1
|
Huang P, Yang H, Kuang H, Yang J, Duan X, Bian H, Wang X. Pancreaticobiliary Maljunction: A Multidimensional Exploration of Pathophysiology, Diagnosis, Classification, Management and Research Prospects. Dig Dis Sci 2025:10.1007/s10620-025-09057-0. [PMID: 40252147 DOI: 10.1007/s10620-025-09057-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/09/2025] [Indexed: 04/21/2025]
Abstract
Pancreaticobiliary maljunction is a congenital malformation in which the pancreatic and bile ducts join anatomically outside the duodenal wall, usually forming a markedly long common channel, which can cause reciprocal reflux between pancreatic juice and bile. Cholangiography, endoscopic ultrasonography, surgery, and autopsy can be used to diagnose pancreaticobiliary maljunction. Elevated amylase levels in bile and extrahepatic bile duct dilatation strongly suggest the existence of pancreaticobiliary maljunction. The regurgitation may lead to the development of various hepatobiliary and pancreatic disorders such as pancreatitis and biliary carcinoma. The pathogenesis of pancreaticobiliary maljunction is the result of a series of pathophysiological changes caused by reflux. Surgery is recommended for patients diagnosed with pancreaticobiliary maljunction irrespective of the presence or absence of symptoms because of its high biliary carcinogenicity, but the treatment strategy is quite different between adult patients with and without biliary dilatation.
Collapse
Affiliation(s)
- Peng Huang
- Department of General Surgery, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, Hubei, China
| | - Hu Yang
- Department of General Surgery, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, Hubei, China
| | - Houfang Kuang
- Department of General Surgery, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, Hubei, China
| | - Jun Yang
- Department of General Surgery, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, Hubei, China
| | - Xufei Duan
- Department of General Surgery, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, Hubei, China
| | - Hongqiang Bian
- Department of General Surgery, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, Hubei, China
| | - Xin Wang
- Department of General Surgery, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, Hubei, China.
| |
Collapse
|
2
|
Vásárhelyi E, Rácz G, Urbányi B, Szabó BP, Szepesi-Bencsik D, Szabó I, Bock I, Volner C, Griffitts JD, Kriszt B, Bakos K, Csenki Z. The acute and sub-chronic toxicological effects of 3-amino-9-ethylcarbazole (AEC) on zebrafish. Hum Exp Toxicol 2025; 44:9603271251318968. [PMID: 39982205 DOI: 10.1177/09603271251318968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
INTRODUCTION In this study, we sought to determine the sub-chronic toxicological effects of AEC on zebrafish embryos. METHODS We utilized fish early life stage (FELS) and fish embryo toxicity (FET) tests, vascular, neurological, and renal transgenic zebrafish lines, and gene expression anal-ysis of the zebrafish tissue. RESULTS In the FET tests, AEC caused several abnormalities in the larvae, with the LC50 at 24 hpf being 4.076 ± 0.221 mg/L and 3.296 ± 0.127 mg/L at 96 hpf. In the FELS test, AEC was shown to be lethal following 16 days of exposure at 0.5 mg/L, 1 mg/L and 2 mg/L. Some of the transgenic zebrafish lines exhibited slight changes in fluorescent signaling pat-terns after exposure to AEC at 1 mg/L and 2 mg/L. Notable results of the gene expression analysis revealed: gpx4b and got2 were downregulated in the liver; HIF1a was downregulated at 0.25 mg/L and 0.5 mg/L concentrations, NOTCH1a and fli-1 genes were downregulated at all concentrations, and A2b was upregulated in the vasculature; a1T, ngn1, elavl3, syn2a, mbp, gap43 were down-regulated in the nervous system; and wt1b was downregulated in the kidney. DISCUCCION Altogether, the results of our study indicate the potential for AEC to cause harm to organisms.
Collapse
Affiliation(s)
- Erna Vásárhelyi
- Department of Environmental Toxicology, Institute of Aquaculture and Environmental Safety, Faculty of Agricultural and Environmental Sciences, Szent István University, Gödöllő, Hungary
| | - Gergely Rácz
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Béla Urbányi
- Department of Aquaculture, Institute of Aquaculture and Environmental Safety, Faculty of Agricultural and Environmental Sciences, Szent István University, Gödöllő, Hungary
| | - Balázs P Szabó
- Department of Food Engineering, Faculty of Engineering, University of Szeged, Szeged, Hungary
| | - Dóra Szepesi-Bencsik
- Department of Food Engineering, Faculty of Engineering, University of Szeged, Szeged, Hungary
| | - István Szabó
- Department of Environmental Toxicology, Institute of Aquaculture and Environmental Safety, Faculty of Agricultural and Environmental Sciences, Szent István University, Gödöllő, Hungary
| | - Illés Bock
- Department of Environmental Toxicology, Institute of Aquaculture and Environmental Safety, Faculty of Agricultural and Environmental Sciences, Szent István University, Gödöllő, Hungary
| | - Cintia Volner
- Department of Environmental Toxicology, Institute of Aquaculture and Environmental Safety, Faculty of Agricultural and Environmental Sciences, Szent István University, Gödöllő, Hungary
| | - Jeffrey Daniel Griffitts
- Department of Environmental Toxicology, Institute of Aquaculture and Environmental Safety, Faculty of Agricultural and Environmental Sciences, Szent István University, Gödöllő, Hungary
| | - Balázs Kriszt
- Department of Environmental Safety, Institute of Aquaculture and Environmental Safety, Faculty of Agricultural and Environmental Sciences, Szent István University, Gödöllő, Hungary
| | - Katalin Bakos
- Department of Environmental Toxicology, Institute of Aquaculture and Environmental Safety, Faculty of Agricultural and Environmental Sciences, Szent István University, Gödöllő, Hungary
- Premonstratensian St Norbert High School, Gödöllő, Hungary
| | - Zsolt Csenki
- Department of Environmental Toxicology, Institute of Aquaculture and Environmental Safety, Faculty of Agricultural and Environmental Sciences, Szent István University, Gödöllő, Hungary
| |
Collapse
|
3
|
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
|
4
|
Rovegno E, Lucon-Xiccato T, Terrin F, Valle LD, Bertolucci C. Knockout in zebrafish reveals the role of the glucocorticoid receptor in shaping behavioral syndromes. Behav Brain Res 2024; 473:115179. [PMID: 39103124 DOI: 10.1016/j.bbr.2024.115179] [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: 05/13/2024] [Revised: 07/03/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
Glucocorticoids (GCs) have a wide spectrum of effects on animal behavior. A recently suggested effect involves determining the structure of individual differences, that is how the behavioral traits of an individual covary, forming the so-called behavioral syndromes. As GCs can exert their action in multiple ways, e.g., via rapid non-genomic effects or via the activation of two highly homologous members of the steroid receptor family acting as transcription factors, it is unclear how the GC modulation of behavioral syndromes takes place. We exploited a zebrafish line with a frameshift mutation in the gene encoding the GC receptor (Gr), to investigate this question. We found that lack of Gr altered the average score of several behavioral traits in the mutant line, determining reduced boldness, and increased activity and sociability. Critically, the pattern of covariation between these traits was also substantially affected by the loss of Gr. The most evident effect was an association of traits involved in boldness in the gr mutant line. This study reveals that, in zebrafish, Gr is not only involved in the modulation of the average value of behavioral traits, but also in how the behavioral traits of an individual are interrelated and determine the behavioral syndromes.
Collapse
Affiliation(s)
- Eleonora Rovegno
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.
| | - Tyrone Lucon-Xiccato
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | | | | | - Cristiano Bertolucci
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| |
Collapse
|
5
|
He X, Xiong D, Zhao L, Fu J, Luo L. Meningeal lymphatic supporting cells govern the formation and maintenance of zebrafish mural lymphatic endothelial cells. Nat Commun 2024; 15:5547. [PMID: 38956047 PMCID: PMC11220022 DOI: 10.1038/s41467-024-49818-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 06/18/2024] [Indexed: 07/04/2024] Open
Abstract
The meninges are critical for the brain functions, but the diversity of meningeal cell types and intercellular interactions have yet to be thoroughly examined. Here we identify a population of meningeal lymphatic supporting cells (mLSCs) in the zebrafish leptomeninges, which are specifically labeled by ependymin. Morphologically, mLSCs form membranous structures that enwrap the majority of leptomeningeal blood vessels and all the mural lymphatic endothelial cells (muLECs). Based on its unique cellular morphologies and transcriptional profile, mLSC is characterized as a unique cell type different from all the currently known meningeal cell types. Because of the formation of supportive structures and production of pro-lymphangiogenic factors, mLSCs not only promote muLEC development and maintain the dispersed distributions of muLECs in the leptomeninges, but also are required for muLEC regeneration after ablation. This study characterizes a newly identified cell type in leptomeninges, mLSC, which is required for muLEC development, maintenance, and regeneration.
Collapse
Affiliation(s)
- Xiang He
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, 400715, China
| | - Daiqin Xiong
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, 400715, China
| | - Lei Zhao
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Jialong Fu
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, 400715, China
| | - Lingfei Luo
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, 400715, China.
- School of Life Sciences, Fudan University, Yangpu, Shanghai, 200438, China.
| |
Collapse
|
6
|
Celeghin R, Risato G, Beffagna G, Cason M, Bueno Marinas M, Della Barbera M, Facchinello N, Giuliodori A, Brañas Casas R, Caichiolo M, Vettori A, Grisan E, Rizzo S, Dalla Valle L, Argenton F, Thiene G, Tiso N, Pilichou K, Basso C. A novel DSP zebrafish model reveals training- and drug-induced modulation of arrhythmogenic cardiomyopathy phenotypes. Cell Death Discov 2023; 9:441. [PMID: 38057295 DOI: 10.1038/s41420-023-01741-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 10/30/2023] [Accepted: 11/23/2023] [Indexed: 12/08/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is an inherited disorder characterized by progressive loss of the ventricular myocardium causing life-threatening ventricular arrhythmias, syncope and sudden cardiac death in young and athletes. About 40% of AC cases carry one or more mutations in genes encoding for desmosomal proteins, including Desmoplakin (Dsp). We present here the first stable Dsp knock-out (KO) zebrafish line able to model cardiac alterations and cell signalling dysregulation, characteristic of the AC disease, on which environmental factors and candidate drugs can be tested. Our stable Dsp knock-out (KO) zebrafish line was characterized by cardiac alterations, oedema and bradycardia at larval stages. Histological analysis of mutated adult hearts showed reduced contractile structures and abnormal shape of the ventricle, with thinning of the myocardial layer, vessels dilation and presence of adipocytes within the myocardium. Moreover, TEM analysis revealed "pale", disorganized and delocalized desmosomes. Intensive physical training protocol caused a global worsening of the cardiac phenotype, accelerating the progression of the disease. Of note, we detected a decrease of Wnt/β-catenin signalling, recently associated with AC pathogenesis, as well as Hippo/YAP-TAZ and TGF-β pathway dysregulation. Pharmacological treatment of mutated larvae with SB216763, a Wnt/β-catenin agonist, rescued pathway expression and cardiac abnormalities, stabilizing the heart rhythm. Overall, our Dsp KO zebrafish line recapitulates many AC features observed in human patients, pointing at zebrafish as a suitable system for in vivo analysis of environmental modulators, such as the physical exercise, and the screening of pathway-targeted drugs, especially related to the Wnt/β-catenin signalling cascade.
