in zebrafish. RESULTS Transgenic constructs were assembled driving dominant-negative, constitutively active, and wild-type versions of Cdc42, RhoA, and Rac1 under 10XUAS control. The self-cleaving viral peptide F2A was utilized to allow bicistronic expression of a fluorescent reporter and Rho GTPase. Global heat shock of hsp70l:gal4(+) transgenic embryos confirmed GAL4-specific construct expression. Western blot analysis indicated myc-tagged Rho GTPases were expressed only in the presence of GAL4. Construct expression was confined to proper cells when combined with pou4f3:gal4 or ptf1a:gal4. Finally, transgene expression resulted in reproducible defects in lens formation, indicating that the transgenes are functional in vivo. CONCLUSIONS We generated and validated 10 transgenic lines, creating a versatile tool kit for the temporal-spatial modulation of Cdc42, RhoA, and Rac1 activity in vivo. These lines will enable systematic analysis of Rho GTPase function in any tissue of interest. Developmental Dynamics 245:844-853, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nicholas J Hanovice
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Emily McMains
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas
| | - Jeffrey M Gross
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
42
|
Dash SN, Hakonen E, Ustinov J, Otonkoski T, Andersson O, Lehtonen S. sept7b is required for the differentiation of pancreatic endocrine progenitors. Sci Rep 2016; 6:24992. [PMID: 27114183 PMCID: PMC4845001 DOI: 10.1038/srep24992] [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: 12/23/2015] [Accepted: 04/05/2016] [Indexed: 12/14/2022] Open
Abstract
Protection or restoration of pancreatic β-cell mass as a therapeutic treatment for type 1 diabetes requires understanding of the mechanisms that drive the specification and development of pancreatic endocrine cells. Septins are filamentous small GTPases that function in the regulation of cell division, cytoskeletal organization and membrane remodeling, and are involved in various tissue-specific developmental processes. However, their role in pancreatic endocrine cell differentiation remains unknown. Here we show by functional manipulation techniques in transgenic zebrafish lines that suppression of sept7b, the zebrafish ortholog of human SEPT7, profoundly increases the number of endocrine progenitors but limits their differentiation, leading to reduction in β- and α-cell mass. Furthermore, we discovered that shh (sonic hedgehog) expression in the endoderm, essential for the development of pancreatic progenitors of the dorsal pancreatic bud, is absent in larvae depleted of sept7b. We also discovered that sept7b is important for the differentiation of ventral pancreatic bud-derived cells: sept7b-depleted larvae exhibit downregulation of Notch receptors notch1a and notch1b and show precocious differentiation of NeuroD-positive endocrine cells in the intrapancreatic duct and gut epithelium. Collectively, this study provides a novel insight into the development of pancreatic endocrine progenitors, revealing an essential role for sept7b in endocrine progenitor differentiation.
Collapse
Affiliation(s)
| | - Elina Hakonen
- Research Program for Molecular Neurology and Biomedicum Stem Cell Center, University of Helsinki, Helsinki, Finland
| | - Jarkko Ustinov
- Research Program for Molecular Neurology and Biomedicum Stem Cell Center, University of Helsinki, Helsinki, Finland
| | - Timo Otonkoski
- Research Program for Molecular Neurology and Biomedicum Stem Cell Center, University of Helsinki, Helsinki, Finland
| | - Olov Andersson
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Sanna Lehtonen
- Department of Pathology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
43
|
Abstract
The zebrafish model is the only available high-throughput vertebrate assessment system, and it is uniquely suited for studies of in vivo cell biology. A sequenced and annotated genome has revealed a large degree of evolutionary conservation in comparison to the human genome. Due to our shared evolutionary history, the anatomical and physiological features of fish are highly homologous to humans, which facilitates studies relevant to human health. In addition, zebrafish provide a very unique vertebrate data stream that allows researchers to anchor hypotheses at the biochemical, genetic, and cellular levels to observations at the structural, functional, and behavioral level in a high-throughput format. In this review, we will draw heavily from toxicological studies to highlight advances in zebrafish high-throughput systems. Breakthroughs in transgenic/reporter lines and methods for genetic manipulation, such as the CRISPR-Cas9 system, will be comprised of reports across diverse disciplines.