Collapse
Affiliation(s)
- Rudy Celeghin
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, 35128, Italy
| | - Giovanni Risato
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, 35128, Italy
- Department of Biology, University of Padova, Padova, 35131, Italy
| | - Giorgia Beffagna
- Department of Biology, University of Padova, Padova, 35131, Italy.
| | - Marco Cason
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, 35128, Italy
| | - Maria Bueno Marinas
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, 35128, Italy
| | - Mila Della Barbera
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, 35128, Italy
| | - Nicola Facchinello
- Neuroscience Institute, Italian National Research Council (CNR), Padova, 35131, Italy
| | - Alice Giuliodori
- Department of Biology, University of Padova, Padova, 35131, Italy
| | | | - Micol Caichiolo
- Department of Biology, University of Padova, Padova, 35131, Italy
| | - Andrea Vettori
- Department of Biotechnology, University of Verona, Verona, 37134, Italy
| | - Enrico Grisan
- School of Engineering, London South Bank University, London, SE1 0AA, UK
| | - Stefania Rizzo
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, 35128, Italy
| | | | | | - Gaetano Thiene
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, 35128, Italy
| | - Natascia Tiso
- Department of Biology, University of Padova, Padova, 35131, Italy.
| | - Kalliopi Pilichou
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, 35128, Italy
| | - Cristina Basso
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, 35128, Italy.
| |
Collapse
|
7
|
Canato E, Grigoletto A, Zanotto I, Tedeschini T, Campara B, Quaglio G, Toffoli G, Mandracchia D, Dinarello A, Tiso N, Argenton F, Sayaf K, Guido M, Gabbia D, De Martin S, Pasut G. Anti-HER2 Super Stealth Immunoliposomes for Targeted-Chemotherapy. Adv Healthc Mater 2023; 12:e2301650. [PMID: 37590033 PMCID: PMC11469322 DOI: 10.1002/adhm.202301650] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/04/2023] [Indexed: 08/18/2023]
Abstract
Liposomes play an important role in the field of drug delivery by virtue of their biocompatibility and versatility as carriers. Stealth liposomes, obtained by surface decoration with hydrophilic polyethylene glycol (PEG) molecules, represent an important turning point in liposome technology, leading to significant improvements in the pharmacokinetic profile compared to naked liposomes. Nevertheless, the generation of effective targeted liposomes-a central issue for cancer therapy-has faced several difficulties and clinical phase failures. Active targeting remains a challenge for liposomes. In this direction, a new Super Stealth Immunoliposomes (SSIL2) composed of a PEG-bi-phospholipids derivative is designed that stabilizes the polymer shielding over the liposomes. Furthermore, its counterpart, conjugated to the fragment antigen-binding of trastuzumab (Fab'TRZ -PEG-bi-phospholipids), is firmly anchored on the liposomes surface and correctly orients outward the targeting moiety. Throughout this study, the performances of SSIL2 are evaluated and compared to classic stealth liposomes and stealth immunoliposomes in vitro in a panel of cell lines and in vivo studies in zebrafish larvae and rodent models. Overall, SSIL2 shows superior in vitro and in vivo outcomes, both in terms of safety and anticancer efficacy, thus representing a step forward in targeted cancer therapy, and valuable for future development.
Collapse
Affiliation(s)
- Elena Canato
- Department Pharmaceutical and Pharmacological SciencesUniversity of PadovaVia Marzolo 5Padova35131Italy
| | - Antonella Grigoletto
- Department Pharmaceutical and Pharmacological SciencesUniversity of PadovaVia Marzolo 5Padova35131Italy
| | - Ilaria Zanotto
- Department Pharmaceutical and Pharmacological SciencesUniversity of PadovaVia Marzolo 5Padova35131Italy
| | - Tommaso Tedeschini
- Department Pharmaceutical and Pharmacological SciencesUniversity of PadovaVia Marzolo 5Padova35131Italy
| | - Benedetta Campara
- Department Pharmaceutical and Pharmacological SciencesUniversity of PadovaVia Marzolo 5Padova35131Italy
| | - Giovanna Quaglio
- Department Pharmaceutical and Pharmacological SciencesUniversity of PadovaVia Marzolo 5Padova35131Italy
| | - Giuseppe Toffoli
- Experimental and Clinical PharmacologyCentro di Riferimento Oncologico di Aviano (CRO) IRCCSVia Franco Gallini n. 2Aviano33081Italy
| | - Delia Mandracchia
- Department of Molecular and Translational MedicineUniversity of BresciaBrescia25123Italy
| | - Alberto Dinarello
- Department of BiologyUniversity of PadovaVia U. Bassi 58/BPadova35131Italy
| | - Natascia Tiso
- Department of BiologyUniversity of PadovaVia U. Bassi 58/BPadova35131Italy
| | - Francesco Argenton
- Department of BiologyUniversity of PadovaVia U. Bassi 58/BPadova35131Italy
| | - Katia Sayaf
- Department Surgery, Oncology and GastroenterologyUniversity of PadovaVia Giustiniani 2Padova35131Italy
| | - Maria Guido
- Department of Medicine‐DIMEDUniversity of PadovaPadua35128Italy
- Department of PathologyAzienda ULSS2 Marca TrevigianaTreviso31100Italy
| | - Daniela Gabbia
- Department Pharmaceutical and Pharmacological SciencesUniversity of PadovaVia Marzolo 5Padova35131Italy
| | - Sara De Martin
- Department Pharmaceutical and Pharmacological SciencesUniversity of PadovaVia Marzolo 5Padova35131Italy
| | - Gianfranco Pasut
- Department Pharmaceutical and Pharmacological SciencesUniversity of PadovaVia Marzolo 5Padova35131Italy
| |
Collapse
|
8
|
Massaquoi MS, Kong GL, Chilin-Fuentes D, Ngo JS, Horve PF, Melancon E, Hamilton MK, Eisen JS, Guillemin K. Cell-type-specific responses to the microbiota across all tissues of the larval zebrafish. Cell Rep 2023; 42:112095. [PMID: 36787219 PMCID: PMC10423310 DOI: 10.1016/j.celrep.2023.112095] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/22/2022] [Accepted: 01/25/2023] [Indexed: 02/15/2023] Open
Abstract
Animal development proceeds in the presence of intimate microbial associations, but the extent to which different host cells across the body respond to resident microbes remains to be fully explored. Using the vertebrate model organism, the larval zebrafish, we assessed transcriptional responses to the microbiota across the entire body at single-cell resolution. We find that cell types across the body, not limited to tissues at host-microbe interfaces, respond to the microbiota. Responses are cell-type-specific, but across many tissues the microbiota enhances cell proliferation, increases metabolism, and stimulates a diversity of cellular activities, revealing roles for the microbiota in promoting developmental plasticity. This work provides a resource for exploring transcriptional responses to the microbiota across all cell types of the vertebrate body and generating new hypotheses about the interactions between vertebrate hosts and their microbiota.
Collapse
Affiliation(s)
- Michelle S Massaquoi
- Institute of Molecular Biology, University of Oregon, 1318 Franklin Boulevard, Eugene, OR 97403, USA; Thermo Fisher Scientific, 29851 Willow Creek Road, Eugene, OR 97402, USA; Thermo Fisher Scientific, 22025 20th Avenue SE, Bothell, WA 98021, USA
| | - Garth L Kong
- Institute of Molecular Biology, University of Oregon, 1318 Franklin Boulevard, Eugene, OR 97403, USA
| | - Daisy Chilin-Fuentes
- Institute of Molecular Biology, University of Oregon, 1318 Franklin Boulevard, Eugene, OR 97403, USA
| | - Julia S Ngo
- Institute of Molecular Biology, University of Oregon, 1318 Franklin Boulevard, Eugene, OR 97403, USA
| | - Patrick F Horve
- Institute of Molecular Biology, University of Oregon, 1318 Franklin Boulevard, Eugene, OR 97403, USA
| | - Ellie Melancon
- Institute of Neuroscience, University of Oregon, 1254 University of Oregon, Eugene, OR 97403, USA
| | - M Kristina Hamilton
- Institute of Molecular Biology, University of Oregon, 1318 Franklin Boulevard, Eugene, OR 97403, USA; Institute of Neuroscience, University of Oregon, 1254 University of Oregon, Eugene, OR 97403, USA; Thermo Fisher Scientific, 29851 Willow Creek Road, Eugene, OR 97402, USA
| | - Judith S Eisen
- Institute of Molecular Biology, University of Oregon, 1318 Franklin Boulevard, Eugene, OR 97403, USA; Institute of Neuroscience, University of Oregon, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Karen Guillemin
- Institute of Molecular Biology, University of Oregon, 1318 Franklin Boulevard, Eugene, OR 97403, USA; Humans and the Microbiome Program, CIFAR, Toronto, ON M5G 1M1, Canada.
| |
Collapse
|
9
|
Hill JH, Massaquoi MS, Sweeney EG, Wall ES, Jahl P, Bell R, Kallio K, Derrick D, Murtaugh LC, Parthasarathy R, Remington SJ, Round JL, Guillemin K. BefA, a microbiota-secreted membrane disrupter, disseminates to the pancreas and increases β cell mass. Cell Metab 2022; 34:1779-1791.e9. [PMID: 36240759 PMCID: PMC9633563 DOI: 10.1016/j.cmet.2022.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/26/2022] [Accepted: 08/31/2022] [Indexed: 01/11/2023]
Abstract
Microbiome dysbiosis is a feature of diabetes, but how microbial products influence insulin production is poorly understood. We report the mechanism of BefA, a microbiome-derived protein that increases proliferation of insulin-producing β cells during development in gnotobiotic zebrafish and mice. BefA disseminates systemically by multiple anatomic routes to act directly on pancreatic islets. We detail BefA's atomic structure, containing a lipid-binding SYLF domain, and demonstrate that it permeabilizes synthetic liposomes and bacterial membranes. A BefA mutant impaired in membrane disruption fails to expand β cells, whereas the pore-forming host defense protein, Reg3, stimulates β cell proliferation. Our work demonstrates that membrane permeabilization by microbiome-derived and host defense proteins is necessary and sufficient for β cell expansion during pancreas development, potentially connecting microbiome composition with diabetes risk.
Collapse
Affiliation(s)
- Jennifer Hampton Hill
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA; Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | | | | | - Elena S Wall
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - Philip Jahl
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA; Department of Physics and Materials Science Institute, University of Oregon, Eugene, OR 97403, USA
| | - Rickesha Bell
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Karen Kallio
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - Daniel Derrick
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - L Charles Murtaugh
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Raghuveer Parthasarathy
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA; Department of Physics and Materials Science Institute, University of Oregon, Eugene, OR 97403, USA
| | - S James Remington
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - June L Round
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Karen Guillemin
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA; Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1Z8, Canada.
| |
Collapse
|
10
|
Arena KA, Zhu Y, Kucenas S. Transforming growth factor-beta signaling modulates perineurial glial bridging following peripheral spinal motor nerve injury in zebrafish. Glia 2022; 70:1826-1849. [PMID: 35616185 PMCID: PMC9378448 DOI: 10.1002/glia.24220] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 11/12/2022]
Abstract
Spinal motor nerves are necessary for organismal locomotion and survival. In zebrafish and most vertebrates, these peripheral nervous system structures are composed of bundles of axons that naturally regenerate following injury. However, the cellular and molecular mechanisms that mediate this process are still only partially understood. Perineurial glia, which form a component of the blood-nerve barrier, are necessary for the earliest regenerative steps by establishing a glial bridge across the injury site as well as phagocytosing debris. Without perineurial glial bridging, regeneration is impaired. In addition to perineurial glia, Schwann cells, the cells that ensheath and myelinate axons within the nerve, are essential for debris clearance and axon guidance. In the absence of Schwann cells, perineurial glia exhibit perturbed bridging, demonstrating that these two cell types communicate during the injury response. While the presence and importance of perineurial glial bridging is known, the molecular mechanisms that underlie this process remain a mystery. Understanding the cellular and molecular interactions that drive perineurial glial bridging is crucial to unlocking the mechanisms underlying successful motor nerve regeneration. Using laser axotomy and in vivo imaging in zebrafish, we show that transforming growth factor-beta (TGFβ) signaling modulates perineurial glial bridging. Further, we identify connective tissue growth factor-a (ctgfa) as a downstream effector of TGF-β signaling that works in a positive feedback loop to mediate perineurial glial bridging. Together, these studies present a new signaling pathway involved in the perineurial glial injury response and further characterize the dynamics of the perineurial glial bridge.
Collapse
Affiliation(s)
- Kimberly A. Arena
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- Program in Fundamental NeuroscienceUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Yunlu Zhu
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Sarah Kucenas
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- Program in Fundamental NeuroscienceUniversity of VirginiaCharlottesvilleVirginiaUSA
| |
Collapse
|
11
|
Ercc2/Xpd deficiency results in failure of digestive organ growth in zebrafish with elevated nucleolar stress. iScience 2022; 25:104957. [PMID: 36065184 PMCID: PMC9440294 DOI: 10.1016/j.isci.2022.104957] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/23/2022] [Accepted: 08/12/2022] [Indexed: 12/09/2022] Open
Abstract
Mutations in ERCC2/XPD helicase, an important component of the TFIIH complex, cause distinct human genetic disorders which exhibit various pathological features. However, the molecular mechanisms underlying many symptoms remain elusive. Here, we have shown that Ercc2/Xpd deficiency in zebrafish resulted in hypoplastic digestive organs with normal bud initiation but later failed to grow. The proliferation of intestinal endothelial cells was impaired in ercc2/xpd mutants, and mitochondrial abnormalities, autophagy, and inflammation were highly induced. Further studies revealed that these abnormalities were associated with the perturbation of rRNA synthesis and nucleolar stress in a p53-independent manner. As TFIIH has only been implicated in RNA polymerase I-dependent transcription in vitro, our results provide the first evidence for the connection between Ercc2/Xpd and rRNA synthesis in an animal model that recapitulates certain key characteristics of ERCC2/XPD-related human genetic disorders, and will greatly advance our understanding of the molecular pathogenesis of these diseases. Ercc2/Xpd deficiency results in failure of digestive organ growth in zebrafish Ercc2/Xpd-deficient intestinal endothelial cells exhibit impaired proliferation Mitochondrial abnormalities, autophagy, and inflammation are highly induced rRNA synthesis perturbation leads to nucleolar stress in a p53-independent manner
Collapse
|
12
|
Risato G, Celeghin R, Brañas Casas R, Dinarello A, Zuppardo A, Vettori A, Pilichou K, Thiene G, Basso C, Argenton F, Visentin S, Cosmi E, Tiso N, Beffagna G. Hyperactivation of Wnt/β-catenin and Jak/Stat3 pathways in human and zebrafish foetal growth restriction models: Implications for pharmacological rescue. Front Cell Dev Biol 2022; 10:943127. [PMID: 36051436 PMCID: PMC9424487 DOI: 10.3389/fcell.2022.943127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
Foetal Growth Restriction (FGR), previously known as Intrauterine Growth Restriction (IUGR), is an obstetrical condition due to placental insufficiency, affecting yearly about 30 million newborns worldwide. In this work, we aimed to identify and pharmacologically target signalling pathways specifically involved in the FGR condition, focusing on FGR-related cardiovascular phenotypes. The transcriptional profile of human umbilical cords from FGR and control cases was compared with the response to hypoxia of zebrafish (Danio rerio) transgenic lines reporting in vivo the activity of twelve signalling pathways involved in embryonic development. Wnt/β-catenin and Jak/Stat3 were found as key pathways significantly dysregulated in both human and zebrafish samples. This information was used in a chemical-genetic analysis to test drugs targeting Wnt/β-catenin and Jak/Stat3 pathways to rescue a set of FGR phenotypes, including growth restriction and cardiovascular modifications. Treatments with the Wnt/β-catenin agonist SB216763 successfully rescued body dimensions, cardiac shape, and vessel organization in zebrafish FGR models. Our data support the Wnt/β-catenin pathway as a key FGR marker and a promising target for pharmacological intervention in the FGR condition.
Collapse
Affiliation(s)
- Giovanni Risato
- Department of Biology, University of Padova, Padova, Italy
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Rudy Celeghin
- Department of Biology, University of Padova, Padova, Italy
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | | | | | | | - Andrea Vettori
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Kalliopi Pilichou
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Gaetano Thiene
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Cristina Basso
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | | | - Silvia Visentin
- Department of Women’s and Children’s Health, University of Padova, Padova, Italy
| | - Erich Cosmi
- Department of Women’s and Children’s Health, University of Padova, Padova, Italy
| | - Natascia Tiso
- Department of Biology, University of Padova, Padova, Italy
| | - Giorgia Beffagna
- Department of Biology, University of Padova, Padova, Italy
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padova, Padova, Italy
| |
Collapse
|
13
|
Faraj N, Duinkerken BHP, Carroll EC, Giepmans BNG. Microscopic modulation and analysis of islets of Langerhans in living zebrafish larvae. FEBS Lett 2022; 596:2497-2512. [DOI: 10.1002/1873-3468.14411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/22/2022] [Accepted: 05/20/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Noura Faraj
- Department of Biomedical Sciences of Cells and Systems, University of Groningen University Medical Center Groningen Groningen 9713AV The Netherlands
| | - B. H. Peter Duinkerken
- Department of Biomedical Sciences of Cells and Systems, University of Groningen University Medical Center Groningen Groningen 9713AV The Netherlands
| | - Elizabeth C. Carroll
- Department of Imaging Physics Delft University of Technology Delft, 2628 CJ The Netherlands
| | - Ben N. G. Giepmans
- Department of Biomedical Sciences of Cells and Systems, University of Groningen University Medical Center Groningen Groningen 9713AV The Netherlands
| |
Collapse
|
14
|
Evidence from oyster suggests an ancient role for Pdx in regulating insulin gene expression in animals. Nat Commun 2021; 12:3117. [PMID: 34035261 PMCID: PMC8149454 DOI: 10.1038/s41467-021-23216-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 04/19/2021] [Indexed: 11/17/2022] Open
Abstract
Hox and ParaHox genes encode transcription factors with similar expression patterns in divergent animals. The Pdx (Xlox) homeobox gene, for example, is expressed in a sharp spatial domain in the endodermal cell layer of the gut in chordates, echinoderms, annelids and molluscs. The significance of comparable gene expression patterns is unclear because it is not known if downstream transcriptional targets are also conserved. Here, we report evidence indicating that a classic transcriptional target of Pdx1 in vertebrates, the insulin gene, is a likely direct target of Pdx in Pacific oyster adults. We show that one insulin-related gene, cgILP, is co-expressed with cgPdx in oyster digestive tissue. Transcriptomic comparison suggests that this tissue plays a similar role to the vertebrate pancreas. Using ATAC-seq and ChIP, we identify an upstream regulatory element of the cgILP gene which shows binding interaction with cgPdx protein in oyster hepatopancreas and demonstrate, using a cell culture assay, that the oyster Pdx can act as a transcriptional activator through this site, possibly in synergy with NeuroD. These data argue that a classic homeodomain-target gene interaction dates back to the origin of Bilateria. In vertebrates insulin is a direct transcriptional target of Pdx: the same is true in Pacific oysters and the authors show insulin-related gene, cgILP, is co-expressed with cgPdx in oyster digestive tissue, showing this gene interaction dates back to the origin of Bilateria.
Collapse
|
15
|
Fontenas L, Kucenas S. Spinal cord precursors utilize neural crest cell mechanisms to generate hybrid peripheral myelinating glia. eLife 2021; 10:64267. [PMID: 33554855 PMCID: PMC7886336 DOI: 10.7554/elife.64267] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 02/05/2021] [Indexed: 12/11/2022] Open
Abstract
During development, oligodendrocytes and Schwann cells myelinate central and peripheral nervous system axons, respectively, while motor exit point (MEP) glia are neural tube-derived, peripheral glia that myelinate axonal territory between these populations at MEP transition zones. From which specific neural tube precursors MEP glia are specified, and how they exit the neural tube to migrate onto peripheral motor axons, remain largely unknown. Here, using zebrafish, we found that MEP glia arise from lateral floor plate precursors and require foxd3 to delaminate and exit the spinal cord. Additionally, we show that similar to Schwann cells, MEP glial development depends on axonally derived neuregulin1. Finally, our data demonstrate that overexpressing axonal cues is sufficient to generate additional MEP glia in the spinal cord. Overall, these studies provide new insight into how a novel population of hybrid, peripheral myelinating glia are generated from neural tube precursors and migrate into the periphery.
Collapse
Affiliation(s)
- Laura Fontenas
- Department of Biology, University of Virginia, Charlottesville, United States
| | - Sarah Kucenas
- Department of Biology, University of Virginia, Charlottesville, United States
| |
Collapse
|
16
|
Efficient clofilium tosylate-mediated rescue of POLG-related disease phenotypes in zebrafish. Cell Death Dis 2021; 12:100. [PMID: 33469036 PMCID: PMC7815880 DOI: 10.1038/s41419-020-03359-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 12/08/2020] [Accepted: 12/16/2020] [Indexed: 02/07/2023]
Abstract
The DNA polymerase gamma (Polg) is a nuclear-encoded enzyme involved in DNA replication in animal mitochondria. In humans, mutations in the POLG gene underlie a set of mitochondrial diseases characterized by mitochondrial DNA (mtDNA) depletion or deletion and multiorgan defects, named POLG disorders, for which an effective therapy is still needed. By applying antisense strategies, ENU- and CRISPR/Cas9-based mutagenesis, we have generated embryonic, larval-lethal and adult-viable zebrafish Polg models. Morphological and functional characterizations detected a set of phenotypes remarkably associated to POLG disorders, including cardiac, skeletal muscle, hepatic and gonadal defects, as well as mitochondrial dysfunctions and, notably, a perturbed mitochondria-to-nucleus retrograde signaling (CREB and Hypoxia pathways). Next, taking advantage of preliminary evidence on the candidate molecule Clofilium tosylate (CLO), we tested CLO toxicity and then its efficacy in our zebrafish lines. Interestingly, at well tolerated doses, the CLO drug could successfully rescue mtDNA and Complex I respiratory activity to normal levels, even in mutant phenotypes worsened by treatment with Ethidium Bromide. In addition, the CLO drug could efficiently restore cardio-skeletal parameters and mitochondrial mass back to normal values. Altogether, these evidences point to zebrafish as a valuable vertebrate organism to faithfully phenocopy multiple defects detected in POLG patients. Moreover, this model represents an excellent platform to screen, at the whole-animal level, candidate molecules with therapeutic effects in POLG disorders.
Collapse
|
17
|
Rastogi A, Severance EG, Jacobs HM, Conlin SM, Islam ST, Timme-Laragy AR. Modulating glutathione thiol status alters pancreatic β-cell morphogenesis in the developing zebrafish (Danio rerio) embryo. Redox Biol 2021; 38:101788. [PMID: 33321464 PMCID: PMC7744774 DOI: 10.1016/j.redox.2020.101788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/26/2020] [Accepted: 11/03/2020] [Indexed: 01/07/2023] Open
Abstract
Emerging evidence suggests that redox-active chemicals perturb pancreatic islet development. To better understand potential mechanisms for this, we used zebrafish (Danio rerio) embryos to investigate roles of glutathione (GSH; predominant cellular redox buffer) and the transcription factor Nrf2a (Nfe2l2a; zebrafish Nrf2 co-ortholog) in islet morphogenesis. We delineated critical windows of susceptibility to redox disruption of β-cell morphogenesis, interrogating embryos at 24, 48 and 72 h post fertilization (hpf) and visualized Nrf2a expression in the pancreas using whole-mount immunohistochemistry at 96 hpf. Chemical GSH modulation at 48 hpf induced significant islet morphology changes at 96 hpf. Pro-oxidant exposures to tert-butylhydroperoxide (77.6 μM; 10-min at 48 hpf) or tert-butylhydroquinone (1 μM; 48-56 hpf) decreased β-cell cluster area at 96 hpf. Conversely, exposures to antioxidant N-acetylcysteine (bolsters GSH pools; 100 μM; 48-72 hpf) or sulforaphane (activates Nrf2a; 20 μM; 48-72 hpf) significantly increased islet areas. Nrf2a was also stabilized in β-cells: 10-min exposures to 77.6 μM tert-butylhydroperoxide significantly increased Nrf2a protein compared to control islet cells that largely lack stabilized Nrf2a; 10-min exposures to higher (776 μM) tert-butylhydroperoxide concentration stabilized Nrf2a throughout the pancreas. Using biotinylated-GSH to visualize in situ protein glutathionylation, islet cells displayed high protein glutathionylation, indicating oxidized GSH pools. The 10-min high (776 μM) tert-butylhydroperoxide exposure (induced Nrf2a globally) decreased global protein glutathionylation at 96 hpf. Mutant fish expressing inactive Nrf2a were protected against tert-butylhydroperoxide-induced abnormal islet morphology. Our data indicate that disrupted redox homeostasis and Nrf2a stabilization during pancreatic β-cell development impact morphogenesis, with implications for disease states at later life stages. Our work identifies a potential molecular target (Nrf2) that mediates abnormal β-cell morphology in response to redox disruptions. Moreover, our findings imply that developmental exposure to exogenous stressors at distinct windows of susceptibility could diminish the reserve redox capacity of β-cells, rendering them vulnerable to later-life stresses and disease.