Collapse
Affiliation(s)
- Gloria R Garcia
- Oregon State University, Department of Environmental and Molecular Toxicology, Environmental Health Sciences Center, Corvallis, OR 97331, USA
| | - Pamela D Noyes
- Oregon State University, Department of Environmental and Molecular Toxicology, Environmental Health Sciences Center, Corvallis, OR 97331, USA
| | - Robert L Tanguay
- Oregon State University, Department of Environmental and Molecular Toxicology, Environmental Health Sciences Center, Corvallis, OR 97331, USA.
| |
Collapse
|
44
|
Beer RL, Parsons MJ, Rovira M. Centroacinar cells: At the center of pancreas regeneration. Dev Biol 2016; 413:8-15. [PMID: 26963675 DOI: 10.1016/j.ydbio.2016.02.027] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/23/2016] [Accepted: 02/26/2016] [Indexed: 10/22/2022]
Abstract
The process of regeneration serves to heal injury by replacing missing cells. Understanding regeneration can help us replace cell populations lost during disease, such as the insulin-producing β cells lost in diabetic patients. Centroacinar cells (CACs) are a specialized ductal pancreatic cell type that act as progenitors to replace β cells in the zebrafish. However, whether CACs contribute to β-cell regeneration in adult mammals remains controversial. Here we review the current understanding of the role of CACs as endocrine progenitors during regeneration in zebrafish and mammals.
Collapse
Affiliation(s)
- Rebecca L Beer
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD, United States.
| | - Michael J Parsons
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD, United States; Department of Surgery, Johns Hopkins University, Baltimore, MD, United States
| | - Meritxell Rovira
- Genomic Programming of Beta-Cells Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain.
| |
Collapse
|
45
|
Li M, Page-McCaw P, Chen W. FGF1 Mediates Overnutrition-Induced Compensatory β-Cell Differentiation. Diabetes 2016; 65:96-109. [PMID: 26420862 PMCID: PMC4686947 DOI: 10.2337/db15-0085] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 09/22/2015] [Indexed: 12/17/2022]
Abstract
Increased insulin demand resulting from insulin resistance and/or overnutrition induces a compensatory increase in β-cell mass. The physiological factors responsible for the compensation have not been fully characterized. In zebrafish, overnutrition rapidly induces compensatory β-cell differentiation through triggering the release of a paracrine signal from persistently activated β-cells. We identified Fgf1 signaling as a key component of the overnutrition-induced β-cell differentiation signal in a small molecule screen. Fgf1 was confirmed as the overnutrition-induced β-cell differentiation signal, as inactivation of fgf1 abolished the compensatory β-cell differentiation. Furthermore, expression of human FGF1 solely in β-cells in fgf1(-/-) animals rescued the compensatory response, indicating that β-cells can be the source of FGF1. Additionally, constitutive secretion of FGF1 with an exogenous signal peptide increased β-cell number in the absence of overnutrition. These results demonstrate that fgf1 is necessary and FGF1 expression in β-cells is sufficient for the compensatory β-cell differentiation. We further show that FGF1 is secreted during prolonged activation of cultured mammalian β-cells and that endoplasmic reticulum stress acts upstream of FGF1 release. Thus, the recently discovered antidiabetes function of FGF1 may act partially through increasing β-cell differentiation.
Collapse
Affiliation(s)
- Mingyu Li
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Patrick Page-McCaw
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Wenbiao Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
46
|
|
47
|
Sparling DP, Yu J, Kim K, Zhu C, Brachs S, Birkenfeld AL, Pajvani UB. Adipocyte-specific blockade of gamma-secretase, but not inhibition of Notch activity, reduces adipose insulin sensitivity. Mol Metab 2015; 5:113-121. [PMID: 26909319 PMCID: PMC4735659 DOI: 10.1016/j.molmet.2015.11.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 11/18/2015] [Accepted: 11/25/2015] [Indexed: 01/12/2023] Open
Abstract
Objective As the obesity pandemic continues to expand, novel molecular targets to reduce obesity-related insulin resistance and Type 2 Diabetes (T2D) continue to be needed. We have recently shown that obesity is associated with reactivated liver Notch signaling, which, in turn, increases hepatic insulin resistance, opening up therapeutic avenues for Notch inhibitors to be repurposed for T2D. Herein, we tested the systemic effects of γ-secretase inhibitors (GSIs), which prevent endogenous Notch activation, and confirmed these effects through creation and characterization of two different adipocyte-specific Notch loss-of-function mouse models through genetic ablation of the Notch transcriptional effector Rbp-Jk (A-Rbpj) and the obligate γ-secretase component Nicastrin (A-Nicastrin). Methods Glucose homeostasis and both local adipose and systemic insulin sensitivity were examined in GSI-treated, A-Rbpj and A-Nicastrin mice, as well as vehicle-treated or control littermates, with complementary in vitro studies in primary hepatocytes and 3T3-L1 adipocytes. Results GSI-treatment increases hepatic insulin sensitivity in obese mice but leads to reciprocal lowering of adipose glucose disposal. While A-Rbpj mice show normal body weight, adipose development and mass and unchanged adipose insulin sensitivity as control littermates, A-Nicastrin mice are relatively insulin-resistant, mirroring the GSI effect on adipose insulin action. Conclusions Notch signaling is dispensable for normal adipocyte function, but adipocyte-specific γ-secretase blockade reduces adipose insulin sensitivity, suggesting that specific Notch inhibitors would be preferable to GSIs for application in T2D. γ-secretase inhibitors (GSIs) are non-specific inhibitors of Notch signaling. GSI-treatment of obese mice increases hepatic, but lowers adipose insulin sensitivity. Adipocyte-specific Notch inhibition does not affect adipose mass or glucose homeostasis. Adipocyte-specific γ-secretase blockade reduces adipose insulin sensitivity. Specific Notch inhibitors may be preferable to GSIs for treatment of Type 2 Diabetes.
Collapse
Affiliation(s)
- David P Sparling
- Departments of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Junjie Yu
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - KyeongJin Kim
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Changyu Zhu
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Sebastian Brachs
- Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Charité - University School of Medicine, Berlin, Germany
| | - Andreas L Birkenfeld
- Section of Metabolic Vascular Medicine, Medical Clinic III and Paul Langerhans Institute Dresden (PLID), a member of the German Center for Diabetes Research (DZD), Technische Universität Dresden, Germany; Section of Diabetes and Nutritional Sciences, Rayne Institute, Denmark Hill Campus, King's College London, UK
| | - Utpal B Pajvani
- Department of Medicine, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
48
|
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
|
49
|
Bielczyk-Maczyńska E, Lam Hung L, Ferreira L, Fleischmann T, Weis F, Fernández-Pevida A, Harvey SA, Wali N, Warren AJ, Barroso I, Stemple DL, Cvejic A. The Ribosome Biogenesis Protein Nol9 Is Essential for Definitive Hematopoiesis and Pancreas Morphogenesis in Zebrafish. PLoS Genet 2015; 11:e1005677. [PMID: 26624285 PMCID: PMC4666468 DOI: 10.1371/journal.pgen.1005677] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 10/26/2015] [Indexed: 12/27/2022] Open
Abstract
Ribosome biogenesis is a ubiquitous and essential process in cells. Defects in ribosome biogenesis and function result in a group of human disorders, collectively known as ribosomopathies. In this study, we describe a zebrafish mutant with a loss-of-function mutation in nol9, a gene that encodes a non-ribosomal protein involved in rRNA processing. nol9sa1022/sa1022 mutants have a defect in 28S rRNA processing. The nol9sa1022/sa1022 larvae display hypoplastic pancreas, liver and intestine and have decreased numbers of hematopoietic stem and progenitor cells (HSPCs), as well as definitive erythrocytes and lymphocytes. In addition, ultrastructural analysis revealed signs of pathological processes occurring in endothelial cells of the caudal vein, emphasizing the complexity of the phenotype observed in nol9sa1022/sa1022 larvae. We further show that both the pancreatic and hematopoietic deficiencies in nol9sa1022/sa1022 embryos were due to impaired cell proliferation of respective progenitor cells. Interestingly, genetic loss of Tp53 rescued the HSPCs but not the pancreatic defects. In contrast, activation of mRNA translation via the mTOR pathway by L-Leucine treatment did not revert the erythroid or pancreatic defects. Together, we present the nol9sa1022/sa1022 mutant, a novel zebrafish ribosomopathy model, which recapitulates key human disease characteristics. The use of this genetically tractable model will enhance our understanding of the tissue-specific mechanisms following impaired ribosome biogenesis in the context of an intact vertebrate.