Collapse
Affiliation(s)
- Archit Rastogi
- Molecular & Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, 01003, USA
| | - Emily G Severance
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Haydee M Jacobs
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Sarah M Conlin
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Sadia T Islam
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Alicia R Timme-Laragy
- Molecular & Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, 01003, USA; Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA, 01003, USA.
| |
Collapse
|
18
|
Hernandez-Perez M, Kulkarni A, Samala N, Sorrell C, El K, Haider I, Aleem AM, Holman TR, Rai G, Tersey SA, Mirmira RG, Anderson RM. A 12-lipoxygenase-Gpr31 signaling axis is required for pancreatic organogenesis in the zebrafish. FASEB J 2020; 34:14850-14862. [PMID: 32918516 PMCID: PMC7606739 DOI: 10.1096/fj.201902308rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 08/21/2020] [Accepted: 08/25/2020] [Indexed: 12/19/2022]
Abstract
12-Lipoxygenase (12-LOX) is a key enzyme in arachidonic acid metabolism, and alongside its major product, 12-HETE, plays a key role in promoting inflammatory signaling during diabetes pathogenesis. Although 12-LOX is a proposed therapeutic target to protect pancreatic islets in the setting of diabetes, little is known about the consequences of blocking its enzymatic activity during embryonic development. Here, we have leveraged the strengths of the zebrafish-genetic manipulation and pharmacologic inhibition-to interrogate the role of 12-LOX in pancreatic development. Lipidomics analysis during zebrafish development demonstrated that 12-LOX-generated metabolites of arachidonic acid increase sharply during organogenesis stages, and that this increase is blocked by morpholino-directed depletion of 12-LOX. Furthermore, we found that either depletion or inhibition of 12-LOX impairs both exocrine pancreas growth and unexpectedly, the generation of insulin-producing β cells. We demonstrate that morpholino-mediated knockdown of GPR31, a purported G-protein-coupled receptor for 12-HETE, largely phenocopies both the depletion and the inhibition of 12-LOX. Moreover, we show that loss of GPR31 impairs pancreatic bud fusion and pancreatic duct morphogenesis. Together, these data provide new insight into the requirement of 12-LOX in pancreatic organogenesis and islet formation, and additionally provide evidence that its effects are mediated via a signaling axis that includes the 12-HETE receptor GPR31.
Collapse
Affiliation(s)
- Marimar Hernandez-Perez
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Abhishek Kulkarni
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Niharika Samala
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cody Sorrell
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kimberly El
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Isra Haider
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ansari Mukhtar Aleem
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Theodore R Holman
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Ganesha Rai
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Sarah A Tersey
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Raghavendra G Mirmira
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Medicine, Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| | - Ryan M Anderson
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Medicine, Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
19
|
Lavergne A, Tarifeño-Saldivia E, Pirson J, Reuter AS, Flasse L, Manfroid I, Voz ML, Peers B. Pancreatic and intestinal endocrine cells in zebrafish share common transcriptomic signatures and regulatory programmes. BMC Biol 2020; 18:109. [PMID: 32867764 PMCID: PMC7457809 DOI: 10.1186/s12915-020-00840-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 08/04/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Endocrine cells of the zebrafish digestive system play an important role in regulating metabolism and include pancreatic endocrine cells (PECs) clustered in the islets of Langerhans and the enteroendocrine cells (EECs) scattered in the intestinal epithelium. Despite EECs and PECs are being located in distinct organs, their differentiation involves shared molecular mechanisms and transcription factors. However, their degree of relatedness remains unexplored. In this study, we investigated comprehensively the similarity of EECs and PECs by defining their transcriptomic landscape and comparing the regulatory programmes controlled by Pax6b, a key player in both EEC and PEC differentiations. RESULTS RNA sequencing was performed on EECs and PECs isolated from wild-type and pax6b mutant zebrafish. Data mining of wild-type zebrafish EEC data confirmed the expression of orthologues for most known mammalian EEC hormones, but also revealed the expression of three additional neuropeptide hormones (Proenkephalin-a, Calcitonin-a and Adcyap1a) not previously reported to be expressed by EECs in any species. Comparison of transcriptomes from EECs, PECs and other zebrafish tissues highlights a very close similarity between EECs and PECs, with more than 70% of genes being expressed in both endocrine cell types. Comparison of Pax6b-regulated genes in EECs and PECs revealed a significant overlap. pax6b loss-of-function does not affect the total number of EECs and PECs but instead disrupts the balance between endocrine cell subtypes, leading to an increase of ghrelin- and motilin-like-expressing cells in both the intestine and pancreas at the expense of other endocrine cells such as beta and delta cells in the pancreas and pyyb-expressing cells in the intestine. Finally, we show that the homeodomain of Pax6b is dispensable for its action in both EECs and PECs. CONCLUSION We have analysed the transcriptomic landscape of wild-type and pax6b mutant zebrafish EECs and PECs. Our study highlights the close relatedness of EECs and PECs at the transcriptomic and regulatory levels, supporting the hypothesis of a common phylogenetic origin and underscoring the potential implication of EECs in metabolic diseases such as type 2 diabetes.
Collapse
Affiliation(s)
- Arnaud Lavergne
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l’Hôpital 1, B34, Sart Tilman, 4000 Liège, Belgium
| | - Estefania Tarifeño-Saldivia
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l’Hôpital 1, B34, Sart Tilman, 4000 Liège, Belgium
- Present Address: Gene Expression and Regulation Laboratory, Department of Biochemistry and Molecular Biology, University of Concepción, Concepción, Chile
| | - Justine Pirson
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l’Hôpital 1, B34, Sart Tilman, 4000 Liège, Belgium
| | - Anne-Sophie Reuter
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l’Hôpital 1, B34, Sart Tilman, 4000 Liège, Belgium
| | - Lydie Flasse
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l’Hôpital 1, B34, Sart Tilman, 4000 Liège, Belgium
| | - Isabelle Manfroid
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l’Hôpital 1, B34, Sart Tilman, 4000 Liège, Belgium
| | - Marianne L. Voz
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l’Hôpital 1, B34, Sart Tilman, 4000 Liège, Belgium
| | - Bernard Peers
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l’Hôpital 1, B34, Sart Tilman, 4000 Liège, Belgium
| |
Collapse
|
20
|
Lu CJ, Fan XY, Guo YF, Cheng ZC, Dong J, Chen JZ, Li LY, Wang MW, Wu ZK, Wang F, Tong XJ, Luo LF, Tang FC, Zhu ZY, Zhang B. Single-cell analyses identify distinct and intermediate states of zebrafish pancreatic islet development. J Mol Cell Biol 2020; 11:435-447. [PMID: 30407522 PMCID: PMC6604604 DOI: 10.1093/jmcb/mjy064] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 10/31/2018] [Accepted: 11/08/2018] [Indexed: 12/13/2022] Open
Abstract
Pancreatic endocrine islets are vital for glucose homeostasis. However, the islet developmental trajectory and its regulatory network are not well understood. To define the features of these specification and differentiation processes, we isolated individual islet cells from TgBAC(neurod1:EGFP) transgenic zebrafish and analyzed islet developmental dynamics across four different embryonic stages using a single-cell RNA-seq strategy. We identified proliferative endocrine progenitors, which could be further categorized by different cell cycle phases with the G1/S subpopulation displaying a distinct differentiation potential. We identified endocrine precursors, a heterogeneous intermediate-state population consisting of lineage-primed alpha, beta and delta cells that were characterized by the expression of lineage-specific transcription factors and relatively low expression of terminally differentiation markers. The terminally differentiated alpha, beta, and delta cells displayed stage-dependent differentiation states, which were related to their functional maturation. Our data unveiled distinct states, events and molecular features during the islet developmental transition, and provided resources to comprehensively understand the lineage hierarchy of islet development at the single-cell level.
Collapse
Affiliation(s)
- Chong-Jian Lu
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Xiao-Ying Fan
- Beijing Advanced Innovation Center for Genomics (ICG), College of Life Sciences, Peking University, Beijing, China
| | - Yue-Feng Guo
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Zhen-Chao Cheng
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Ji Dong
- Beijing Advanced Innovation Center for Genomics (ICG), College of Life Sciences, Peking University, Beijing, China
| | - Jin-Zi Chen
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Chongqing, China
| | - Lian-Yan Li
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Mei-Wen Wang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Ze-Kai Wu
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Fei Wang
- National Center for Protein Sciences, Peking University, Beijing, China
| | - Xiang-Jun Tong
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Ling-Fei Luo
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Chongqing, China
| | - Fu-Chou Tang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China.,Beijing Advanced Innovation Center for Genomics (ICG), College of Life Sciences, Peking University, Beijing, China
| | - Zuo-Yan Zhu
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Bo Zhang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| |
Collapse
|
21
|
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
|
22
|
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
|
23
|
Adusumilli L, Facchinello N, Teh C, Busolin G, Le MTN, Yang H, Beffagna G, Campanaro S, Tam WL, Argenton F, Lim B, Korzh V, Tiso N. miR-7 Controls the Dopaminergic/Oligodendroglial Fate through Wnt/β-catenin Signaling Regulation. Cells 2020; 9:cells9030711. [PMID: 32183236 PMCID: PMC7140713 DOI: 10.3390/cells9030711] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 03/07/2020] [Accepted: 03/11/2020] [Indexed: 12/23/2022] Open
Abstract
During the development of the central nervous system, the proliferation of neural progenitors and differentiation of neurons and glia are tightly regulated by different transcription factors and signaling cascades, such as the Wnt and Shh pathways. This process takes place in cooperation with several microRNAs, some of which evolutionarily conserved in vertebrates, from teleosts to mammals. We focused our attention on miR-7, as its role in the regulation of cell signaling during neural development is still unclear. Specifically, we used human stem cell cultures and whole zebrafish embryos to study, in vitro and in vivo, the role of miR-7 in the development of dopaminergic (DA) neurons, a cell type primarily affected in Parkinson’s disease. We demonstrated that the zebrafish homologue of miR-7 (miR-7a) is expressed in the forebrain during the development of DA neurons. Moreover, we identified 143 target genes downregulated by miR-7, including the neural fate markers TCF4 and TCF12, as well as the Wnt pathway effector TCF7L2. We then demonstrated that miR-7 negatively regulates the proliferation of DA-progenitors by inhibiting Wnt/β-catenin signaling in zebrafish embryos. In parallel, miR-7 positively regulates Shh signaling, thus controlling the balance between oligodendroglial and DA neuronal cell fates. In summary, this study identifies a new molecular cross-talk between Wnt and Shh signaling pathways during the development of DA-neurons. Being mediated by a microRNA, this mechanism represents a promising target in cell differentiation therapies for Parkinson’s disease.
Collapse
Affiliation(s)
- Lavanya Adusumilli
- Genome Institute of Singapore, A-STAR, Singapore 138672, Singapore; (L.A.); (H.Y.); (W.L.T.)
| | - Nicola Facchinello
- Department of Biology, University of Padova, 35131 Padova, Italy; (N.F.); (G.B.); (G.B.); (S.C.); (F.A.)
| | - Cathleen Teh
- Institute of Molecular and Cell Biology, A-STAR, Singapore 138632, Singapore;
- Department of Biological Sciences, National University of Singapore, Singapore 117558, Singapore
| | - Giorgia Busolin
- Department of Biology, University of Padova, 35131 Padova, Italy; (N.F.); (G.B.); (G.B.); (S.C.); (F.A.)
| | - Minh TN Le
- Department of Pharmacology, National University of Singapore, Singapore 117559, Singapore;
| | - Henry Yang
- Genome Institute of Singapore, A-STAR, Singapore 138672, Singapore; (L.A.); (H.Y.); (W.L.T.)
| | - Giorgia Beffagna
- Department of Biology, University of Padova, 35131 Padova, Italy; (N.F.); (G.B.); (G.B.); (S.C.); (F.A.)
| | - Stefano Campanaro
- Department of Biology, University of Padova, 35131 Padova, Italy; (N.F.); (G.B.); (G.B.); (S.C.); (F.A.)
| | - Wai Leong Tam
- Genome Institute of Singapore, A-STAR, Singapore 138672, Singapore; (L.A.); (H.Y.); (W.L.T.)
| | - Francesco Argenton
- Department of Biology, University of Padova, 35131 Padova, Italy; (N.F.); (G.B.); (G.B.); (S.C.); (F.A.)
| | - Bing Lim
- Genome Institute of Singapore, A-STAR, Singapore 138672, Singapore; (L.A.); (H.Y.); (W.L.T.)
- Correspondence: (B.L.); (V.K.); (N.T.); Tel.: +1-781-484-7643 (B.L.); +48-22-597-07-65 (V.K.); +39-049-827-6302 (N.T.)
| | - Vladimir Korzh
- Institute of Molecular and Cell Biology, A-STAR, Singapore 138632, Singapore;
- International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
- Correspondence: (B.L.); (V.K.); (N.T.); Tel.: +1-781-484-7643 (B.L.); +48-22-597-07-65 (V.K.); +39-049-827-6302 (N.T.)
| | - Natascia Tiso
- Department of Biology, University of Padova, 35131 Padova, Italy; (N.F.); (G.B.); (G.B.); (S.C.); (F.A.)
- Correspondence: (B.L.); (V.K.); (N.T.); Tel.: +1-781-484-7643 (B.L.); +48-22-597-07-65 (V.K.); +39-049-827-6302 (N.T.)
| |
Collapse
|
24
|
Zhu Y, Crowley SC, Latimer AJ, Lewis GM, Nash R, Kucenas S. Migratory Neural Crest Cells Phagocytose Dead Cells in the Developing Nervous System. Cell 2019; 179:74-89.e10. [PMID: 31495570 DOI: 10.1016/j.cell.2019.08.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 07/16/2019] [Accepted: 08/01/2019] [Indexed: 12/20/2022]
Abstract
During neural tube closure and spinal cord development, many cells die in both the central and peripheral nervous systems (CNS and PNS, respectively). However, myeloid-derived professional phagocytes have not yet colonized the trunk region during early neurogenesis. How apoptotic cells are removed from this region during these stages remains largely unknown. Using live imaging in zebrafish, we demonstrate that neural crest cells (NCCs) respond rapidly to dying cells and phagocytose cellular debris around the neural tube. Additionally, NCCs have the ability to enter the CNS through motor exit point transition zones and clear debris in the spinal cord. Surprisingly, NCCs phagocytosis mechanistically resembles macrophage phagocytosis and their recruitment toward cellular debris is mediated by interleukin-1β. Taken together, our results reveal a role for NCCs in phagocytosis of debris in the developing nervous system before the presence of professional phagocytes.
Collapse
Affiliation(s)
- Yunlu Zhu
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Samantha C Crowley
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Andrew J Latimer
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Gwendolyn M Lewis
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Rebecca Nash
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Sarah Kucenas
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA.
| |
Collapse
|
25
|
Vetrivel S, Tiso N, Kügler A, Irmler M, Horsch M, Beckers J, Hladik D, Giesert F, Gailus-Durner V, Fuchs H, Sabrautzki S, Hrabě de Angelis M, Graw J. Mutation in the mouse histone gene Hist2h3c1 leads to degeneration of the lens vesicle and severe microphthalmia. Exp Eye Res 2019; 188:107632. [PMID: 30991053 PMCID: PMC6876282 DOI: 10.1016/j.exer.2019.03.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 03/20/2019] [Accepted: 03/30/2019] [Indexed: 12/21/2022]
Abstract
During an ENU (N-ethyl-N-nitrosourea) mutagenesis screen, we observed a dominant small-eye mutant mouse with viable homozygotes. A corresponding mutant line was established and referred to as Aey69 (abnormality of the eye #69). Comprehensive phenotyping of the homozygous Aey69 mutants in the German Mouse Clinic revealed only a subset of statistically significant alterations between wild types and homozygous mutants. The mutation causes microphthalmia without a lens but with retinal hyperproliferation. Linkage was demonstrated to mouse chromosome 3 between the markers D3Mit188 and D3Mit11. Sequencing revealed a 358 A-> C mutation (Ile120Leu) in the Hist2h3c1 gene and a 71 T-> C (Val24Ala) mutation in the Gja8 gene. Detailed analysis of eye development in the homozygous mutant mice documented a perturbed lens development starting from the lens vesicle stage including decreasing expression of crystallins as well as of lens-specific transcription factors like PITX3 and FOXE3. In contrast, we observed an early expression of retinal progenitor cells characterized by several markers including BRN3 (retinal ganglion cells) and OTX2 (cone photoreceptors). The changes in the retina at the early embryonic stages of E11.5-E15.5 happen in parallel with apoptotic processes in the lens at the respective stages. The excessive retinal hyperproliferation is characterized by an increased level of Ki67. The hyperproliferation, however, does not disrupt the differentiation and appearance of the principal retinal cell types at postnatal stages, even if the overgrowing retina covers finally the entire bulbus of the eye. Morpholino-mediated knock-down of the hist2h3ca1 gene in zebrafish leads to a specific perturbation of lens development. When injected into zebrafish zygotes, only the mutant mouse mRNA leads to severe malformations, ranging from cyclopia to severe microphthalmia. The wild-type Hist2h3c1 mRNA can rescue the morpholino-induced defects corroborating its specific function in lens development. Based upon these data, it is concluded that the ocular function of the Hist2h3c1 gene (encoding a canonical H3.2 variant) is conserved throughout evolution. Moreover, the data highlight also the importance of Hist2h3c1 in the coordinated formation of lens and retina during eye development. A dominant small-eye mutant mouse is caused by a mutation in the histone gene Hist2H3c1. Morpholino-mediated knock-down of hist2h3ca1 in the zebrafish validated this finding. The mutation leads to degeneration of the lens vesicle and retina hyperproliferation.
Collapse
Affiliation(s)
- Sharmilee Vetrivel
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Developmental Genetics, D-85764 Neuherberg, Germany
| | - Natascia Tiso
- Department of Biology, University of Padova, I-35131 Padova, Italy.
| | - Andrea Kügler
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Developmental Genetics, D-85764 Neuherberg, Germany
| | - Martin Irmler
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Experimental Genetics, D-85764 Neuherberg, Germany
| | - Marion Horsch
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Experimental Genetics, D-85764 Neuherberg, Germany
| | - Johannes Beckers
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Experimental Genetics, D-85764 Neuherberg, Germany; Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, D-85354 Freising, Germany; German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany
| | - Daniela Hladik
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Developmental Genetics, D-85764 Neuherberg, Germany
| | - Florian Giesert
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Developmental Genetics, D-85764 Neuherberg, Germany
| | - Valerie Gailus-Durner
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Experimental Genetics, D-85764 Neuherberg, Germany
| | - Helmut Fuchs
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Experimental Genetics, D-85764 Neuherberg, Germany
| | - Sibylle Sabrautzki
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Experimental Genetics, D-85764 Neuherberg, Germany; Helmholtz Center Munich, German Research Center for Environmental Health, Research Unit Comparative Medicine, D-85764 Neuherberg, Germany
| | - Martin Hrabě de Angelis
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Experimental Genetics, D-85764 Neuherberg, Germany; Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, D-85354 Freising, Germany; German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany
| | - Jochen Graw
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Developmental Genetics, D-85764 Neuherberg, Germany.
| |
Collapse
|
26
|
Small BC, Quiniou SM, Kaiya H, Bledsoe JW, Musungu B. Characterization of a third ghrelin receptor, GHS-R3a, in channel catfish reveals novel expression patterns and a high affinity for homologous ligand. Comp Biochem Physiol A Mol Integr Physiol 2019; 229:1-9. [DOI: 10.1016/j.cbpa.2018.11.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/16/2018] [Accepted: 11/18/2018] [Indexed: 11/30/2022]
|
27
|
Jacobs HM, Sant KE, Basnet A, Williams LM, Moss JB, Timme-Laragy AR. Embryonic exposure to Mono(2-ethylhexyl) phthalate (MEHP) disrupts pancreatic organogenesis in zebrafish (Danio rerio). CHEMOSPHERE 2018; 195:498-507. [PMID: 29277029 PMCID: PMC5788038 DOI: 10.1016/j.chemosphere.2017.12.094] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 12/14/2017] [Accepted: 12/15/2017] [Indexed: 05/22/2023]
Abstract
Mono(2-ethylhexyl) phthalate (MEHP) is the bioactive metabolite of di(2-ethylhexyl) phthalate, a plasticizing agent and persistent environmental contaminant associated with obesity, developmental abnormalities, and oxidative stress. Nrf2 (Nfe2l2) is a transcription factor that regulates cytoprotective genes as part of the adaptive antioxidant response. We previously identified the pancreas as a sensitive target of oxidative stress during embryonic development. The goals of this study were to 1) characterize the effects of MEHP exposure on pancreatic development, and 2) determine whether oxidative stress contributes to MEHP embryotoxicity. Zebrafish (Danio rerio) embryos from AB wildtype and Tg(ins:GFP;nrf2afh318/fh318) were exposed to 0 or 200 μg/L MEHP at 3 h post fertilization (hpf) through 168 hpf to assess pancreatic organogenesis. MEHP exposure significantly decreased β-cell area at all timepoints (48, 72, 96, 168 hpf), but Nrf2a did not significantly protect against islet hypomorphism. Tg(gcga:GFP) embryos exposed to MEHP showed a decrease in α-cell area in the islet across the same timepoints. Tg(ptf1a:GFP) embryos were assessed at 80 and 168 hpf for exocrine pancreas length. MEHP exposure decreased growth of the exocrine pancreas. Expression of pancreas genes insa, sst2 and ptf1a was significantly reduced by MEHP exposure compared to controls. Glutathione (GSH) concentrations and redox potentials were quantified at 72 hpf by HPLC, but no significant changes were observed. However, expression of the GSH-related genes gstp1 and gsr were significantly altered by MEHP exposure. These data indicate that the developing pancreas is a sensitive target tissue of embryonic exposure to MEHP.
Collapse
Affiliation(s)
- Haydee M Jacobs
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, MA 01003, USA
| | - Karilyn E Sant
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, MA 01003, USA
| | - Aviraj Basnet
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, MA 01003, USA
| | | | - Jennifer B Moss
- Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27701, USA
| | - Alicia R Timme-Laragy
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, MA 01003, USA.
| |
Collapse
|
28
|
Troll JV, Hamilton MK, Abel ML, Ganz J, Bates JM, Stephens WZ, Melancon E, van der Vaart M, Meijer AH, Distel M, Eisen JS, Guillemin K. Microbiota promote secretory cell determination in the intestinal epithelium by modulating host Notch signaling. Development 2018; 145:145/4/dev155317. [PMID: 29475973 DOI: 10.1242/dev.155317] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 01/19/2018] [Indexed: 12/15/2022]
Abstract
Resident microbes promote many aspects of host development, although the mechanisms by which microbiota influence host tissues remain unclear. We showed previously that the microbiota is required for allocation of appropriate numbers of secretory cells in the zebrafish intestinal epithelium. Because Notch signaling is crucial for secretory fate determination, we conducted epistasis experiments to establish whether the microbiota modulates host Notch signaling. We also investigated whether innate immune signaling transduces microbiota cues via the Myd88 adaptor protein. We provide the first evidence that microbiota-induced, Myd88-dependent signaling inhibits host Notch signaling in the intestinal epithelium, thereby promoting secretory cell fate determination. These results connect microbiota activity via innate immune signaling to the Notch pathway, which also plays crucial roles in intestinal homeostasis throughout life and when impaired can result in chronic inflammation and cancer.