Collapse
Affiliation(s)
- Ewa Bielczyk-Maczyńska
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge, United Kingdom
- NHS Blood and Transplant, Cambridge, United Kingdom
| | - Laure Lam Hung
- Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Lauren Ferreira
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Tobias Fleischmann
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Cambridge Institute for Medical Research, Cambridge, United Kingdom
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Félix Weis
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Cambridge Institute for Medical Research, Cambridge, United Kingdom
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Antonio Fernández-Pevida
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Cambridge Institute for Medical Research, Cambridge, United Kingdom
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Steven A. Harvey
- Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Neha Wali
- Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Alan J. Warren
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Cambridge Institute for Medical Research, Cambridge, United Kingdom
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Inês Barroso
- Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Derek L. Stemple
- Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Ana Cvejic
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge, United Kingdom
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
50
|
Delaspre F, Beer RL, Rovira M, Huang W, Wang G, Gee S, Vitery MDC, Wheelan SJ, Parsons MJ. Centroacinar Cells Are Progenitors That Contribute to Endocrine Pancreas Regeneration. Diabetes 2015; 64:3499-509. [PMID: 26153247 PMCID: PMC4587647 DOI: 10.2337/db15-0153] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 06/24/2015] [Indexed: 12/17/2022]
Abstract
Diabetes is associated with a paucity of insulin-producing β-cells. With the goal of finding therapeutic routes to treat diabetes, we aim to find molecular and cellular mechanisms involved in β-cell neogenesis and regeneration. To facilitate discovery of such mechanisms, we use a vertebrate organism where pancreatic cells readily regenerate. The larval zebrafish pancreas contains Notch-responsive progenitors that during development give rise to adult ductal, endocrine, and centroacinar cells (CACs). Adult CACs are also Notch responsive and are morphologically similar to their larval predecessors. To test our hypothesis that adult CACs are also progenitors, we took two complementary approaches: 1) We established the transcriptome for adult CACs. Using gene ontology, transgenic lines, and in situ hybridization, we found that the CAC transcriptome is enriched for progenitor markers. 2) Using lineage tracing, we demonstrated that CACs do form new endocrine cells after β-cell ablation or partial pancreatectomy. We concluded that CACs and their larval predecessors are the same cell type and represent an opportune model to study both β-cell neogenesis and β-cell regeneration. Furthermore, we show that in cftr loss-of-function mutants, there is a deficiency of larval CACs, providing a possible explanation for pancreatic complications associated with cystic fibrosis.
Collapse
Affiliation(s)
- Fabien Delaspre
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD
| | - Rebecca L Beer
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD
| | - Meritxell Rovira
- Genomic Programming of Beta-Cells Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Wei Huang
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD
| | - Guangliang Wang
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD
| | - Stephen Gee
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD
| | | | - Sarah J Wheelan
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD Department of Oncology, Johns Hopkins University, Baltimore, MD
| | - Michael J Parsons
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD Department of Surgery, Johns Hopkins University, Baltimore, MD
| |
Collapse
|