Collapse
Affiliation(s)
- Joshua V Troll
- Institute of Molecular Biology, Department of Biology, 1229 University of Oregon, Eugene, OR 97403, USA
| | - M Kristina Hamilton
- Institute of Neuroscience, Department of Biology, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Melissa L Abel
- Institute of Molecular Biology, Department of Biology, 1229 University of Oregon, Eugene, OR 97403, USA
| | - Julia Ganz
- Institute of Neuroscience, Department of Biology, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Jennifer M Bates
- Institute of Molecular Biology, Department of Biology, 1229 University of Oregon, Eugene, OR 97403, USA
| | - W Zac Stephens
- Institute of Molecular Biology, Department of Biology, 1229 University of Oregon, Eugene, OR 97403, USA
| | - Ellie Melancon
- Institute of Neuroscience, Department of Biology, 1254 University of Oregon, Eugene, OR 97403, USA
| | | | - Annemarie H Meijer
- Institute of Biology, Leiden University, 2300 RA Leiden, The Netherlands
| | - Martin Distel
- Children's Cancer Research Institute, 1090 Vienna, Austria
| | - Judith S Eisen
- Institute of Neuroscience, Department of Biology, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Karen Guillemin
- Institute of Molecular Biology, Department of Biology, 1229 University of Oregon, Eugene, OR 97403, USA .,Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, Ontario, Canada M5G 1Z8
| |
Collapse
|
29
|
SPLICS: a split green fluorescent protein-based contact site sensor for narrow and wide heterotypic organelle juxtaposition. Cell Death Differ 2017; 25:1131-1145. [PMID: 29229997 PMCID: PMC5988678 DOI: 10.1038/s41418-017-0033-z] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 10/17/2017] [Accepted: 10/31/2017] [Indexed: 01/06/2023] Open
Abstract
Contact sites are discrete areas of organelle proximity that coordinate essential physiological processes across membranes, including Ca2+ signaling, lipid biosynthesis, apoptosis, and autophagy. However, tools to easily image inter-organelle proximity over a range of distances in living cells and in vivo are lacking. Here we report a split-GFP-based contact site sensor (SPLICS) engineered to fluoresce when organelles are in proximity. Two SPLICS versions efficiently measured narrow (8–10 nm) and wide (40–50 nm) juxtapositions between endoplasmic reticulum and mitochondria, documenting the existence of at least two types of contact sites in human cells. Narrow and wide ER–mitochondria contact sites responded differently to starvation, ER stress, mitochondrial shape modifications, and changes in the levels of modulators of ER–mitochondria juxtaposition. SPLICS detected contact sites in soma and axons of D. rerio Rohon Beard (RB) sensory neurons invivo, extending its use to analyses of organelle juxtaposition in the whole animal.
Collapse
|
30
|
Identification of the principal transcriptional regulators for low-fat and high-fat meal responsive genes in small intestine. Nutr Metab (Lond) 2017; 14:66. [PMID: 29075307 PMCID: PMC5654052 DOI: 10.1186/s12986-017-0221-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 10/16/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND High-fat (HF) diet is a well-known cause of obesity. To identify principle transcriptional regulators that could be therapeutic targets of obesity, we investigated transcriptomic modulation in the duodenal mucosa following low-fat (LF) and HF meal ingestion. METHODS Whereas one group of mice was sacrificed after fasting, the others were fed ad libitum with LF or HF meal, and sacrificed 30 min, 1 h and 3 h after the beginning of the meal. A transcriptome analysis of the duodenal mucosa of the 7 groups was conducted using both microarray and serial analysis of gene expression (SAGE) method followed by an Ingenuity Pathways Analysis (IPA). RESULTS SAGE and microarray showed that the modulation of a total of 896 transcripts in the duodenal mucosa after LF and/or HF meal, compared to the fasting condition. The IPA identified lipid metabolism, molecular transport, and small molecule biochemistry as top three molecular and cellular functions for the HF-responsive, HF-specific, HF-delay, and LF-HF different genes. Moreover, the top transcriptional regulator for the HF-responsive and HF-specific genes was peroxisome proliferator-activated receptor alpha (PPARα). On the other hand, the LF-responsive and LF-specific genes were related to carbohydrate metabolism, cellular function and maintenance, and cell death/cellular growth and proliferation, and the top transcriptional regulators were forkhead box protein O1 (FOXO1) and cAMP response element binding protein 1 (CREB1), respectively. CONCLUSIONS These results will help to understand the molecular mechanisms of intestinal response after LF and HF ingestions, and contribute to identify therapeutic targets for obesity and obesity-related diseases.
Collapse
|
31
|
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
|
32
|
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
|
33
|
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
|
34
|
|
35
|
Astone M, Pizzi M, Peron M, Domenichini A, Guzzardo V, Töchterle S, Tiso N, Rugge M, Meyer D, Argenton F, Vettori A. A GFP-Tagged Gross Deletion on Chromosome 1 Causes Malignant Peripheral Nerve Sheath Tumors and Carcinomas in Zebrafish. PLoS One 2015; 10:e0145178. [PMID: 26695815 PMCID: PMC4687860 DOI: 10.1371/journal.pone.0145178] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 11/30/2015] [Indexed: 11/19/2022] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are highly aggressive soft-tissue sarcomas, characterized by complex karyotypes. The molecular bases of such malignancy are poorly understood and efficient targeted molecular therapies are currently lacking. Here we describe a novel zebrafish model of MPNSTs, represented by the transgenic mutant line Tg(-8.5nkx2.2a:GFP)ia2. ia2 homozygous animals displayed embryonic lethality by 72 hpf, while the heterozygotes develop visible tumor masses with high frequency in adulthood. Histological and immunohistochemical examination revealed aggressive tumors with either mesenchymal or epithelial features. The former (54% of the cases) arose either in the abdominal cavity, or as intrathecal/intraspinal lesions and is composed of cytokeratin-negative spindle cells with fascicular/storiform growth pattern consistent with zebrafish MPNSTs. The second histotype was composed by polygonal or elongated cells, immunohistochemically positive for the pan-cytokeratin AE1/AE3. The overall histologic and immunohistochemical features were consistent with a malignant epithelial neoplasm of possible gastrointestinal/pancreatic origin. With an integrated approach, based on microsatellite (VNTR) and STS markers, we showed that ia2 insertion, in Tg(-8.5nkx2.2a:GFP)ia2 embryos, is associated with a deletion of 15.2 Mb in the telomeric portion of chromosome 1. Interestingly, among ia2 deleted genes we identified the presence of the 40S ribosomal protein S6 gene that may be one of the possible drivers for the MPNSTs in ia2 mutants. Thanks to the peculiar features of zebrafish as animal model of human cancer (cellular and genomic similarity, transparency and prolificacy) and the GFP tag, the Tg(-8.5nkx2.2a:GFP)ia2 line provides a manageable tool to study in vivo with high frequency MPNST biology and genetics, and to identify, in concert with the existing zebrafish MPNST models, conserved relevant mechanisms in zebrafish and human cancer development.
Collapse
Affiliation(s)
- Matteo Astone
- Department of Biology, University of Padova, Padova, Italy
| | - Marco Pizzi
- General Pathology & Cytopathology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | | | | | - Vincenza Guzzardo
- General Pathology & Cytopathology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Sonja Töchterle
- Institute for Molecular Biology/ CMBI, University of Innsbruck, 6020 Innsbruck, Austria
| | - Natascia Tiso
- Department of Biology, University of Padova, Padova, Italy
| | - Massimo Rugge
- General Pathology & Cytopathology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Dirk Meyer
- Institute for Molecular Biology/ CMBI, University of Innsbruck, 6020 Innsbruck, Austria
| | | | - Andrea Vettori
- Department of Biology, University of Padova, Padova, Italy
| |
Collapse
|
36
|
Ye L, Robertson MA, Mastracci TL, Anderson RM. An insulin signaling feedback loop regulates pancreas progenitor cell differentiation during islet development and regeneration. Dev Biol 2015; 409:354-69. [PMID: 26658317 DOI: 10.1016/j.ydbio.2015.12.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 11/12/2015] [Accepted: 12/02/2015] [Indexed: 02/06/2023]
Abstract
As one of the key nutrient sensors, insulin signaling plays an important role in integrating environmental energy cues with organism growth. In adult organisms, relative insufficiency of insulin signaling induces compensatory expansion of insulin-secreting pancreatic beta (β) cells. However, little is known about how insulin signaling feedback might influence neogenesis of β cells during embryonic development. Using genetic approaches and a unique cell transplantation system in developing zebrafish, we have uncovered a novel role for insulin signaling in the negative regulation of pancreatic progenitor cell differentiation. Blocking insulin signaling in the pancreatic progenitors hastened the expression of the essential β cell genes insulin and pdx1, and promoted β cell fate at the expense of alpha cell fate. In addition, loss of insulin signaling promoted β cell regeneration and destabilization of alpha cell character. These data indicate that insulin signaling constitutes a tunable mechanism for β cell compensatory plasticity during early development. Moreover, using a novel blastomere-to-larva transplantation strategy, we found that loss of insulin signaling in endoderm-committed blastomeres drove their differentiation into β cells. Furthermore, the extent of this differentiation was dependent on the function of the β cell mass in the host. Altogether, our results indicate that modulation of insulin signaling will be crucial for the development of β cell restoration therapies for diabetics; further clarification of the mechanisms of insulin signaling in β cell progenitors will reveal therapeutic targets for both in vivo and in vitro β cell generation.
Collapse
Affiliation(s)
- Lihua Ye
- Herman B Wells Center for Pediatric Research in the Department of Pediatrics and the Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, 635 Barnhill Drive, Van Nuys Medical Sciences Building MS2043, Indianapolis, IN 46202, USA; Department of Cellular and Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Van Nuys Medical Sciences Building MS2043, Indianapolis, IN 46202, USA
| | - Morgan A Robertson
- Herman B Wells Center for Pediatric Research in the Department of Pediatrics and the Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, 635 Barnhill Drive, Van Nuys Medical Sciences Building MS2043, Indianapolis, IN 46202, USA
| | - Teresa L Mastracci
- Herman B Wells Center for Pediatric Research in the Department of Pediatrics and the Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, 635 Barnhill Drive, Van Nuys Medical Sciences Building MS2043, Indianapolis, IN 46202, USA
| | - Ryan M Anderson
- Herman B Wells Center for Pediatric Research in the Department of Pediatrics and the Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, 635 Barnhill Drive, Van Nuys Medical Sciences Building MS2043, Indianapolis, IN 46202, USA; Department of Cellular and Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Van Nuys Medical Sciences Building MS2043, Indianapolis, IN 46202, USA.
| |
Collapse
|
37
|
Guerrera MC, De Pasquale F, Muglia U, Caruso G. Digestive enzymatic activity during ontogenetic development in zebrafish (Danio rerio). JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2015; 324:699-706. [PMID: 26477613 DOI: 10.1002/jez.b.22658] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Accepted: 10/05/2015] [Indexed: 12/13/2022]
Abstract
Despite the growing importance of zebrafish (Danio rerio) as an experimental model in biomedical research, some aspect of physiological and related morphological age dependent changes in digestive system during larval development are still unknown. In this paper, a biochemical and morphological study of the digestive tract of zebrafish was undertaken to record the functional changes occurring in this species during its ontogenetic development, particularly from 24 hr to 47 days post fertilization (dpf). Endo- and exo-proteases, as well as α-amylase enzymes, were quantified in zebrafish larvae before first feeding (7 dpf). The most morphologically significant events during the ontogenesis of the gut occurred between 3 dpf (mouth opening) and 7 dpf (end of exocrine pancreas differentiation). The presence of a wide range of digestive enzymes, already active at earlier zebrafish larval stages, closely related with the omnivorous diet of this species. Increasing enzyme activities were found with increasing age, probably in relation with intestinal mucosa folding and consequent absorption surface increase. J. Exp. Zool. (Mol. Dev. Evol.) 324B: 699-706, 2015. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Francesca De Pasquale
- Institute for Coastal Marine Environment (IAMC), National Research Council, Messina, Italy
| | - Ugo Muglia
- Department of Veterinary Sciences, University of Messina, Messina, Italy
| | - Gabriella Caruso
- Institute for Coastal Marine Environment (IAMC), National Research Council, Messina, Italy
| |
Collapse
|
38
|
Mastracci TL, Robertson MA, Mirmira RG, Anderson RM. Polyamine biosynthesis is critical for growth and differentiation of the pancreas. Sci Rep 2015; 5:13269. [PMID: 26299433 PMCID: PMC4547391 DOI: 10.1038/srep13269] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 06/30/2015] [Indexed: 02/03/2023] Open
Abstract
The pancreas, in most studied vertebrates, is a compound organ with both exocrine and endocrine functions. The exocrine compartment makes and secretes digestive enzymes, while the endocrine compartment, organized into islets of Langerhans, produces hormones that regulate blood glucose. High concentrations of polyamines, which are aliphatic amines, are reported in exocrine and endocrine cells, with insulin-producing β cells showing the highest concentrations. We utilized zebrafish as a model organism, together with pharmacological inhibition or genetic manipulation, to determine how polyamine biosynthesis functions in pancreatic organogenesis. We identified that inhibition of polyamine biosynthesis reduces exocrine pancreas and β cell mass, and that these reductions are at the level of differentiation. Moreover, we demonstrate that inhibition of ornithine decarboxylase (ODC), the rate-limiting enzyme in polyamine biosynthesis, phenocopies inhibition or knockdown of the enzyme deoxyhypusine synthase (DHS). These data identify that the pancreatic requirement for polyamine biosynthesis is largely mediated through a requirement for spermidine for the downstream posttranslational modification of eIF5A by its enzymatic activator DHS, which in turn impacts mRNA translation. Altogether, we have uncovered a role for polyamine biosynthesis in pancreatic organogenesis and identified that it may be possible to exploit polyamine biosynthesis to manipulate pancreatic cell differentiation.
Collapse
Affiliation(s)
- Teresa L Mastracci
- Department of Pediatrics, Indiana University School of Medicine, USA.,Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, USA
| | - Morgan A Robertson
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, USA
| | - Raghavendra G Mirmira
- Department of Pediatrics, Indiana University School of Medicine, USA.,Department of Physiology, Indiana University School of Medicine, USA.,Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, USA
| | - Ryan M Anderson
- Department of Pediatrics, Indiana University School of Medicine, USA.,Department of Physiology, Indiana University School of Medicine, USA.,Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, USA
| |
Collapse
|
39
|
Nishimura Y, Murakami S, Ashikawa Y, Sasagawa S, Umemoto N, Shimada Y, Tanaka T. Zebrafish as a systems toxicology model for developmental neurotoxicity testing. Congenit Anom (Kyoto) 2015; 55:1-16. [PMID: 25109898 DOI: 10.1111/cga.12079] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 07/29/2014] [Indexed: 12/18/2022]
Abstract
The developing brain is extremely sensitive to many chemicals. Exposure to neurotoxicants during development has been implicated in various neuropsychiatric and neurological disorders, including autism spectrum disorder, attention deficit hyperactive disorder, schizophrenia, Parkinson's disease, and Alzheimer's disease. Although rodents have been widely used for developmental neurotoxicity testing, experiments using large numbers of rodents are time-consuming, expensive, and raise ethical concerns. Using alternative non-mammalian animal models may relieve some of these pressures by allowing testing of large numbers of subjects while reducing expenses and minimizing the use of mammalian subjects. In this review, we discuss some of the advantages of using zebrafish in developmental neurotoxicity testing, focusing on central nervous system development, neurobehavior, toxicokinetics, and toxicodynamics in this species. We also describe some important examples of developmental neurotoxicity testing using zebrafish combined with gene expression profiling, neuroimaging, or neurobehavioral assessment. Zebrafish may be a systems toxicology model that has the potential to reveal the pathways of developmental neurotoxicity and to provide a sound basis for human risk assessments.
Collapse
Affiliation(s)
- Yuhei Nishimura
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine, Tsu, Japan; Mie University Medical Zebrafish Research Center, Tsu, Japan; Depertment of Systems Pharmacology, Mie University Graduate School of Medicine, Tsu, Japan; Department of Omics Medicine, Mie University Industrial Technology Innovation Institute, Tsu, Japan; Department of Bioinformatics, Mie University Life Science Research Center, Tsu, Japan
| | | | | | | | | | | | | |
Collapse
|
40
|
Navis A, Bagnat M. Loss of cftr function leads to pancreatic destruction in larval zebrafish. Dev Biol 2015; 399:237-48. [PMID: 25592226 DOI: 10.1016/j.ydbio.2014.12.034] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 12/29/2014] [Accepted: 12/30/2014] [Indexed: 12/14/2022]
Abstract
The development and function of many internal organs requires precisely regulated fluid secretion. A key regulator of vertebrate fluid secretion is an anion channel, the cystic fibrosis transmembrane conductance regulator (CFTR). Loss of CFTR function leads to defects in fluid transport and cystic fibrosis (CF), a complex disease characterized by a loss of fluid secretion and mucus buildup in many organs including the lungs, liver, and pancreas. Several animal models including mouse, ferret and pig have been generated to investigate the pathophysiology of CF. However, these models have limited accessibility to early processes in the development of CF and are not amenable for forward genetic or chemical screens. Here, we show that Cftr is expressed and localized to the apical membrane of the zebrafish pancreatic duct and that loss of cftr function leads to destruction of the exocrine pancreas and a cystic fibrosis phenotype that mirrors human disease. Our analyses reveal that the cftr mutant pancreas initially develops normally, then rapidly loses pancreatic tissue during larval life, reflecting pancreatic disease in CF. Altogether, we demonstrate that the cftr mutant zebrafish is a powerful new model for pancreatitis and pancreatic destruction in CF. This accessible model will allow more detailed investigation into the mechanisms that drive CF of the pancreas and facilitate development of new therapies to treat the disease.
Collapse
Affiliation(s)
- Adam Navis
- Department of Cell Biology, Duke University Medical Center, Durham, NC, United States
| | - Michel Bagnat
- Department of Cell Biology, Duke University Medical Center, Durham, NC, United States.
| |
Collapse
|
41
|
Zhang D, Golubkov VS, Han W, Correa RG, Zhou Y, Lee S, Strongin AY, Dong PDS. Identification of Annexin A4 as a hepatopancreas factor involved in liver cell survival. Dev Biol 2014; 395:96-110. [PMID: 25176043 DOI: 10.1016/j.ydbio.2014.08.025] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 08/19/2014] [Accepted: 08/20/2014] [Indexed: 01/27/2023]
Abstract
To gain insight into liver and pancreas development, we investigated the target of 2F11, a monoclonal antibody of unknown antigen, widely used in zebrafish studies for labeling hepatopancreatic ducts. Utilizing mass spectrometry and in vivo assays, we determined the molecular target of 2F11 to be Annexin A4 (Anxa4), a calcium binding protein. We further found that in both zebrafish and mouse endoderm, Anxa4 is broadly expressed in the developing liver and pancreas, and later becomes more restricted to the hepatopancreatic ducts and pancreatic islets, including the insulin producing ß-cells. Although Anxa4 is a known target of several monogenic diabetes genes and its elevated expression is associated with chemoresistance in malignancy, its in vivo role is largely unexplored. Knockdown of Anxa4 in zebrafish leads to elevated expression of caspase 8 and Δ113p53, and liver bud specific activation of Caspase 3 and apoptosis. Mosaic knockdown reveal that Anxa4 is required cell-autonomously in the liver bud for cell survival. This finding is further corroborated with mosaic anxa4 knockout studies using the CRISPR/Cas9 system. Collectively, we identify Anxa4 as a new, evolutionarily conserved hepatopancreatic factor that is required in zebrafish for liver progenitor viability, through inhibition of the extrinsic apoptotic pathway. A role for Anxa4 in cell survival may have implications for the mechanism of diabetic ß-cell apoptosis and cancer cell chemoresistance.
Collapse
Affiliation(s)
- Danhua Zhang
- Sanford Children's Health Research Center, Programs in Genetic Disease, Development and Aging, and Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA; Graduate School of Biomedical Sciences, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Vladislav S Golubkov
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Wenlong Han
- NCI-Designated Cancer Center, Tumor Microenvironment Program, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Ricardo G Correa
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Ying Zhou
- Sanford Children's Health Research Center, Programs in Genetic Disease, Development and Aging, and Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA; Graduate School of Biomedical Sciences, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Sunyoung Lee
- NCI-Designated Cancer Center, Tumor Microenvironment Program, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Alex Y Strongin
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - P Duc Si Dong
- Sanford Children's Health Research Center, Programs in Genetic Disease, Development and Aging, and Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA; Graduate School of Biomedical Sciences, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA.
| |
Collapse
|
42
|
Abstract
The islets of Langerhans are key regulators of glucose homeostasis and have been known as a structure for almost one and a half centuries. During the twentieth century several different cell types were described in the islets of different species and at different developmental stages. Six cell types with identified hormonal product have been described so far by the use of histochemical staining methods, transmission electron microscopy, and immunohistochemistry. Thus, glucagon-producing α-cells, insulin-producing β-cells, somatostatin-producing δ-cells, pancreatic polypeptide-producing PP-cells, serotonin-producing enterochromaffin-cells, and gastrin-producing G-cells have all been found in the mammalian pancreas at least at some developmental stage. Species differences are at hand and age-related differences are also to be considered. Eleven years ago a novel cell type, the ghrelin cell, was discovered in the human islets. Subsequent studies have shown the presence of islet ghrelin cells in several animals, including mouse, rat, gerbils, and fish. The developmental regulation of ghrelin cells in the islets of mice has gained a lot of interest and several studies have added important pieces to the puzzle of molecular mechanisms and the genetic regulation that lead to differentiation into mature ghrelin cells. A body of evidence has shown that ghrelin is an insulinostatic hormone, and the potential for blockade of ghrelin signalling as a therapeutic avenue for type 2 diabetes is intriguing. Furthermore, ghrelin-expressing pancreatic tumours have been reported and ghrelin needs to be taken into account when diagnosing pancreatic tumours. In this review article, we summarise the knowledge about islet ghrelin cells obtained so far.
Collapse
Affiliation(s)
- Nils Wierup
- Unit of Neuroendocrine Cell Biology, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Clinical Research Centre, Scania University Hospital, Jan Waldenströms gata 35, SE 205 02 Malmö, Sweden Imaging Team, Novo Nordisk A/S, Novo Nordisk Park, DK2760 Måløv, Denmark
| | | | | |
Collapse
|
43
|
Hepatocyte growth factor signaling in intrapancreatic ductal cells drives pancreatic morphogenesis. PLoS Genet 2013; 9:e1003650. [PMID: 23935514 PMCID: PMC3723531 DOI: 10.1371/journal.pgen.1003650] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 06/04/2013] [Indexed: 12/17/2022] Open
Abstract
In a forward genetic screen for regulators of pancreas development in zebrafish, we identified donut(s908) , a mutant which exhibits failed outgrowth of the exocrine pancreas. The s908 mutation leads to a leucine to arginine substitution in the ectodomain of the hepatocyte growth factor (HGF) tyrosine kinase receptor, Met. This missense mutation impedes the proteolytic maturation of the receptor, its trafficking to the plasma membrane, and diminishes the phospho-activation of its kinase domain. Interestingly, during pancreatogenesis, met and its hgf ligands are expressed in pancreatic epithelia and mesenchyme, respectively. Although Met signaling elicits mitogenic and migratory responses in varied contexts, normal proliferation rates in donut mutant pancreata together with dysmorphic, mislocalized ductal cells suggest that met primarily functions motogenically in pancreatic tail formation. Treatment with PI3K and STAT3 inhibitors, but not with MAPK inhibitors, phenocopies the donut pancreatic defect, further indicating that Met signals through migratory pathways during pancreas development. Chimera analyses showed that Met-deficient cells were excluded from the duct, but not acinar, compartment in the pancreatic tail. Conversely, wild-type intrapancreatic duct and "tip cells" at the leading edge of the growing pancreas rescued the donut phenotype. Altogether, these results reveal a novel and essential role for HGF signaling in the intrapancreatic ducts during exocrine morphogenesis.
Collapse
|
44
|
Flasse LC, Pirson JL, Stern DG, Von Berg V, Manfroid I, Peers B, Voz ML. Ascl1b and Neurod1, instead of Neurog3, control pancreatic endocrine cell fate in zebrafish. BMC Biol 2013; 11:78. [PMID: 23835295 PMCID: PMC3726459 DOI: 10.1186/1741-7007-11-78] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 06/28/2013] [Indexed: 12/15/2022] Open
Abstract
Background NEUROG3 is a key regulator of pancreatic endocrine cell differentiation in mouse, essential for the generation of all mature hormone producing cells. It is repressed by Notch signaling that prevents pancreatic cell differentiation by maintaining precursors in an undifferentiated state. Results We show that, in zebrafish, neurog3 is not expressed in the pancreas and null neurog3 mutant embryos do not display any apparent endocrine defects. The control of endocrine cell fate is instead fulfilled by two basic helix-loop-helix factors, Ascl1b and Neurod1, that are both repressed by Notch signaling. ascl1b is transiently expressed in the mid-trunk endoderm just after gastrulation and is required for the generation of the first pancreatic endocrine precursor cells. Neurod1 is expressed afterwards in the pancreatic anlagen and pursues the endocrine cell differentiation program initiated by Ascl1b. Their complementary role in endocrine differentiation of the dorsal bud is demonstrated by the loss of all hormone-secreting cells following their simultaneous inactivation. This defect is due to a blockage of the initiation of endocrine cell differentiation. Conclusions This study demonstrates that NEUROG3 is not the unique pancreatic endocrine cell fate determinant in vertebrates. A general survey of endocrine cell fate determinants in the whole digestive system among vertebrates indicates that they all belong to the ARP/ASCL family but not necessarily to the Neurog3 subfamily. The identity of the ARP/ASCL factor involved depends not only on the organ but also on the species. One could, therefore, consider differentiating stem cells into insulin-producing cells without the involvement of NEUROG3 but via another ARP/ASCL factor.
Collapse
Affiliation(s)
- Lydie C Flasse
- Laboratory of zebrafish development and disease models, University of Liege (ULg), Liege 4000, Belgium
| | | | | | | | | | | | | |
Collapse
|
45
|
Yee NS, Kazi AA, Yee RK. Translating discovery in zebrafish pancreatic development to human pancreatic cancer: biomarkers, targets, pathogenesis, and therapeutics. Zebrafish 2013; 10:132-146. [PMID: 23682805 PMCID: PMC3673615 DOI: 10.1089/zeb.2012.0817] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Abstract Experimental studies in the zebrafish have greatly facilitated understanding of genetic regulation of the early developmental events in the pancreas. Various approaches using forward and reverse genetics, chemical genetics, and transgenesis in zebrafish have demonstrated generally conserved regulatory roles of mammalian genes and discovered novel genetic pathways in exocrine pancreatic development. Accumulating evidence has supported the use of zebrafish as a model of human malignant diseases, including pancreatic cancer. Studies have shown that the genetic regulators of exocrine pancreatic development in zebrafish can be translated into potential clinical biomarkers and therapeutic targets in human pancreatic adenocarcinoma. Transgenic zebrafish expressing oncogenic K-ras and zebrafish tumor xenograft model have emerged as valuable tools for dissecting the pathogenetic mechanisms of pancreatic cancer and for drug discovery and toxicology. Future analysis of the pancreas in zebrafish will continue to advance understanding of the genetic regulation and biological mechanisms during organogenesis. Results of those studies are expected to provide new insights into how aberrant developmental pathways contribute to formation and growth of pancreatic neoplasia, and hopefully generate valid biomarkers and targets as well as effective and safe therapeutics in pancreatic cancer.
Collapse
Affiliation(s)
- Nelson S Yee
- Division of Hematology-Oncology, Program of Experimental Therapeutics, Department of Medicine, Penn State Milton S. Hershey Medical Center, Penn State College of Medicine, Penn State Hershey Cancer Institute, Pennsylvania State University , Hershey, PA 17033-0850, USA.
| | | | | |
Collapse
|
46
|
Zhang X, Gong Z. Fluorescent transgenic zebrafish Tg(nkx2.2a:mEGFP) provides a highly sensitive monitoring tool for neurotoxins. PLoS One 2013; 8:e55474. [PMID: 23383332 PMCID: PMC3562320 DOI: 10.1371/journal.pone.0055474] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 12/23/2012] [Indexed: 11/25/2022] Open
Abstract
Previously a standard toxicological test termed as DarT (Danio rerio Teratogenic assay) using wild type zebrafish embryos has been established and it is widely applied in toxicological and chemical screenings. As an increasing number of fluorescent transgenic zebrafish lines with specific fluorescent protein expression specifically expressed in different organs and tissues, we envision that the fluorescent markers may provide more sensitive endpoints for monitoring chemical induced phenotypical changes. Here we employed Tg(nkx2.2a:mEGFP) transgenic zebrafish which have GFP expression in the central nervous system to investigate its potential for screening neurotoxic chemicals. Five potential neurotoxins (acetaminophen, atenolol, atrazine, ethanol and lindane) and one neuroprotectant (mefenamic acid) were tested. We found that the GFP-labeled ventral axons from trunk motoneurons, which were easily observed in live fry and measured for quantification, were a highly sensitive to all of the five neurotoxins and the length of axons was significantly reduced in fry which looked normal based on DarT endpoints at low concentrations of neurotoxins. Compared to the most sensitive endpoints of DarT, ventral axon marker could improve the detection limit of these neurotoxins by about 10 fold. In contrast, there was no improvement for detection of the mefenamic acid compared to all DarT endpoints. Thus, ventral axon lengths provide a convenient and measureable marker specifically for neurotoxins. Our study may open a new avenue to use other fluorescent transgenic zebrafish embryos/fry to develop sensitive and specific toxicological tests for different categories of chemicals.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | | |
Collapse
|
47
|
Djiotsa J, Verbruggen V, Giacomotto J, Ishibashi M, Manning E, Rinkwitz S, Manfroid I, Voz ML, Peers B. Pax4 is not essential for beta-cell differentiation in zebrafish embryos but modulates alpha-cell generation by repressing arx gene expression. BMC DEVELOPMENTAL BIOLOGY 2012; 12:37. [PMID: 23244389 PMCID: PMC3563606 DOI: 10.1186/1471-213x-12-37] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 12/14/2012] [Indexed: 01/03/2023]
Abstract
BACKGROUND Genetic studies in mouse have demonstrated the crucial function of PAX4 in pancreatic cell differentiation. This transcription factor specifies β- and δ-cell fate at the expense of α-cell identity by repressing Arx gene expression and ectopic expression of PAX4 in α-cells is sufficient to convert them into β-cells. Surprisingly, no Pax4 orthologous gene can be found in chicken and Xenopus tropicalis raising the question of the function of pax4 gene in lower vertebrates such as in fish. In the present study, we have analyzed the expression and the function of the orthologous pax4 gene in zebrafish. RESULTS pax4 gene is transiently expressed in the pancreas of zebrafish embryos and is mostly restricted to endocrine precursors as well as to some differentiating δ- and ε-cells but was not detected in differentiating β-cells. pax4 knock-down in zebrafish embryos caused a significant increase in α-cells number while having no apparent effect on β- and δ-cell differentiation. This rise of α-cells is due to an up-regulation of the Arx transcription factor. Conversely, knock-down of arx caused to a complete loss of α-cells and a concomitant increase of pax4 expression but had no effect on the number of β- and δ-cells. In addition to the mutual repression between Arx and Pax4, these two transcription factors negatively regulate the transcription of their own gene. Interestingly, disruption of pax4 RNA splicing or of arx RNA splicing by morpholinos targeting exon-intron junction sites caused a blockage of the altered transcripts in cell nuclei allowing an easy characterization of the arx- and pax4-deficient cells. Such analyses demonstrated that arx knock-down in zebrafish does not lead to a switch of cell fate, as reported in mouse, but rather blocks the cells in their differentiation process towards α-cells. CONCLUSIONS In zebrafish, pax4 is not required for the generation of the first β- and δ-cells deriving from the dorsal pancreatic bud, unlike its crucial role in the differentiation of these cell types in mouse. On the other hand, the mutual repression between Arx and Pax4 is observed in both mouse and zebrafish. These data suggests that the main original function of Pax4 during vertebrate evolution was to modulate the number of pancreatic α-cells and its role in β-cells differentiation appeared later in vertebrate evolution.
Collapse
Affiliation(s)
- Joachim Djiotsa
- Molecular Biology and Genetic Engineering, Giga-Research, University of Liège, 1 avenue de l'Hôpital B34, Sart-Tilman B-4000, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Persistent nutrient excess results in a compensatory increase in the β-cell number in mammals. It is unknown whether this response occurs in nonmammalian vertebrates, including zebrafish, a model for genetics and chemical genetics. We investigated the response of zebrafish β-cells to nutrient excess and the underlying mechanisms by culturing transgenic zebrafish larvae in solutions of different nutrient composition. The number of β-cells rapidly increases after persistent, but not intermittent, exposure to glucose or a lipid-rich diet. The response to glucose, but not the lipid-rich diet, required mammalian target of rapamycin activity. In contrast, inhibition of insulin/IGF-1 signaling in β-cells blocked the response to the lipid-rich diet, but not to glucose. Lineage tracing and marker expression analyses indicated that the new β-cells were not from self-replication but arose through differentiation of postmitotic precursor cells. On the basis of transgenic markers, we identified two groups of newly formed β-cells: one with nkx2.2 promoter activity and the other with mnx1 promoter activity. Thus, nutrient excess in zebrafish induces a rapid increase in β-cells though differentiation of two subpopulations of postmitotic precursor cells. This occurs through different mechanisms depending on the nutrient type and likely involves paracrine signaling between the differentiated β-cells and the precursor cells.
Collapse
Affiliation(s)
- Lisette A. Maddison
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Wenbiao Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
49
|
Provost E, Wehner KA, Zhong X, Ashar F, Nguyen E, Green R, Parsons MJ, Leach SD. Ribosomal biogenesis genes play an essential and p53-independent role in zebrafish pancreas development. Development 2012; 139:3232-41. [PMID: 22872088 PMCID: PMC3413166 DOI: 10.1242/dev.077107] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2012] [Indexed: 11/20/2022]
Abstract
Mutations in the human Shwachman-Bodian-Diamond syndrome (SBDS) gene cause defective ribosome assembly and are associated with exocrine pancreatic insufficiency, chronic neutropenia and skeletal defects. However, the mechanism underlying these phenotypes remains unclear. Here we show that knockdown of the zebrafish sbds ortholog fully recapitulates the spectrum of developmental abnormalities observed in the human syndrome, and further implicate impaired proliferation of ptf1a-expressing pancreatic progenitor cells as the basis for the observed pancreatic phenotype. It is thought that diseases of ribosome assembly share a p53-dependent mechanism. However, loss of p53 did not rescue the developmental defects associated with loss of zebrafish sbds. To clarify the molecular mechanisms underlying the observed organogenesis defects, we performed transcriptional profiling to identify candidate downstream mediators of the sbds phenotype. Among transcripts displaying differential expression, functional group analysis revealed marked enrichment of genes related to ribosome biogenesis, rRNA processing and translational initiation. Among these, ribosomal protein L3 (rpl3) and pescadillo (pes) were selected for additional analysis. Similar to knockdown of sbds, knockdown or mutation of either rpl3 or pes resulted in impaired expansion of pancreatic progenitor cells. The pancreatic phenotypes observed in rpl3- and pes-deficient embryos were also independent of p53. Together, these data suggest novel p53-independent roles for ribosomal biogenesis genes in zebrafish pancreas development.
Collapse
Affiliation(s)
- Elayne Provost
- Department of Surgery, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Karen A. Wehner
- The Howard Hughes Medical Institute and Department of Molecular Biology and Genetics, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Xiangang Zhong
- Department of Surgery, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Foram Ashar
- Institute for Genetic Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Rachel Green
- The Howard Hughes Medical Institute and Department of Molecular Biology and Genetics, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Michael J. Parsons
- Institute for Genetic Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Steven D. Leach
- Department of Surgery, Johns Hopkins University, Baltimore, MD 21205, USA
- Institute for Genetic Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
50
|
Abstract
A full understanding of embryonic endocrine pancreas development will be key to the establishment of islet replacement strategies. In particular, it is important to identify molecular pathways that establish the correct balance of specific endocrine pancreatic islet cell types. Recently, our work in the zebrafish has revealed that the correct ratio of α and β cell fates depends on the homeodomain transcription factor Mnx1 (Hb9); in the absence of functional Mnx1, β cell precursors give rise to α cells. ( 1) Our study suggests that mnx1 may function in β cell precursors to suppress the α cell fate. Here we consider how Mnx1 may interact with other endocrine-specific transcription factors to specify β cells. Our work emphasizes the vital importance of Mnx1 for β cell development, and suggests that identifying Mnx1 transcriptional targets in β cell precursors may provide important new information of direct relevance to stem cell-based protocols to cure diabetes.
Collapse
|